1
|
Wang W, Zhang S, Dou C, Hu B, Song H, Qi F, Zhao Y, Li X, Zhou M, Xie J, Deng K, Wu Q, Ye L, Cui C, Liu L, Huang J, Yang G. Pharmacokinetics, Safety, and Immunogenicity of a Biosimilar of Nivolumab (LY01015): A Randomized, Double-Blind, Parallel-Controlled Phase I Clinical Trial in Healthy Chinese Male Subjects. BioDrugs 2024; 38:855-865. [PMID: 39317850 DOI: 10.1007/s40259-024-00679-w] [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] [Accepted: 08/30/2024] [Indexed: 09/26/2024]
Abstract
BACKGROUND Nivolumab (Opdivo®) is the first anti-PD-1 antibody approved in the world. LY01015 is a potential biosimilar of nivolumab. OBJECTIVES This phase I study aimed to establish the pharmacokinetic equivalence between LY01015 and the original investigational nivolumab (Opdivo®) in healthy Chinese male subjects. Additionally, safety and immunogenicity were assessed. PATIENTS AND METHODS A randomized, double-blind, parallel-controlled, phase I trial was conducted with 176 healthy male adults receiving a single intravenous infusion of LY01015 or nivolumab at 0.3 mg/kg. Pharmacokinetics, safety, and immunogenicity were evaluated over a 99-day period. The primary pharmacokinetics endpoint was AUC0-∞, and the secondary pharmacokinetic endpoints included AUC0-t and Cmax. Pharmacokinetic bioequivalence was confirmed using standard equivalence margins of 80.00-125.00%. RESULTS This study is the first to report on the pharmacokinetics, safety, and immunogenicity of Opdivo® in healthy individuals. The pharmacokinetics profiles of LY01015 and Opdivo® were found to be comparable. The geometric mean ratios (90% confidence intervals) for the AUC0-∞, AUC0-t, and Cmax of LY01015 to Opdivo® were 94.49% (90.29-98.88%), 94.92% (88.73-101.54%), and 96.55% (93.32-99.90%), respectively, falling within the conventional bioequivalence criteria of 80.00-125.00%. The safety and immunogenicity were also comparable between the two groups. CONCLUSIONS LY01015 demonstrated highly similar pharmacokinetics to nivolumab in healthy Chinese male subjects. Both drugs exhibited comparable safety and immunogenicity profiles. TRIAL REGISTRATION This trial is registered at the Chinese Clinical Trial Registry website ( https://www.chictr.org.cn/ #ChiCTR2200064771).
Collapse
Affiliation(s)
- Wei Wang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Shengnan Zhang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Changlin Dou
- Shandong Boan Biotechnology Co. Ltd, Luye Life Sciences Group, Beijing, China
| | - Baihui Hu
- Clinical Research Center of Luye Pharma Group Ltd, Luye Life Sciences Group, Beijing, China
| | - Hongtao Song
- Clinical Research Center of Luye Pharma Group Ltd, Luye Life Sciences Group, Beijing, China
| | - Fan Qi
- Shandong Boan Biotechnology Co. Ltd, Luye Life Sciences Group, Beijing, China
| | - Yanyan Zhao
- Shandong Boan Biotechnology Co. Ltd, Luye Life Sciences Group, Beijing, China
| | - Xiaojing Li
- Shandong Boan Biotechnology Co. Ltd, Luye Life Sciences Group, Beijing, China
| | - Ming Zhou
- Shandong Boan Biotechnology Co. Ltd, Luye Life Sciences Group, Beijing, China
| | - Jinlian Xie
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Kunhong Deng
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Qian Wu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Ling Ye
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Chang Cui
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Li Liu
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Jie Huang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Guoping Yang
- Center of Clinical Pharmacology, The Third Xiangya Hospital, Central South University, Changsha, China.
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China.
- Department of Pharmacy, The Third Xiangya Hospital, Central South University, Changsha, China.
- Research Center of Drug Clinical Evaluation, Central South University, Changsha, China.
| |
Collapse
|
2
|
Xiong J, Ouyang W, Yang M, Gao Z, Zhou H, Lou H, Guo Y, Xu Z, Zheng L, Liu Y, Wang Z, Sun P, Niyazi H, Wang J, Chen Y, Zhang B, Li L, Kang X, Guo W. Efficacy and Safety of Iparomlimab, an Anti-PD-1 Antibody, in Patients with Advanced Solid Tumors: A Phase 1c Study. Adv Ther 2024; 41:4153-4171. [PMID: 39276185 DOI: 10.1007/s12325-024-02981-z] [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: 06/19/2024] [Accepted: 08/22/2024] [Indexed: 09/16/2024]
Abstract
INTRODUCTION Iparomlimab (QL1604) is a humanized immunoglobulin G4 mAb against programmed cell death protein 1 (PD-1). Here, we report the preliminary efficacy, safety, pharmacokinetics, and immunogenicity of iparomlimab in patients with advanced solid tumors. METHODS In this open-label, phase 1c study, patients with advanced or metastatic solid tumors, either failed or had no standard therapies available, were enrolled and received intravenous iparomlimab at 3 mg/kg once every 3 weeks. The primary efficacy endpoint was the objective response rate (ORR) assessed by the investigator per Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1. RESULTS Between July 20, 2020, and September 6, 2021, 71 patients were enrolled and received at least one dose of iparomlimab. The ORR was 9.9% (7/71) and disease control rate was 36.6% (26/71). Median duration of response of all responders was 10.7 months [95% confidence interval (CI), 1.4-not estimable]. Additionally, the median time to progression, progression-free survival, and overall survival were 1.4 months (95% CI, 1.4-2.8), 1.4 months (95% CI, 1.4-2.7), and 9.7 months (95% CI, 7.2-15.3), respectively. A total of 52 (73.2%) patients experienced treatment-related adverse events (TRAEs) (grade ≥ 3, 19.7%). The most common TRAE (≥ 10%) was anemia (18.3%). A total of 20 (28.2%) experienced immune-related adverse events (grade ≥ 3, 7.0%). TRAEs leading to discontinuation of study drug occurred in 4 (5.6%) patients, including immune-mediated myocarditis (2 patients), Guillain-Barré syndrome (1 patient), and diarrhea (1 patient). CONCLUSIONS Iparomlimab showed preliminary clinical activity and had a manageable safety profile in patients with advanced solid tumors. These results support further investigation of iparomlimab as monotherapy or in combination therapy in advanced solid tumors. TRIAL REGISTRATION ClinicalTrials.gov identifier, NCT05801094. Retrospectively registered in 2023-03-24.
Collapse
Affiliation(s)
- Jianping Xiong
- Department of Medical Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiwei Ouyang
- Phase I Ward, Guizhou Cancer Hospital, Guiyang, China
| | - Mengxiang Yang
- Department of Oncology, Liaocheng People's Hospital, Liaocheng, China
| | - Zhenyuan Gao
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Huan Zhou
- Department of Medical Oncology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Hanmei Lou
- Phase I Ward, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yabing Guo
- Liver Cancer Center/Phase I Clinical Research Laboratory, Nanfang Hospital, Guangzhou, China
| | - Zhongyuan Xu
- Liver Cancer Center/Phase I Clinical Research Laboratory, Nanfang Hospital, Guangzhou, China
| | - Ling Zheng
- Phase I Ward, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Ying Liu
- The Third Ward of Digestive Diseases, Henan Cancer Hospital, Zhengzhou, China
| | - Zhongfeng Wang
- Henan Cancer Hospital, The First Hospital of Jilin University, Changchun, China
| | - Ping Sun
- Department of Medical Oncology, Yantai Yuhuangding Hospital, Yantai, China
| | - Huerxidan Niyazi
- Department of Oncology/Phase I Ward, The First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, China
| | - Jianhua Wang
- Department of Oncology/Phase I Ward, The First Affiliated Hospital of Xinjiang Medical University, Wulumuqi, China
| | - Yan Chen
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Baihui Zhang
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Lingyan Li
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Xiaoyan Kang
- Clinical Research and Development Center, Qilu Pharmaceutical Co., Ltd, Jinan, China
| | - Weijian Guo
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
3
|
Roozitalab G, Abedi B, Imani S, Farghadani R, Jabbarzadeh Kaboli P. Comprehensive assessment of TECENTRIQ® and OPDIVO®: analyzing immunotherapy indications withdrawn in triple-negative breast cancer and hepatocellular carcinoma. Cancer Metastasis Rev 2024; 43:889-918. [PMID: 38409546 DOI: 10.1007/s10555-024-10174-x] [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: 08/17/2023] [Accepted: 02/05/2024] [Indexed: 02/28/2024]
Abstract
Atezolizumab (TECENTRIQ®) and nivolumab (OPDIVO®) are both immunotherapeutic indications targeting programmed cell death 1 ligand 1 (PD-L1) and programmed cell death 1 (PD-1), respectively. These inhibitors hold promise as therapies for triple-negative breast cancer (TNBC) and hepatocellular carcinoma (HCC) and have demonstrated encouraging results in reducing the progression and spread of tumors. However, due to their adverse effects and low response rates, the US Food and Drug Administration (FDA) has withdrawn the approval of atezolizumab in TNBC and nivolumab in HCC treatment. The withdrawals of atezolizumab and nivolumab have raised concerns regarding their effectiveness and the ability to predict treatment responses. Therefore, the current study aims to investigate the immunotherapy withdrawal of PD-1/PD-L1 inhibitors, specifically atezolizumab for TNBC and nivolumab for HCC. This study will examine both the structural and clinical aspects. This review provides detailed insights into the structure of the PD-1 receptor and its ligands, the interactions between PD-1 and PD-L1, and their interactions with the withdrawn antibodies (atezolizumab and nivolumab) as well as PD-1 and PD-L1 modifications. In addition, this review further assesses these antibodies in the context of TNBC and HCC. It seeks to elucidate the factors that contribute to diverse responses to PD-1/PD-L1 therapy in different types of cancer and propose approaches for predicting responses, mitigating the potential risks linked to therapy withdrawals, and optimizing patient outcomes. By better understanding the mechanisms underlying responses to PD-1/PD-L1 therapy and developing strategies to predict these responses, it is possible to create more efficient treatments for TNBC and HCC.
Collapse
Affiliation(s)
- Ghazaal Roozitalab
- Noncommunicable Diseases Research Center, Fasa University of Medical Sciences, Fasa, Iran
| | - Behnaz Abedi
- Department of Basic Sciences, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Saber Imani
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, People's Republic of China
| | - Reyhaneh Farghadani
- Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, Bandar Sunway, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| | - Parham Jabbarzadeh Kaboli
- Graduate Institute of Biomedical Sciences, Institute of Biochemistry and Molecular Biology, Research Center for Cancer Biology, Cancer Biology and Precision Therapeutics Center, and Center for Molecular Medicine, China Medical University, Taichung, 406, Taiwan.
| |
Collapse
|
4
|
Usdin M, Quarmby V, Zanghi J, Bernaards C, Liao L, Laxamana J, Wu B, Swanson S, Song Y, Siguenza P. Immunogenicity of Atezolizumab: Influence of Testing Method and Sampling Frequency on Reported Anti-drug Antibody Incidence Rates. AAPS J 2024; 26:84. [PMID: 39009791 DOI: 10.1208/s12248-024-00954-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Measurement of anti-drug antibodies (ADA) to assess the incidence of ADA in a clinical trial is a critical step in immunogenicity assessment during the development of a protein therapeutic. We developed novel graphical approaches to illustrate clinical trial ADA data for the PD-L1 inhibitor atezolizumab (Tecentriq) that included a systematic analysis of the impact of the timing of ADA sampling and ADA assay drug tolerance on reported ADA incidence. We found that approaches used across the industry for ADA incidence analysis provide a limited view of immunogenicity in oncology studies, where ADA detection may be confounded by both drug dosage and patient attrition. Moreover, these approaches can miss important temporal information about the immune response. Our results demonstrated that the methodology of ADA assessment for the atezolizumab program was specifically designed to capture most ADA responses to ensure accurate reporting of ADA incidence. We further showed that the use of sparse sampling and/or ADA test methods with insufficient drug tolerance may result in a significant underreporting of ADA incidence. We conclude that the comparison of ADA incidence between different drugs can be highly misleading and that a test method with appropriate sensitivity in the presence of the drug and a clinical sampling scheme that is aligned with ADA responses to a drug is required to accurately report ADA incidence.
Collapse
Affiliation(s)
- Maxime Usdin
- Genentech Research and Early Development (gRED) Computational Sciences, Genentech, Building 45, 620 E Grand Ave, South San Francisco, California, 94080, USA
| | - Valerie Quarmby
- Independent Consultant, South San Francisco, California, USA
| | - James Zanghi
- Independent Consultant, South San Francisco, California, USA.
- BioAnalytical Sciences, Genentech, Building 47, 660 E Grand Ave, South San Francisco, California, 94080, USA.
| | - Coen Bernaards
- Product Development Data Sciences, Genentech, Building 40, 600 E Grand Ave, South San Francisco, California, 94080, USA
| | - Laura Liao
- Product Development Data Sciences, Genentech, Building 40, 600 E Grand Ave, South San Francisco, California, 94080, USA
| | - Joel Laxamana
- Product Development Data Sciences, Genentech, Building 40, 600 E Grand Ave, South San Francisco, California, 94080, USA
| | - Benjamin Wu
- Clinical Pharmacology, Genentech, Building 46, 640 E Grand Ave, South San Francisco, California, 94080, USA
| | - Steven Swanson
- BioAnalytical Sciences, Genentech, Building 47, 660 E Grand Ave, South San Francisco, California, 94080, USA
| | - Yuan Song
- BioAnalytical Sciences, Genentech, Building 47, 660 E Grand Ave, South San Francisco, California, 94080, USA
| | - Patty Siguenza
- BioAnalytical Sciences, Genentech, Building 47, 660 E Grand Ave, South San Francisco, California, 94080, USA
| |
Collapse
|
5
|
Zheng Y, Zhu J, Xiong J, Jiang O, Wang H, Xie Y, Zhou Y, Xu N. Phase 1b/2 study of penpulimab (AK105), an antiprogrammed cell death-1 immunoglobulin G1 antibody, in advanced or metastatic solid tumors (AK105-204). Cancer 2024; 130:2180-2190. [PMID: 38412283 DOI: 10.1002/cncr.35259] [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: 05/05/2023] [Revised: 12/11/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND Penpulimab, a new-generation antiprogrammed cell death-1 immunoglobulin G1 monoclonal antibody, was engineered to optimize receptor occupancy and eliminate fragment crystallizable γ-mediated effector function. In this multicenter, phase 1b/2, multicohort study, the objective was to investigate the efficacy, safety, and immunogenicity of penpulimab in advanced solid tumors. METHODS Patients who had unresectable, advanced solid tumors were enrolled from six centers and received 200 mg penpulimab on day 1 every 2 weeks for up to 24 months. The primary end point was the objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors, version criteria 1.1. RESULTS Between September 2, 2019, and January 1, 2020, 65 patients were enrolled and received penpulimab. At the time of data cutoff (May 11, 2022), the median follow-up was 12.6 months (range, 1.1-28.6 months). The ORR was 12.3 (95% confidence interval [CI], 5.5%-22.8%), with three (4.6%) complete responses and five (7.7%) partial responses. Twelve patients (18.5%) achieved stable disease, resulting in a disease control rate of 30.8% (95% CI, 19.9%-43.4%). The median duration of response was not reached (95% CI, 6.70 months to not estimable). In all cohorts, the median progression-free survival was 1.74 months (95% CI, 1.41-2.69 months), and the median overall survival was 16.59 months (95% CI, 7.82-22.18 months). Grade 3 or greater treatment-related adverse events and immune-related adverse events occurred in 9.2% and 27.7% of patients, respectively. Positive antidrug antibody responses to penpulimab were observed in one patient (1.8%). CONCLUSIONS Penpulimab showed promising antitumor activity with an acceptable safety profile, offering a potential new treatment approach for solid tumors. These findings supported the evaluation of penpulimab's durable activity and safety, as monotherapy or in combination therapy, in specific malignancies.
Collapse
Affiliation(s)
- Yulong Zheng
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianqing Zhu
- Department of Gynecologic Oncology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ou Jiang
- Department of Tumor Surgery, Neijiang Second People's Hospital, Neijiang, Sichuan, China
| | - Hong Wang
- Department of Oncology, The Third Hospital of Nanchang, Nanchang, Jiangxi, China
| | - Yanru Xie
- Department of Oncology, Lishui City Central Hospital, Lishui, Zhejiang, China
| | - Yuefen Zhou
- Department of Oncology, Lishui City Central Hospital, Lishui, Zhejiang, China
| | - Nong Xu
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
6
|
Ladd AD, Duarte S, Sahin I, Zarrinpar A. Mechanisms of drug resistance in HCC. Hepatology 2024; 79:926-940. [PMID: 36680397 DOI: 10.1097/hep.0000000000000237] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 11/21/2022] [Indexed: 01/22/2023]
Abstract
HCC comprises ∼80% of primary liver cancer. HCC is the only major cancer for which death rates have not improved over the last 10 years. Most patients are diagnosed with advanced disease when surgical and locoregional treatments are not feasible or effective. Sorafenib, a multikinase inhibitor targeting cell growth and angiogenesis, was approved for advanced unresectable HCC in 2007. Since then, other multikinase inhibitors have been approved. Lenvatinib was found to be noninferior to sorafenib as a first-line agent. Regorafenib, cabozantinib, and ramucirumab were shown to prolong survival as second-line agents. Advances in immunotherapy for HCC have also added hope for patients, but their efficacy remains limited. A large proportion of patients with advanced HCC gain no long-term benefit from systemic therapy due to primary and acquired drug resistance, which, combined with its rising incidence, keeps HCC a highly fatal disease. This review summarizes mechanisms of primary and acquired resistance to therapy and includes methods for bypassing resistance. It addresses recent advancements in immunotherapy, provides new perspectives on the linkage between drug resistance and molecular etiology of HCC, and evaluates the role of the microbiome in drug resistance. It also discusses alterations in signaling pathways, dysregulation of apoptosis, modulations in the tumor microenvironment, involvement of cancer stem cells, changes in drug metabolism/transport, tumor hypoxia, DNA repair, and the role of microRNAs in drug resistance. Understanding the interplay among these factors will provide guidance on the development of new therapeutic strategies capable of improving patient outcomes.
Collapse
Affiliation(s)
- Alexandra D Ladd
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Sergio Duarte
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ilyas Sahin
- Division of Hematology/Oncology, Department of Medicine, College of Medicine, University of Florida, Gainesville, Florida, USA
| | - Ali Zarrinpar
- Department of Surgery, College of Medicine, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
7
|
Hu P, Dai HI, Bourdage J, Zhou D, Trang K, Kowalski K, Bello C, Hibma J, Khandelwal A, Cowan K, Dong J, Venkatakrishnan K, Gao W. Immunogenicity of avelumab in patients with metastatic Merkel cell carcinoma or advanced urothelial carcinoma. Clin Transl Sci 2024; 17:e13730. [PMID: 38411318 PMCID: PMC10897865 DOI: 10.1111/cts.13730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 01/04/2024] [Accepted: 01/16/2024] [Indexed: 02/28/2024] Open
Abstract
Like other monoclonal antibodies, immune checkpoint inhibitors may be immunogenic in some patients, potentially affecting pharmacokinetics (PKs) and clinical outcomes. In post hoc analyses, we characterized antidrug antibody (ADA) development with avelumab monotherapy in patients with metastatic Merkel cell carcinoma (mMCC) from the JAVELIN Merkel 200 trial (first-line [1L; N = 116] and second-line or later [≥2L; N = 88] cohorts) or with advanced urothelial carcinoma (aUC) from the JAVELIN Bladder 100 (1L maintenance [N = 350]) and JAVELIN Solid Tumor (≥2L [N = 249]) trials. Treatment-emergent ADAs developed in a numerically higher proportion of patients with aUC (1L maintenance, 19.1%; ≥2L, 18.1%) versus mMCC (1L, 8.2%; ≥2L, 8.9%); incidences within tumor types were similar by line of therapy. In PK analyses, numerically lower avelumab trough concentration and higher baseline clearance were observed in treatment-emergent ADA+ versus ADA- subgroups; however, differences were not clinically relevant. Numerical differences in overall survival, progression-free survival, or objective response rate by ADA status were observed; however, no clinically meaningful trends were identified. Proportions of patients with treatment-emergent adverse events (TEAEs; any grade or grade 3/4), serious TEAEs, TEAEs leading to treatment discontinuation, or infusion-related reactions were similar, with overlapping 80% confidence intervals between ADA subgroups. Efficacy and safety observations were similar in subgroups defined by early development of ADA+ status during treatment. In conclusion, no meaningful differences in PKs, efficacy, and safety were observed between subgroups of avelumab-treated patients with different ADA status. Overall, these data suggest that ADAs are not relevant for treatment decisions with avelumab.
Collapse
Affiliation(s)
- Ping Hu
- EMD SeronoBillericaMassachusettsUSA
| | - Haiqing Isaac Dai
- EMD SeronoBillericaMassachusettsUSA
- Present address:
BioNTechCambridgeMassachusettsUSA
| | | | - Dongli Zhou
- the healthcare business of Merck KGaADarmstadtGermany
| | - Ky Trang
- the healthcare business of Merck KGaADarmstadtGermany
| | | | | | | | - Akash Khandelwal
- the healthcare business of Merck KGaADarmstadtGermany
- Present address:
UCB BIOSCIENCES GmbHMonheim am RheinGermany
| | - Kyra Cowan
- the healthcare business of Merck KGaADarmstadtGermany
| | | | | | - Wei Gao
- EMD SeronoBillericaMassachusettsUSA
| |
Collapse
|
8
|
Guo Y, Remaily BC, Thomas J, Kim K, Kulp SK, Mace TA, Ganesan LP, Owen DH, Coss CC, Phelps MA. Antibody Drug Clearance: An Underexplored Marker of Outcomes with Checkpoint Inhibitors. Clin Cancer Res 2024; 30:942-958. [PMID: 37921739 PMCID: PMC10922515 DOI: 10.1158/1078-0432.ccr-23-1683] [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: 06/13/2023] [Revised: 08/23/2023] [Accepted: 10/13/2023] [Indexed: 11/04/2023]
Abstract
Immune-checkpoint inhibitor (ICI) therapy has dramatically changed the clinical landscape for several cancers, and ICI use continues to expand across many cancer types. Low baseline clearance (CL) and/or a large reduction of CL during treatment correlates with better clinical response and longer survival. Similar phenomena have also been reported with other monoclonal antibodies (mAb) in cancer and other diseases, highlighting a characteristic of mAb clinical pharmacology that is potentially shared among various mAbs and diseases. Though tempting to attribute poor outcomes to low drug exposure and arguably low target engagement due to high CL, such speculation is not supported by the relatively flat exposure-response relationship of most ICIs, where a higher dose or exposure is not likely to provide additional benefit. Instead, an elevated and/or increasing CL could be a surrogate marker of the inherent resistant phenotype that cannot be reversed by maximizing drug exposure. The mechanisms connecting ICI clearance, therapeutic efficacy, and resistance are unclear and likely to be multifactorial. Therefore, to explore the potential of ICI CL as an early marker for efficacy, this review highlights the similarities and differences of CL characteristics and CL-response relationships for all FDA-approved ICIs, and we compare and contrast these to selected non-ICI mAbs. We also discuss underlying mechanisms that potentially link mAb CL with efficacy and highlight existing knowledge gaps and future directions where more clinical and preclinical investigations are warranted to clearly understand the value of baseline and/or time-varying CL in predicting response to ICI-based therapeutics.
Collapse
Affiliation(s)
- Yizhen Guo
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Bryan C. Remaily
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Justin Thomas
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Kyeongmin Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Samuel K. Kulp
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Thomas A. Mace
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Latha P. Ganesan
- Department of Internal Medicine, Division of Rheumatology and Immunology, Division of Nephrology, The Ohio State University Wexner Medical Center, Columbus, OH
| | - Dwight H. Owen
- Division of Medical Oncology, Ohio State University Wexner Medical Center, James Cancer Hospital and Solove Research Institute, Columbus, OH
| | - Christopher C. Coss
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| | - Mitch A. Phelps
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH
| |
Collapse
|
9
|
Kong Y, Wang X, Qie R. Immunotherapy-associated cardiovascular toxicities: insights from preclinical and clinical studies. Front Oncol 2024; 14:1347140. [PMID: 38482205 PMCID: PMC10932998 DOI: 10.3389/fonc.2024.1347140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/12/2024] [Indexed: 11/02/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have become a widely accepted and effective treatment for various types of solid tumors. Recent studies suggest that cardiovascular immune-related adverse events (irAEs) specifically have an incidence rate ranging from 1.14% to more than 5%. Myocarditis is the most common observed cardiovascular irAE. Others include arrhythmias, pericardial diseases, vasculitis, and a condition resembling takotsubo cardiomyopathy. Programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway, cytotoxic T-lymphocyte antigen-4 (CTLA-4) pathway, and the recently discovered lymphocyte-activation gene 3 (LAG-3) pathway, play a critical role in boosting the body's natural immune response against cancer cells. While ICIs offer significant benefits in terms of augmenting immune function, they can also give rise to unwanted inflammatory side effects known as irAEs. The occurrence of irAEs can vary in severity, ranging from mild to severe, and can impact the overall clinical efficacy of these agents. This review aims to summarize the underlying mechanisms of cardiovascular irAE from both preclinical and clinical studies for a better understanding of cardiovascular irAE in clinical application.
Collapse
Affiliation(s)
- Youqian Kong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rui Qie
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
10
|
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
|
11
|
Majenka P, Loquai C, Schöning T, Enk A, Hassel J. Acute low back pain as infusion-related reaction to monoclonal antibodies. Front Oncol 2023; 13:1161818. [PMID: 37941549 PMCID: PMC10627944 DOI: 10.3389/fonc.2023.1161818] [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: 06/02/2023] [Accepted: 09/20/2023] [Indexed: 11/10/2023] Open
Abstract
Monoclonal antibodies, such as PD-1 inhibitors, are increasingly used in various cancers. Acute low back pain as infusion-related reaction (IRR) to monoclonal antibodies is poorly described. We report a bicentric series of 10 cases of acute low back pain due to administration of monoclonal antibodies directed against PD-1/PD-L1 for skin cancer treatment in patients treated at University Hospital Heidelberg and University Medical Center Mainz (Germany). The management of IRR symptoms was immediate interruption of infusion and analgesia leading to quick improvement and complete symptom relief in all patients. Our findings suggest that the risk of developing low back pain as IRR is depending on the concentration of the administered drug. Low back pain as IRR can be managed by early interruption of infusion and by decreasing the infusion rate or concentration in following administrations.
Collapse
Affiliation(s)
- Pawel Majenka
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Carmen Loquai
- Department of Dermatology, University Medical Center Mainz, Mainz, Germany
| | - Tilman Schöning
- Department of Pharmacy, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Enk
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Jessica Hassel
- Department of Dermatology and National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
12
|
Sangro B, Yau T, El‐Khoueiry AB, Kudo M, Shen Y, Tschaika M, Roy A, Feng Y, Gao L, Aras U. Exposure-response analysis for nivolumab plus ipilimumab combination therapy in patients with advanced hepatocellular carcinoma (CheckMate 040). Clin Transl Sci 2023; 16:1445-1457. [PMID: 37165980 PMCID: PMC10432868 DOI: 10.1111/cts.13544] [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: 02/07/2023] [Revised: 04/17/2023] [Accepted: 04/30/2023] [Indexed: 05/12/2023] Open
Abstract
This analysis was conducted to inform dose selection of a combination of nivolumab plus ipilimumab for the treatment of sorafenib-experienced patients with hepatocellular carcinoma (HCC). CheckMate 040 is an open-label, multicohort, phase I/II trial in adults with advanced HCC that evaluated nivolumab monotherapy (0.1-10 mg/kg once every 2 weeks [q2w]) and the following three combinations of nivolumab plus ipilimumab: (1) nivolumab 1 mg/kg plus ipilimumab 3 mg/kg every 3 weeks (q3w) for four doses, followed by nivolumab monotherapy 240 mg q2w (arm A); (2) nivolumab 3 mg/kg plus ipilimumab 1 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w (arm B); and (3) nivolumab 3 mg/kg q2w plus ipilimumab 1 mg/kg every 6 weeks continuously (arm C). Exposure-response relationships (efficacy and safety) were characterized using nivolumab and ipilimumab concentrations after the first dose (Cavg1) as the exposure measure. Objective tumor response (OTR) and overall survival (OS) improvements were associated with increased ipilimumab exposure (OTR: odds ratio 1.45, 95% confidence interval [CI], 1.13-1.86; OS: hazard ratio 0.86, 95% CI 0.75-0.98), but not nivolumab exposure (OTR: odds ratio 0.99, 95% CI 0.97-1.02; OS: hazard ratio 1.08, 95% CI 0.89-1.32). Hepatic treatment-related and immune-mediated adverse events were more common in arm A than in arms B or C. Nivolumab 1 mg/kg plus ipilimumab 3 mg/kg q3w for four doses, followed by nivolumab monotherapy 240 mg q2w had the most favorable benefit:risk profile in patients with advanced HCC.
Collapse
Affiliation(s)
- Bruno Sangro
- Liver UnitClinica Universidad de Navarra‐IDISNA and CIBEREHDPamplonaSpain
| | - Thomas Yau
- University of Hong Kong, Hong Kong Special Administrative RegionPokfulamChina
| | | | - Masatoshi Kudo
- Department of Gastroenterology and HepatologyKindai University Faculty of MedicineOsakaJapan
| | - Yun Shen
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | | | - Amit Roy
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Yan Feng
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Ling Gao
- Bristol Myers SquibbPrincetonNew JerseyUSA
| | - Urvi Aras
- Bristol Myers SquibbPrincetonNew JerseyUSA
| |
Collapse
|
13
|
Yu SJ. Immunotherapy for hepatocellular carcinoma: Recent advances and future targets. Pharmacol Ther 2023; 244:108387. [PMID: 36948423 DOI: 10.1016/j.pharmthera.2023.108387] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 02/12/2023] [Accepted: 03/15/2023] [Indexed: 03/24/2023]
Abstract
Immunotherapy is a promising approach to treating various types of cancers, including hepatocellular carcinoma (HCC). While single immunotherapy drugs show limited effectiveness on a small subset of patients, the combination of the anti PD-L1 atezolizumab and anti-vascular endothelial growth factor bevacizumab has shown significant improvement in survival compared to sorafenib as a first-line treatment. However, the current treatment options still have a low success rate of about 30%. Thus, more effective treatments for HCC are urgently required. Several novel immunotherapeutic methods, including the use of novel immune checkpoint inhibitors, innovative immune cell therapies like chimeric antigen receptor T cells (CAR-T), TCR gene-modified T cells and stem cells, as well as combination strategies are being tested in clinical trials for the treatment of HCC. However, some crucial issues still exist such as the presence of heterogeneous antigens in solid tumors, the immune-suppressive environment within tumors, the risk of on-target/off-tumor, infiltrating CAR-T cells, immunosuppressive checkpoint molecules, and cytokines. Overall, immunotherapy is on the brink of major advancements in the fight against HCC.
Collapse
Affiliation(s)
- Su Jong Yu
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
14
|
Kawakami N, Saito H, Takahashi S, Kajie S, Kato R, Shimaya K, Wakai Y, Saito K, Sakashita M. Airway disorders associated with immune checkpoint inhibitor therapy: Two case reports and a systematic review. Semin Oncol 2022; 49:439-455. [PMID: 36759235 DOI: 10.1053/j.seminoncol.2023.01.003] [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: 01/13/2023] [Revised: 01/15/2023] [Accepted: 01/16/2023] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors (ICI) are widely used for the treatment of various malignant neoplasms. Interstitial lung disease is a well-known immune-related adverse event, however, ICI-induced airway disease remains under-recognized. Herein, we report two similar cases of pembrolizumab-induced tracheobronchitis presenting as persistent chronic cough and dyspnea. Blood tests revealed elevated C-reactive protein levels without eosinophilia. Spirometry demonstrated mild airflow obstruction. Computed tomography revealed diffuse thickening of the tracheobronchial walls and bronchiectasis predominantly in the lower lobes. Bronchoscopy revealed edematous and erythematous tracheobronchial mucosa, and bronchial biopsy tissue exhibited marked inflammation with predominant infiltration of CD8+ lymphocytes. Subsequently, pembrolizumab-induced tracheobronchitis was diagnosed in both cases. Cessation of pembrolizumab and initiation of erythromycin, inhaled corticosteroids, and long-acting beta-agonists gradually improved the symptoms, airflow obstruction, and radiographic findings. These were completely resolved in one case. The other case initially showed a poor response to systemic corticosteroids combined with the aforementioned drugs, but improved gradually and almost completely. These cases exemplify ICI-induced airway disease that is, an under-recognized manifestation of immune-related adverse events. In addition, we have systematically searched the PubMed database for articles on ICI-induced airway disease, categorized the retrieved articles as eosinophilic and non-eosinophilic airway diseases, and reviewed the differences in treatment and prognoses between these two categories.
Collapse
Affiliation(s)
- Naoki Kawakami
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan.
| | - Hiroaki Saito
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Susumu Takahashi
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Shinpei Kajie
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Rina Kato
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Kazuhiro Shimaya
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Yoko Wakai
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Kazuhito Saito
- Department of Respiratory Medicine, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| | - Mai Sakashita
- Department of Pathology, Tsuchiura Kyodo General Hospital, Tsuchiura, Ibaraki, Japan
| |
Collapse
|
15
|
Pallozzi M, Di Tommaso N, Maccauro V, Santopaolo F, Gasbarrini A, Ponziani FR, Pompili M. Non-Invasive Biomarkers for Immunotherapy in Patients with Hepatocellular Carcinoma: Current Knowledge and Future Perspectives. Cancers (Basel) 2022; 14:cancers14194631. [PMID: 36230554 PMCID: PMC9559710 DOI: 10.3390/cancers14194631] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/16/2022] Open
Abstract
Simple Summary The search for non-invasive biomarkers is a hot topic in modern oncology, since a tissue biopsy has significant limitations in terms of cost and invasiveness. The treatment perspectives have been significantly improved after the approval of immunotherapy for patients with hepatocellular carcinoma; therefore, the quick identification of responders is crucial to define the best therapeutic strategy. In this review, the current knowledge on the available non-invasive biomarkers of the response to immunotherapy is described. Abstract The treatment perspectives of advanced hepatocellular carcinoma (HCC) have deeply changed after the introduction of immunotherapy. The results in responders show improved survival compared with Sorafenib, but only one-third of patients achieve a significant benefit from treatment. As the tumor microenvironment exerts a central role in shaping the response to immunotherapy, the future goal of HCC treatment should be to identify a proxy of the hepatic tissue condition that is easy to use in clinical practice. Therefore, the search for biomarkers that are accurate in predicting prognosis will be the hot topic in the therapeutic management of HCC in the near future. Understanding the mechanisms of resistance to immunotherapy may expand the patient population that will benefit from it, and help researchers to find new combination regimens to improve patients’ outcomes. In this review, we describe the current knowledge on the prognostic non-invasive biomarkers related to treatment with immune checkpoint inhibitors, focusing on serological markers and gut microbiota.
Collapse
Affiliation(s)
- Maria Pallozzi
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Natalia Di Tommaso
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Valeria Maccauro
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Santopaolo
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Translational Medicine and Surgery Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Translational Medicine and Surgery Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence: (F.R.P.); (M.P.)
| | - Maurizio Pompili
- Internal Medicine and Gastroenterology-Hepatology Unit, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
- Translational Medicine and Surgery Department, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence: (F.R.P.); (M.P.)
| |
Collapse
|
16
|
Lee CK, Chan SL, Chon HJ. Could We Predict the Response of Immune Checkpoint Inhibitor Treatment in Hepatocellular Carcinoma? Cancers (Basel) 2022; 14:3213. [PMID: 35804984 PMCID: PMC9264773 DOI: 10.3390/cancers14133213] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
The use of anti-programmed cell-death protein (ligand)-1 (PD-[L]1) is an important strategy for treating hepatocellular carcinoma (HCC). However, the treatment only benefits 10-20% of patients when used as a monotherapy. Therefore, the selection of patients for anti-PD-1/PD-L1 treatment is crucial for both patients and clinicians. This review aimed to explore the existing literature on tissue or circulating markers for the identification of responders or non-responders to anti-PD-1/PD-L1 in HCC. For the clinically available markers, both etiological factors (viral versus non-viral) and disease extent (intra-hepatic vs. extrahepatic) impact the responses to anti-PD-1/PD-L1, warranting further studies. Preliminary data suggested that inflammatory indices (e.g., neutrophil-lymphocyte ratio) may be associated with clinical outcomes of HCC during the anti-PD-1/PD-L1 treatment. Finally, although PD-L1 expression in tumor tissues is a predictive marker for multiple cancer types, its clinical application is less clear in HCC due to the lack of a clear-cut association with responders to anti-PD-1/PD-L1 treatment. Although all translational markers are not routinely measured in HCC, recent data suggest their potential roles in selecting patients for anti-PD-1/PD-L1 treatment. Such markers, including the immune classification of HCC, selected signaling pathways, tumor-infiltrating lymphocytes, and auto-antibodies, were discussed in this review.
Collapse
Affiliation(s)
- Choong-kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul 03722, Korea;
| | - Stephen L. Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Hong Jae Chon
- Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea
| |
Collapse
|
17
|
Mosch R, Guchelaar HJ. Immunogenicity of Monoclonal Antibodies and the Potential Use of HLA Haplotypes to Predict Vulnerable Patients. Front Immunol 2022; 13:885672. [PMID: 35784343 PMCID: PMC9249215 DOI: 10.3389/fimmu.2022.885672] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/24/2022] [Indexed: 01/14/2023] Open
Abstract
The use of monoclonal antibodies (mAbs) in the clinic has successfully expanded to treatment of cancer, viral infections, inflammations, and other indications. However, some of the classes of mAbs that are used in the clinic show the formation of anti-drug antibodies (ADAs) leading to loss of efficacy. This review describes ADA formation for the various mAbs, and its clinical effect. Lastly, this review considers the use of HLA-haplotypes as biomarkers to predict vulnerability of patients sensitive to formation of ADAs.
Collapse
|
18
|
In vitro preliminary study on different anti-PD-1 antibody concentrations on T cells activation. Sci Rep 2022; 12:8370. [PMID: 35589776 PMCID: PMC9120143 DOI: 10.1038/s41598-022-12136-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 05/06/2022] [Indexed: 11/12/2022] Open
Abstract
Lung adenocarcinoma predominates among diagnosed nonsmall cell lung cancer subtypes in nonsmokers. The introduction of immune checkpoint inhibitors into clinical practice offered patients prolonged progression-free survival and overall survival times. However, the results demonstrate that the benefits do not apply to all patients. Nivolumab is a monoclonal antibody against the PD-1 protein expressed mainly on T lymphocytes and is widely used in cancer therapy in different settings. Tumor cells often express the PD-L1 molecule and can effectively block the action of PD-1-positive lymphocytes. A body of knowledge regarding the high expression of PD-L1 on tumor cells highlights that it does not always correlate with the effectiveness of anti-PD-1 therapy. The side effects of the therapy also constitute a significant issue. These side effects can occur at any time during anti-PD-1 treatment and lead to discontinuation and even the death of the patient. In these situations, it is possible to delay the dosage. Nevertheless, unfortunately, it is not possible to reduce the dose of anti-PD-1 antibody, which would undoubtedly minimize side effects, leaving the patient's immune system active. In our preliminary study, we analyzed the effect of different concentrations of nivolumab on the functioning of T lymphocytes. Activation and proliferation markers were investigated on T cells after being cultured with antigen-stimulated autologous dendritic cells. This process may indicate an appropriate concentration of nivolumab, which shows clinical activity with minimal side effects.
Collapse
|
19
|
Kasichayanula S, Mandlekar S, Shivva V, Patel M, Girish S. Evolution of Preclinical Characterization and Insights into Clinical Pharmacology of Checkpoint Inhibitors Approved for Cancer Immunotherapy. Clin Transl Sci 2022; 15:1818-1837. [PMID: 35588531 PMCID: PMC9372426 DOI: 10.1111/cts.13312] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 05/05/2022] [Accepted: 05/09/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer immunotherapy has significantly advanced the treatment paradigm in oncology, with approvals of immuno‐oncology agents for over 16 indications, many of them first line. Checkpoint inhibitors (CPIs) are recognized as an essential backbone for a successful anticancer therapy regimen. This review focuses on the US Food and Drug Administration (FDA) regulatory approvals of major CPIs and the evolution of translational advances since their first approval close to a decade ago. In addition, critical preclinical and clinical pharmacology considerations, an overview of the pharmacokinetic and dose/regimen aspects, and a discussion of the future of CPI translational and clinical pharmacology as combination therapy becomes a mainstay of industrial immunotherapy development and in clinical practice are also discussed.
Collapse
Affiliation(s)
| | | | - Vittal Shivva
- Genentech, 1 DNA Way, South San Francisco, 94080, CA
| | - Maulik Patel
- AbbVie Inc., 1000 Gateway Blvd, South San Francisco, 94080, CA
| | - Sandhya Girish
- Gilead Sciences, 310 Lakeside Drive, Foster City, 94404, CA
| |
Collapse
|
20
|
Anti-drug antibodies in the current management of cancer. Cancer Chemother Pharmacol 2022; 89:577-584. [PMID: 35333967 DOI: 10.1007/s00280-022-04418-2] [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: 11/05/2021] [Accepted: 03/08/2022] [Indexed: 11/02/2022]
Abstract
Monoclonal antibodies (mAbs) have become one of the main therapeutic weapons in modern oncology, mainly as targeted therapies, and immune checkpoint inhibitors. The generation of anti-drug antibodies (ADAs) after their administration can alter their pharmacokinetic, pharmacodynamic, efficacy and safety profile causing infusion-related reactions. Several risk factors have been associated with ADAs development, notably host genetics and immune status, comorbidity, concomitant medications, mAbs molecular structure, dose and route of administration. ADAs are not usually tested on daily clinical practice, being their analysis generally placed in early stages of drug development. ELISA-type assay the most common method. ADAs detection can involve important implications for treatment strategies of cancer patients, guiding therapeutic adjustment. In oncology, some studies about ADAs synthesis related to targeted therapies and immune checkpoint inhibitors have been recently published. Several strategies are proposed to reduce mAbs immunogenicity, such as different schedules, routes of administration or even the use of immunosuppressants. Another question that arises in relation to ADAs generation is the need to measure the concentration levels of active drug to guide the administration schedule. In this review, we will discuss all the aspects that are currently under discussion in relation with ADAs in oncology.
Collapse
|
21
|
Wieleba I, Wojas-Krawczyk K, Krawczyk P, Milanowski J. Clinical Application Perspectives of Lung Cancers 3D Tumor Microenvironment Models for In Vitro Cultures. Int J Mol Sci 2022; 23:ijms23042261. [PMID: 35216378 PMCID: PMC8876687 DOI: 10.3390/ijms23042261] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the enormous progress and development of modern therapies, lung cancer remains one of the most common causes of death among men and women. The key element in the development of new anti-cancer drugs is proper planning of the preclinical research phase. The most adequate basic research exemplary for cancer study are 3D tumor microenvironment in vitro models, which allow us to avoid the use of animal models and ensure replicable culture condition. However, the question tormenting the scientist is how to choose the best tool for tumor microenvironment research, especially for extremely heterogenous lung cancer cases. In the presented review we are focused to explain the key factors of lung cancer biology, its microenvironment, and clinical gaps related to different therapies. The review summarized the most important strategies for in vitro culture models mimicking the tumor–tumor microenvironmental interaction, as well as all advantages and disadvantages were depicted. This knowledge could facilitate the right decision to designate proper pre-clinical in vitro study, based on available analytical tools and technical capabilities, to obtain more reliable and personalized results for faster introduction them into the future clinical trials.
Collapse
|
22
|
Jawa V, Maamary J, Swanson M, Zhang S, Montgomery D. Implementing a Clinical Immunogenicity Strategy using Preclinical Risk Assessment Outputs. J Pharm Sci 2022; 111:960-969. [PMID: 35122828 DOI: 10.1016/j.xphs.2022.01.032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/27/2022]
Abstract
Immunogenicity to biologics is often observed following dosing in human subjects during clinical trials. Both product and host specific factors may be implicated in contributing to a potential immune response. However, even if such risk factors are identified and eliminated as part of the rational quality by design approaches, the outcome in clinic can be uncertain and challenging to predict. Several tools have been employed to identify these risk factors and consequent mitigation approaches implemented prior to dosing in humans. However, the complexity of the immune system with an interplay of network of immune cells involved in driving a long- term immune response as well as patient characteristics, can make it challenging to predict the outcome in clinic. This perspective will provide an insight into recent advances in the risk assessment approaches that are utilized during preclinical stage of development of a biologic. The outputs from such tools can help to rank order and select the most optimal candidate with the least likelihood of an immune response and can further drive the development of a clinical bioanalytical and immunogenicity monitoring strategy. Such a strategy can be proactively shared with the regulators along with the proposal to streamline clinical immunogenicity and personalizing the outcome based on pharmacogenomics and other patient-related factors. This paper provides a roadmap on performing risk assessments through a systematic identification of risks and their mitigations wherever possible. Recommendations on incorporating the key components of such risk assessments as part of the new regulatory submissions are also provided. Shorter abstract Immunogenicity to biologics is common during clinical trials. Both product and host specific factors have been implicated. Several risk assessment tools can be used to identify and mitigate the risk factors responsible for immunogenicity. An insight into recent advances in the risk assessment approaches will be presented. The outputs can define a risk score and guide the clinical bioanalytical and immunogenicity monitoring strategy. A roadmap on performing risk assessments through a systematic identification of risks and their mitigations wherever possible is provided. Best practices for a risk assessment strategy and recommendations on the content for IND and the Integrated summary of Immunogenicity are also provided.
Collapse
Affiliation(s)
- Vibha Jawa
- Nonclinical Disposition and Bioanalysis, Bristol-Myers Squibb, Princeton, NJ 08648, USA.
| | - Jad Maamary
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Michael Swanson
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Shuli Zhang
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| | - Diana Montgomery
- Pharmacokinetics, Pharmacodynamics & Drug Metabolism (PPDM), Merck & Co., Inc., Kenilworth, NJ 07033 USA
| |
Collapse
|
23
|
Dong Y, Wong JSL, Sugimura R, Lam KO, Li B, Kwok GGW, Leung R, Chiu JWY, Cheung TT, Yau T. Recent Advances and Future Prospects in Immune Checkpoint (ICI)-Based Combination Therapy for Advanced HCC. Cancers (Basel) 2021; 13:1949. [PMID: 33919570 PMCID: PMC8072916 DOI: 10.3390/cancers13081949] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 04/11/2021] [Accepted: 04/16/2021] [Indexed: 12/30/2022] Open
Abstract
Advanced, unresectable hepatocellular carcinoma has a dismal outcome. Multiple immune checkpoint inhibitors (ICIs) targeting the programmed-cell death 1 pathway (PD-1/L1) have been approved for the treatment of advanced HCC. However, outcomes remain undesirable and unpredictable on a patient-to-patient basis. The combination of anti-PD-1/L1 with alternative agents, chiefly cytotoxic T-lymphocyte antigen-4 (CTLA-4) ICIs or agents targeting other oncogenic pathways such as the vascular endothelial growth factor (VEGF) pathway and the c-MET pathway, has, in addition to the benefit of directly targeting alterative oncogenic pathways, in vitro evidence of synergism through altering the genomic and function signatures of T cells and expression of immune checkpoints. Several trials have been completed or are underway evaluating such combinations. Finally, studies utilizing transcriptomics and organoids are underway to establish biomarkers to predict ICI response. This review aims to discuss the biological rationale and clinical advances in ICI-based combinations in HCCs, as well as the progress and prospects of the search for the aforementioned biomarkers in ICI treatment of HCC.
Collapse
Affiliation(s)
- Yawen Dong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
- Department of Surgery, Klinik Favoriten, Wiener Gesundheitsverbund, 1100 Vienna, Austria
| | - Jeffrey Sum Lung Wong
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| | - Ryohichi Sugimura
- School of Biomedical Science, The University of Hong Kong, Hong Kong, China;
| | - Ka-On Lam
- Department of Clinical Oncology, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China;
| | - Bryan Li
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| | - Gerry Gin Wai Kwok
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| | - Roland Leung
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| | - Joanne Wing Yan Chiu
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| | - Tan To Cheung
- Department of Surgery, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China;
| | - Thomas Yau
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, China; (Y.D.); (J.S.L.W.); (B.L.); (G.G.W.K.); (R.L.); (J.W.Y.C.)
| |
Collapse
|
24
|
Labella M, Castells M. Hypersensitivity reactions and anaphylaxis to checkpoint inhibitor-monoclonal antibodies and desensitization. Ann Allergy Asthma Immunol 2021; 126:623-629. [PMID: 33781937 DOI: 10.1016/j.anai.2021.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/07/2021] [Accepted: 03/16/2021] [Indexed: 02/08/2023]
Abstract
OBJECTIVE To review type 1 hypersensitivity reactions and anaphylaxis to checkpoint inhibitor-monoclonal antibodies and its management with drug desensitization. DATA SOURCES English-language literature on MEDLINE regarding hypersensitivity, anaphylaxis, and checkpoint inhibitor-monoclonal antibodies. STUDY SELECTIONS References were selected based on relevance, novelty, robustness, and applicability. RESULTS There are well-known tissue toxicities associated to checkpoint inhibitors, but hypersensitivity reactions and anaphylaxis have been underreported. The presentation of these reactions is based on clinical phenotypes with underlying endotypes identified by specific biomarkers. Drug desensitizations have been successfully applied to checkpoint inhibitor drugs to allow patients with cancer to receive first-line therapies. This review provides current best practices for the recognition and diagnosis of hypersensitivity reactions and anaphylaxis to checkpoint inhibitors and their management using drug desensitization. CONCLUSION Hypersensitivity reactions and anaphylaxis have been identified as potential adverse effects induced by checkpoint inhibitor-monoclonal antibodies. Drug desensitization is a safe and effective treatment option for patients who experience hypersensitivity reactions in need of these monoclonal antibodies to improve cancer outcomes.
Collapse
Affiliation(s)
- Marina Labella
- Allergy Clinical Unit, Hospital Regional Universitario de Málaga, Málaga, Spain; Allergy Research Group, Instituto de Investigación Biomédica de Málaga-IBIMA, Málaga, Spain.
| | - Mariana Castells
- Division of Rheumatology, Immunology, and Allergy, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
25
|
Junker F, Gulati P, Wessels U, Seeber S, Stubenrauch KG, Codarri-Deak L, Markert C, Klein C, Camillo Teixeira P, Kao H. A human receptor occupancy assay to measure anti-PD-1 binding in patients with prior anti-PD-1. Cytometry A 2021; 99:832-843. [PMID: 33704890 PMCID: PMC8451911 DOI: 10.1002/cyto.a.24334] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 02/08/2021] [Accepted: 03/04/2021] [Indexed: 12/19/2022]
Abstract
Receptor occupancy (RO) assessment by flow cytometry is an important pharmacodynamic (PD) biomarker in the clinical development of large molecules such as monoclonal therapeutic antibodies (mAbs). The total‐drug‐bound RO assay format directly assesses mAb binding to cell surface targets using anti‐drug detection antibodies. Here, we generated a flow cytometry detection antibody specifically binding to mAbs of the IgG1 P329GLALA backbone. Using this reagent, we developed a total‐drug‐bound RO assay format for RG7769, a bi‐specific P329GLALA containing mAb targeting PD‐1 and TIM3 on T cells. In its fit‐for‐purpose validated version, this RO assay has been used in the Phase‐I dose escalation study of RG7769, informing on peripheral T cell RO and RG7769 antibody binding capacity (ABC). We assessed RG7769 RO in checkpoint‐inhibitor (CPI) naïve patients and anti‐PD‐1 CPI experienced patients using our novel assay. Here, we show that in both groups, complete T cell RO can be achieved (~100%). However, we found that the maximum number of T cell binding sites for RG7769 pre‐dosing was roughly twofold lower in patients recently having undergone anti‐PD‐1 treatment. We show that this is due to steric hindrance exerted by competing mAbs masking the available drug binding sites. Our findings highlight the importance of quantitative mAb assessment in addition to relative RO especially in the context of patients who have previously received anti‐PD‐1 treatment.
Collapse
Affiliation(s)
- Fabian Junker
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Pratiksha Gulati
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Uwe Wessels
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, F. Hoffmann-La Roche Ltd, Penzberg, Germany
| | - Stefan Seeber
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, F. Hoffmann-La Roche Ltd, Penzberg, Germany
| | - Kay-Gunnar Stubenrauch
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Munich, F. Hoffmann-La Roche Ltd, Penzberg, Germany
| | - Laura Codarri-Deak
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | | | - Christian Klein
- Roche Pharma Research and Early Development, Discovery Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Priscila Camillo Teixeira
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Henry Kao
- Roche Pharma Research and Early Development, Early Biomarker Development Oncology, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| |
Collapse
|
26
|
Nakhoda SK, Olszanski AJ. Addressing Recent Failures in Immuno-Oncology Trials to Guide Novel Immunotherapeutic Treatment Strategies. Pharmaceut Med 2021; 34:83-91. [PMID: 32157638 DOI: 10.1007/s40290-020-00326-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The incorporation of checkpoint inhibitors into the treatment armamentarium of oncologic therapeutics has revolutionized the course of disease in many cancers. This has spurred the evaluation of other novel immunotherapy agents in clinical trials with varying levels of success. This review explores possible explanations for differences in efficacy in clinical outcomes among currently US FDA-approved immunotherapy agents, lessons learned from clinical trial failures of investigational immunotherapies, and methods to improve success in the future. An inherent challenge of early phase immunotherapy trials is identifying the maximum tolerated dose and improving understanding of the pharmacokinetics/pharmacodynamics of immunotherapies as they exert their effects indirectly via T cells rather than directly via dose-dependent cytotoxic activity. The wide heterogeneity of the immune system among patients and within an individual patient over time largely affects the results of optimal dose- and toxicity-finding studies as well as the effectiveness of immunotherapy. Therefore, optimization of phase I/II study design is crucial for clinical trial success. These differences may also help elucidate the lack of immunotherapy benefit in certain disease subtypes despite the presence of specific biomarkers. Broader investigation of the tumor microenvironment and its dynamic nature can help in the identification of alternative pathways for targeted therapies, mechanisms of immunotherapy resistance, and more correlative biomarkers. Finally, manipulation of the tumor microenvironment via a single agonist or antagonist may be inadequate, so combination therapies and sequencing of agents must be further assessed while balancing cumulative toxicity risk.
Collapse
Affiliation(s)
- Shazia K Nakhoda
- Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111, USA
| | | |
Collapse
|
27
|
Sensi M, Berto M, Gentile S, Pinti M, Conti A, Pellacani G, Salvarani C, Cossarizza A, Bortolotti CA, Biscarini F. Anti-drug antibody detection with label-free electrolyte-gated organic field-effect transistors. Chem Commun (Camb) 2021; 57:367-370. [PMID: 33325465 DOI: 10.1039/d0cc03399e] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The efficacy of immunotherapy can be undermined by the development of an immune response against a drug/antibody mediated by anti-drug antibodies (ADAs) in treated patients. We present the first label-free EGOFET immunosensor that integrates a biological drug, Nivolumab (Opdivo©), as a specific recognition moiety to quantitatively and selectively detect ADAs against the drug. The limit of detection is 100 fM. This demonstration is a prelude to the detection of ADAs in a clinical setting in the treatment of different pathologies, and it also enables rapid screening of biological drugs for immunogenicity.
Collapse
Affiliation(s)
- Matteo Sensi
- Dipartimento di Scienze della Vita - Università di Modena e Reggio Emilia, Via Campi 103, 41125 Modena, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Schofield DJ, Percival-Alwyn J, Rytelewski M, Hood J, Rothstein R, Wetzel L, McGlinchey K, Adjei G, Watkins A, Machiesky L, Chen W, Andrews J, Groves M, Morrow M, Stewart RA, Leinster A, Wilkinson RW, Hammond SA, Luheshi N, Dobson C, Oberst M. Activity of murine surrogate antibodies for durvalumab and tremelimumab lacking effector function and the ability to deplete regulatory T cells in mouse models of cancer. MAbs 2021; 13:1857100. [PMID: 33397194 PMCID: PMC7831362 DOI: 10.1080/19420862.2020.1857100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 11/12/2020] [Accepted: 11/24/2020] [Indexed: 12/13/2022] Open
Abstract
Preclinical studies of PD-L1 and CTLA-4 blockade have relied heavily on mouse syngeneic tumor models with intact immune systems, which facilitate dissection of immunosuppressive mechanisms in the tumor microenvironment. Commercially developed monoclonal antibodies (mAbs) targeting human PD-L1, PD-1, and CTLA-4 may not demonstrate cross-reactive binding to their mouse orthologs, and surrogate anti-mouse antibodies are often used in their place to inhibit these immune checkpoints. In each case, multiple choices exist for surrogate antibodies, which differ with respect to species of origin, affinity, and effector function. To develop relevant murine surrogate antibodies for the anti-human PD-L1 mAb durvalumab and the anti-human CTLA-4 mAb tremelimumab, rat/mouse chimeric or fully murine mAbs engineered for reduced effector function were developed and compared with durvalumab and tremelimumab. Characterization included determination of target affinity, in vivo effector function, pharmacokinetic profile, and anti-tumor efficacy in mouse syngeneic tumor models. Results showed that anti-PD-L1 and anti-CTLA-4 murine surrogates with pharmacologic properties similar to those of durvalumab and tremelimumab demonstrated anti-tumor activity in a subset of commonly used mouse syngeneic tumor models. This activity was not entirely dependent on antibody-dependent cellular cytotoxicity, antibody-dependent cellular phagocytosis effector function, or regulatory T-cell depletion, as antibodies engineered to lack these features showed activity in models historically sensitive to checkpoint inhibition, albeit at a significantly lower level than antibodies with intact effector function.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/immunology
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antineoplastic Agents, Immunological/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- B7-H1 Antigen/immunology
- CTLA-4 Antigen/immunology
- Cell Line, Tumor
- Female
- Humans
- Kaplan-Meier Estimate
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Rats, Sprague-Dawley
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- Tumor Burden/drug effects
- Tumor Burden/immunology
- Mice
- Rats
Collapse
Affiliation(s)
- Darren J. Schofield
- Antibody Development and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jennifer Percival-Alwyn
- Antibody Development and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | | | - John Hood
- Clinical and Quantitative Pharmacology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Raymond Rothstein
- Discovery Biosciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Leslie Wetzel
- Discovery Biosciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Kelly McGlinchey
- Translational Medicine Department in Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Grace Adjei
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Amanda Watkins
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | - LeeAnn Machiesky
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Weimin Chen
- Analytical Sciences, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - John Andrews
- Antibody Development and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Maria Groves
- Antibody Development and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Michelle Morrow
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Ross A. Stewart
- Translational Medicine Department in Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Andrew Leinster
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | | | - Scott A. Hammond
- Discovery Biosciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Nadia Luheshi
- Discovery Biosciences, Oncology R&D, AstraZeneca, Cambridge, UK
| | - Claire Dobson
- Antibody Development and Protein Engineering, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Michael Oberst
- Discovery Biosciences, Oncology R&D, AstraZeneca, Gaithersburg, MD, USA
| |
Collapse
|
29
|
Strategies to develop highly drug-tolerant cell-based neutralizing antibody assay: neutralizing antidrug antibodies extraction and drug depletion. Bioanalysis 2020; 12:1279-1293. [PMID: 32945693 DOI: 10.4155/bio-2020-0091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Aim: Drug interference poses great analytical challenges for cell-based neutralizing antidrug antibodies (NAb) assay. The work aimed to improve assay drug tolerance through biotin-drug extraction with acid dissociation method optimization and developing new approach. Results: The NAb extraction with biotin-drug extraction with acid dissociation approach has been optimized by reducing biotinylated drug leaching and improving NAb elution efficiency, resulting in drug tolerance of up to 160 μg/ml. To circumvent the low acid elution efficiency of NAb from drug, a novel drug depletion approach was developed, which combined acid dissociation and drug targeted crosslinked capture, achieved drug tolerance up to 400 μg/ml. At last, a strategy workflow for sample pretreatment approach selection and optimization was established for improving drug tolerance of NAb assay. Conclusion: We demonstrated that reduced biotinylated drug leaching and the high NAb elution efficiency was critical for improving assay drug tolerance. Drug depletion offers an alternative approach to overcome low NAb elution efficiency.
Collapse
|
30
|
Geraud A, Gougis P, Vozy A, Anquetil C, Allenbach Y, Romano E, Funck-Brentano E, Moslehi JJ, Johnson DB, Salem JE. Clinical Pharmacology and Interplay of Immune Checkpoint Agents: A Yin-Yang Balance. Annu Rev Pharmacol Toxicol 2020; 61:85-112. [PMID: 32871087 DOI: 10.1146/annurev-pharmtox-022820-093805] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
T cells have a central role in immune system balance. When activated, they may lead to autoimmune diseases. When too anergic, they contribute to infection spread and cancer proliferation. Immune checkpoint proteins regulate T cell function, including cytotoxic T lymphocyte antigen-4 (CTLA-4) and programmed cell death-1 (PD-1) and its ligand (PD-L1). These nodes of self-tolerance may be exploited pharmacologically to downregulate (CTLA-4 agonists) and activate [CTLA-4 and PD-1/PD-L1 antagonists, also called immune checkpoint inhibitors (ICIs)] the immune system.CTLA-4 agonists are used to treat rheumatologic immune disorders and graft rejection. CTLA-4, PD-1, and PD-L1 antagonists are approved for multiple cancer types and are being investigated for chronic viral infections. Notably, ICIs may be associated with immune-related adverse events (irAEs), which can be highly morbid or fatal. CTLA-4 agonism has been a promising method to reverse such life-threatening irAEs. Herein, we review the clinical pharmacology of these immune checkpoint agents with a focus on their interplay in human diseases.
Collapse
Affiliation(s)
- Arthur Geraud
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France; .,Department of Drug Development (DITEP), Gustave Roussy, 94805 Villejuif, France
| | - Paul Gougis
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France;
| | - Aurore Vozy
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France;
| | - Celine Anquetil
- Sorbonne Université, INSERM, Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France
| | - Yves Allenbach
- Sorbonne Université, INSERM, Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France
| | - Emanuela Romano
- Center for Cancer Immunotherapy, INSERM U932, Institut Curie, 75248 Paris Cedex 05, France
| | - Elisa Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise-Paré Hospital, AP-HP, EA 4340, Université Paris-Saclay, 92100 Boulogne-Billancourt, France
| | - Javid J Moslehi
- Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Douglas B Johnson
- Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | - Joe-Elie Salem
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France; .,Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| |
Collapse
|
31
|
Vaisman-Mentesh A, Gutierrez-Gonzalez M, DeKosky BJ, Wine Y. The Molecular Mechanisms That Underlie the Immune Biology of Anti-drug Antibody Formation Following Treatment With Monoclonal Antibodies. Front Immunol 2020; 11:1951. [PMID: 33013848 PMCID: PMC7461797 DOI: 10.3389/fimmu.2020.01951] [Citation(s) in RCA: 114] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/20/2020] [Indexed: 12/25/2022] Open
Abstract
Monoclonal antibodies (mAbs) are a crucial asset for human health and modern medicine, however, the repeated administration of mAbs can be highly immunogenic. Drug immunogenicity manifests in the generation of anti-drug antibodies (ADAs), and some mAbs show immunogenicity in up to 70% of patients. ADAs can alter a drug's pharmacokinetic and pharmacodynamic properties, reducing drug efficacy. In more severe cases, ADAs can neutralize the drug's therapeutic effects or cause severe adverse events to the patient. While some contributing factors to ADA formation are known, the molecular mechanisms of how therapeutic mAbs elicit ADAs are not completely clear. Accurate ADA detection is necessary to provide clinicians with sufficient information for patient monitoring and clinical intervention. However, ADA assays present unique challenges because both the analyte and antigen are antibodies, so most assays are cumbersome, costly, time consuming, and lack standardization. This review will discuss aspects related to ADA formation following mAb drug administration. First, we will provide an overview of the prevalence of ADA formation and the available diagnostic tools for their detection. Next, we will review studies that support possible molecular mechanisms causing the formation of ADA. Finally, we will summarize recent approaches used to decrease the propensity of mAbs to induce ADAs.
Collapse
Affiliation(s)
- Anna Vaisman-Mentesh
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| | | | - Brandon J. DeKosky
- Department of Pharmaceutical Chemistry, The University of Kansas, Lawrence, KS, United States
- Department of Chemical and Petroleum Engineering, The University of Kansas, Lawrence, KS, United States
| | - Yariv Wine
- George S. Wise Faculty of Life Sciences, School of Molecular Cell Biology and Biotechnology, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
32
|
Lin WW, Lu YC, Chuang CH, Cheng TL. Ab locks for improving the selectivity and safety of antibody drugs. J Biomed Sci 2020; 27:76. [PMID: 32586313 PMCID: PMC7318374 DOI: 10.1186/s12929-020-00652-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/22/2020] [Indexed: 02/06/2023] Open
Abstract
Monoclonal antibodies (mAbs) are a major targeted therapy for malignancies, infectious diseases, autoimmune diseases, transplant rejection and chronic inflammatory diseases due to their antigen specificity and longer half-life than conventional drugs. However, long-term systemic antigen neutralization by mAbs may cause severe adverse events. Improving the selectivity of mAbs to distinguish target antigens at the disease site from normal healthy tissue and reducing severe adverse events caused by the mechanisms-of-action of mAbs is still a pressing need. Development of pro-antibodies (pro-Abs) by installing a protease-cleavable Ab lock is a novel and advanced recombinant Ab-based strategy that efficiently masks the antigen binding ability of mAbs in the normal state and selectively "turns on" the mAb activity when the pro-Ab reaches the proteolytic protease-overexpressed diseased tissue. In this review, we discuss the design and advantages/disadvantages of different Ab lock strategies, focusing particularly on spatial-hindrance-based and affinity peptide-based approaches. We expect that the development of different masking strategies for mAbs will benefit the local reactivity of mAbs at the disease site, increase the therapeutic efficacy and safety of long-term treatment with mAbs in chronic diseases and even permit scientists to develop Ab drugs for formerly undruggable targets and satisfy the unmet medical needs of mAb therapy.
Collapse
Affiliation(s)
- Wen-Wei Lin
- Department of Laboratory Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Yun-Chi Lu
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Biomedical and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan
| | - Chih-Hung Chuang
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Health Sciences, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tian-Lu Cheng
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung, Taiwan.
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan.
- Department of Biomedical and Environmental Biology, Kaohsiung Medical University, 100 Shih-Chuan 1st Road, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
33
|
Sehgal K, Costa DB, Rangachari D. Extended-Interval Dosing Strategy of Immune Checkpoint Inhibitors in Lung Cancer: Will it Outlast the COVID-19 Pandemic? Front Oncol 2020; 10:1193. [PMID: 32714874 PMCID: PMC7344199 DOI: 10.3389/fonc.2020.01193] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Accepted: 06/12/2020] [Indexed: 12/19/2022] Open
Abstract
Patients with lung cancer are particularly vulnerable to complications from coronavirus disease-2019 (COVID-19). Recurrent hospital visits and hospital admission are potential risk factors for acquiring infection with its causative pathogen, severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). As immune checkpoint inhibitors (ICIs) constitute the therapeutic backbone for the vast majority of patients with advanced lung cancer in the absence of actionable driver oncogenes, there have been intense discussions within the oncology community regarding risk-benefit of delaying these treatments or use of alternative extended-interval treatment strategies to minimize the risk of viral transmission secondary to unintended nosocomial exposures. In the midst of the COVID-19 pandemic, the U.S. Food and Drug Administration (FDA) granted accelerated approval for extended-interval strategy of pembrolizumab at a dose of 400 mg every 6 weeks for all already approved oncologic indications. Herein, we summarize the evidence from the in silico pharmacokinetic modeling/simulation studies supporting extended-interval dosing strategies for the ICIs used in lung cancer. We further review the evolving clinical evidence behind these approaches and predict that they will continue to be used in routine practice even long after the pandemic, particularly for patients with durable disease control.
Collapse
Affiliation(s)
- Kartik Sehgal
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States
| | - Daniel B. Costa
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Deepa Rangachari
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
34
|
Ramos-Casals M, Brahmer JR, Callahan MK, Flores-Chávez A, Keegan N, Khamashta MA, Lambotte O, Mariette X, Prat A, Suárez-Almazor ME. Immune-related adverse events of checkpoint inhibitors. Nat Rev Dis Primers 2020; 6:38. [PMID: 32382051 PMCID: PMC9728094 DOI: 10.1038/s41572-020-0160-6] [Citation(s) in RCA: 720] [Impact Index Per Article: 180.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapies have changed the landscape of cancer treatment during the past few decades. Among them, immune checkpoint inhibitors, which target PD-1, PD-L1 and CTLA-4, are increasingly used for certain cancers; however, this increased use has resulted in increased reports of immune-related adverse events (irAEs). These irAEs are unique and are different to those of traditional cancer therapies, and typically have a delayed onset and prolonged duration. IrAEs can involve any organ or system. These effects are frequently low grade and are treatable and reversible; however, some adverse effects can be severe and lead to permanent disorders. Management is primarily based on corticosteroids and other immunomodulatory agents, which should be prescribed carefully to reduce the potential of short-term and long-term complications. Thoughtful management of irAEs is important in optimizing quality of life and long-term outcomes.
Collapse
Affiliation(s)
- Manuel Ramos-Casals
- Department of Autoimmune Diseases, ICMiD, Barcelona, Spain. .,Laboratory of Autoimmune Diseases Josep Font, IDIBAPS-CELLEX, Barcelona, Spain. .,Department of Medicine, University of Barcelona, Hospital Clínic, Barcelona, Spain.
| | - Julie R. Brahmer
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Margaret K. Callahan
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Weill Cornell Medical College, New York, NY, USA,Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Niamh Keegan
- Melanoma and Immunotherapeutics Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA,Weill Cornell Medical College, New York, NY, USA
| | - Munther A. Khamashta
- Lupus Clinic, Rheumatology Department, Dubai Hospital, Dubai, United Arab Emirates
| | - Olivier Lambotte
- APHP Médecine Interne/Immunologie Clinique, Hôpital Bicêtre, Paris, France,Université Paris-Saclay – INSERM U1184 - CEA, Immunology of Viral Infections and Autoimmune Diseases, IDMIT Department, IBFJ, Fontenay-aux-Roses and Le Kremlin-Bicêtre, France
| | - Xavier Mariette
- Université Paris-Saclay, INSERM, CEA, Centre de recherche en Immunologie des infections virales et des maladies auto-immunes ; AP-HP.Université Paris-Saclay, Hôpital Bicêtre, Rheumatology Department, Le Kremlin Bicêtre, France
| | - Aleix Prat
- Translational Genomic and Targeted Therapeutics in Solid Tumors, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain,Department of Medical Oncology, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Maria E. Suárez-Almazor
- Section of Rheumatology/Clinical Immunology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
35
|
Desnoyer A, Broutin S, Delahousse J, Maritaz C, Blondel L, Mir O, Chaput N, Paci A. Pharmacokinetic/pharmacodynamic relationship of therapeutic monoclonal antibodies used in oncology: Part 2, immune checkpoint inhibitor antibodies. Eur J Cancer 2020; 128:119-128. [PMID: 32037060 DOI: 10.1016/j.ejca.2020.01.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/02/2020] [Accepted: 01/07/2020] [Indexed: 10/25/2022]
Abstract
Immune checkpoint inhibitors are monoclonal antibodies (mAbs) directed against negative immunologic regulators that are used to restore the immune response against cancer. Approved drugs include anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4), anti-programmed cell death 1 (PD-1) and anti-programmed cell death-ligand 1 (PD-L1) antibodies exhibiting pharmacokinetic (PK) characteristics typical of mAbs. Most factors such as age, sex, ethnicity, tumour burden, performance status and immunogenicity, but not body weight, do not seem to affect drug clearance clinically. However, an exposure-response relation has been described for both the efficacy and toxicity of anti-CTLA-4 and anti-PD-1 agents. The change in clearance over time is associated with overall response at least for nivolumab and pembrolizumab. Few PK/pharmacodynamic (PD) data are available for anti-PD-L1 mAbs, but time-varying clearance has been described for these drugs, and the high immunogenicity rate observed with atezolizumab may affect PK parameters and should be further studied. These data suggest the need for additional PK/PD studies. In this review, we summarise studies of the PKs of immune checkpoint inhibitors, exploring possible interactions with PD considerations.
Collapse
Affiliation(s)
- Aude Desnoyer
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, Villejuif, F-94805, France.
| | - Sophie Broutin
- Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| | - Julia Delahousse
- Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| | - Christophe Maritaz
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France.
| | - Louis Blondel
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France.
| | - Olivier Mir
- Gustave Roussy Cancer Campus, Department of Ambulatory Care, Villejuif, F-94805, France.
| | - Nathalie Chaput
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Laboratory of Immunomonitoring in Oncology, Villejuif, F-94805, France.
| | - Angelo Paci
- University Paris-Saclay, Faculty of Pharmacy, Chatenay-Malabry, F-92290, France; Gustave Roussy Cancer Campus, Department of Pharmacology, Villejuif, F-94805, France.
| |
Collapse
|
36
|
Enrico D, Paci A, Chaput N, Karamouza E, Besse B. Antidrug Antibodies Against Immune Checkpoint Blockers: Impairment of Drug Efficacy or Indication of Immune Activation? Clin Cancer Res 2019; 26:787-792. [PMID: 31757876 DOI: 10.1158/1078-0432.ccr-19-2337] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 10/07/2019] [Accepted: 11/14/2019] [Indexed: 11/16/2022]
Abstract
The generation of antibodies following exposure to therapeutic drugs has been widely studied, however in oncology, data in relation to their clinical relevance are limited. Antidrug antibodies (ADAs) can cause a decrease in the amount of drug available, resulting in some cases in decreased antitumor activity and a consequent impact on clinical outcomes. Several immunologic factors can influence the development of ADAs, and in addition, the sensitivity of the different testing methods used in different studies can vary, representing an additional potential confounding factor. The reported frequency of ADA-positive patients following treatment with immune checkpoint inhibitors varies from as low as 1.5% for pembrolizumab to 54% for atezolizumab. This latter drug is the only immune checkpoint inhibitor to have undergone an expanded analysis of the clinical implications of ADAs, but with discordant results. Given that immune checkpoint inhibitors can modify the immune response and potentially impact ADA formation, data from published as well as prospective trials need to be evaluated for a better understanding of the clinical implications of ADAs in this setting.
Collapse
Affiliation(s)
- Diego Enrico
- Gustave Roussy Cancer Campus, Villejuif, France.,Medical Oncology Department, Gustave Roussy Cancer Center, Villejuif, France
| | - Angelo Paci
- Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.,Laboratory of Pharmacology and Drug Analysis, Gustave Roussy Cancer Center, Villejuif, France
| | - Nathalie Chaput
- Gustave Roussy Cancer Campus, Villejuif, France.,Faculty of Pharmacy, University Paris-Saclay, Chatenay-Malabry, France.,Laboratory for Immunomonitoring in Oncology, UMS 3655 CNRS/US 23 INSERM, Gustave Roussy Cancer Campus, Villejuif, France
| | - Eleni Karamouza
- Gustave Roussy Cancer Campus, Villejuif, France.,Ligue Nationale Contre le Cancer Meta-Analysis Platform, Biostatistics and Epidemiology Unit, INSERM U1018, CESP, Villejuif, France
| | - Benjamin Besse
- Gustave Roussy Cancer Campus, Villejuif, France. .,Medical Oncology Department, Gustave Roussy Cancer Center, Villejuif, France.,Faculty of Medicine, University Paris-Saclay, Le Kremlin-Bicêtre, France
| |
Collapse
|
37
|
Is there an Exposure-Response Relationship for Nivolumab in Real-World NSCLC Patients? Cancers (Basel) 2019; 11:cancers11111784. [PMID: 31766292 PMCID: PMC6895963 DOI: 10.3390/cancers11111784] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 11/08/2019] [Indexed: 11/26/2022] Open
Abstract
Pharmacokinetic/pharmacodynamic data from real-world cohort are sparse in non small–cell lung cancer (NSCLC) patients treated with nivolumab. The aim of this prospective observational study was to explore the exposure-response relationship for effectiveness and toxicity of nivolumab in 81 outpatients with metastatic lung cancer. Nivolumab plasma trough concentrations (Cmin) were assayed at days 14, 28, and 42. Prognostic factors (including Cmin) regarding progression-free survival (PFS) and overall survival (OS) were explored using a multivariate Cox model. A Spearman’s rank test was used to investigate the relationship between Cmin and grade >2 immune-related adverse events (irAE). Mean nivolumab Cmin was 16.2 ± 6.0 µg/mL (n = 76), 25.6 ± 10.2 µg/mL (n = 64) and 33.4 ± 11.3 µg/mL (n = 53) at days 14, 28, and 42, respectively. No pharmacokinetic/pharmacodynamic (PK/PD) relationship was observed with either survival or onset of irAE. Multivariable Cox regression analysis identified Eastern Cooperative Oncology Group Performance Status (hazard ratio 1.85, 95%confidence interval 1.02–3.38, p-value = 0.043) and baseline use of corticosteroids (HR 8.08, 95%CI 1.78–36.62, p-value = 0.007) as independent risk factor for PFS and only baseline use of corticosteroids (HR 6.29, 95%CI 1.46–27.08, p-value = 0.013) for OS. No PK/PD relationship for nivolumab was observed in real-world NSCLC patients. This supports the recent use of flat dose regimens without plasma drug monitoring.
Collapse
|
38
|
Current In Vitro Assays for Prediction of T Cell Mediated Immunogenicity of Biotherapeutics and Manufacturing Impurities. J Pharm Innov 2019. [DOI: 10.1007/s12247-019-09412-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
39
|
Ferreira M, Pichon E, Carmier D, Bouquet E, Pageot C, Bejan-Angoulvant T, Campana M, Vermes E, Marchand-Adam S. Coronary Toxicities of Anti-PD-1 and Anti-PD-L1 Immunotherapies: a Case Report and Review of the Literature and International Registries. Target Oncol 2019; 13:509-515. [PMID: 30006825 DOI: 10.1007/s11523-018-0579-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Immunotherapy medications that target programmed death 1 protein (PD-1) and programmed death-ligand 1 (PD-L1), such as nivolumab, pembrolizumab, and atezolizumab, are currently used in the first- or second-line treatment of non-small cell lung cancers, among other indications. However, these agents are associated with immune-related side effects, the most common of which are endocrinopathies, colitis, hepatitis, and interstitial pneumonitis. In contrast, coronary toxicities are rarely reported and remain poorly understood. Here, we describe the case of a patient who developed an acute coronary syndrome when treated with nivolumab as second-line therapy for metastatic pulmonary adenocarcinoma. A review of the literature, the French pharmacovigilance registry, and the World Health Organization pharmacovigilance database led to the identification of four cases of patients with coronary manifestations attributable to anti-PD1 immunotherapy (with no reported cases of patients undergoing anti-PD-L1 immunotherapy), which we describe herein. The potential mechanisms causing adverse coronary reactions to this type of therapy, which is used to treat lung cancer as well as other solid and hematological neoplastic diseases, are also discussed.
Collapse
Affiliation(s)
- Marion Ferreira
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France. .,, Impasse du Clos Simon, Building A, Apartment 36, 37520, La Riche, France.
| | - Eric Pichon
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France
| | - Delphine Carmier
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France
| | - Emilie Bouquet
- Department of Clinical Pharmacology and Regional Pharmacovigilance Center, University Hospital of Tours, Tours, France
| | - Cécile Pageot
- Regional Pharmacovigilance Center of Bordeaux, Bordeaux, France
| | - Theodora Bejan-Angoulvant
- Medical Pharmacology Department, University Hospital of Tours, Tours, France.,GICC UMR CNRS 7292, Faculty of Medicine of Tours, François Rabelais University, Tours, France
| | - Marion Campana
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France
| | - Emmanuelle Vermes
- Department of Radiology, University Hospital of Tours, Tours, France
| | - Sylvain Marchand-Adam
- Department of Pneumology and Respiratory Functional Exploration, University Hospital of Tours, Tours, France.,INSERM U-1100, Faculty of Medicine of Tours, François Rabelais University, Tours, France
| |
Collapse
|
40
|
Wang J, Fei K, Jing H, Wu Z, Wu W, Zhou S, Ni H, Chen B, Xiong Y, Liu Y, Peng B, Yu D, Jiang H, Liu J. Durable blockade of PD-1 signaling links preclinical efficacy of sintilimab to its clinical benefit. MAbs 2019; 11:1443-1451. [PMID: 31402780 PMCID: PMC6816392 DOI: 10.1080/19420862.2019.1654303] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Blockade of immune checkpoint pathways by programmed cell death protein 1 (PD-1) antibodies has demonstrated broad clinical efficacy against a variety of malignancies. Sintilimab, a highly selective, fully human monoclonal antibody (mAb), blocks the interaction of PD-1 and its ligands and has demonstrated clinical benefit in various clinical studies. Here, we evaluated the affinity of sintilimab to human PD-1 by surface plasmon resonance and mesoscale discovery and evaluated PD-1 receptor occupancy and anti-tumor efficacy of sintilimab in a humanized NOD/Shi-scid-IL2rgamma (null) (NOG) mouse model. We also assessed the receptor occupancy and immunogenicity of sintilimab from clinical studies in humans (9 patients with advanced solid tumor and 381 patients from 4 clinical studies, respectively). Sintilimab bound to human PD-1 with greater affinity than nivolumab (Opdivo®, MDX-1106) and pembrolizumab (Keytruda®, MK-3475). The high affinity of sintilimab is explained by its distinct structural binding mode to PD-1. The pharmacokinetic behavior of sintilimab did not show any significant differences compared to the other two anti-PD-1 mAbs. In the humanized NOG mouse model, sintilimab showed superior PD-1 occupancy on circulating T cells and a stronger anti-tumor effect against NCI-H292 tumors. The strong anti-tumor response correlated with increased interferon-γ-secreting, tumor-specific CD8+ T cells, but not with CD4+ Tregs in tumor tissue. Pharmacodynamics testing indicated a sustained mean occupancy of ≥95% of PD-1 molecules on circulating T cells in patients following sintilimab infusion, regardless of infusion dose. Sintilimab infusion was associated with 0.52% (2/381 patients) of anti-drug antibodies and 0.26% (1/381 patients) neutralizing antibodies. These data validate sintilimab as a novel, safe, and efficacious anti-PD-1 mAb for cancer immunotherapy.
Collapse
Affiliation(s)
- Jie Wang
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Keke Fei
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Hua Jing
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Zhihai Wu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Weiwei Wu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Shuaixiang Zhou
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Haiqing Ni
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Bingliang Chen
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Yan Xiong
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Yanpeng Liu
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Bo Peng
- Department of Translational Science, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Dechao Yu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| | - Haiping Jiang
- Department of Medical Oncology, First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou , Zhejiang Province , China
| | - Junjian Liu
- Department of Biologics Discovery, Innovent Biologics (Suzhou) Co , Jiangsu , China
| |
Collapse
|
41
|
Guo W, Liu L, Xiang C, Chen J, Liang XJ. Engineered nanoparticles circumvent the adaptive treatment tolerance to immune-checkpoint blockade therapy. Sci China Chem 2019. [DOI: 10.1007/s11426-019-9588-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
42
|
Development of an Enzyme-Linked Immune Sorbent Assay to Measure Nivolumab and Pembrolizumab Serum Concentrations. Ther Drug Monit 2019; 40:596-601. [PMID: 29847460 DOI: 10.1097/ftd.0000000000000534] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Treatment with monoclonal antibodies (mAbs) against programmed cell death protein 1 receptor is subject to high variation in treatment outcome among cancer patients. For these agents, no exposure-response (ER) relationships have been investigated in routine health care settings. However, ER relationships have been identified for several other mAbs used in oncology. Methods to conveniently measure serum concentrations of anti-programmed cell death protein 1 mAbs in routine health care may clarify possible ER relationships. Therefore, the authors aimed to develop an enzyme-linked immune sorbent assay (ELISA) for the measurement of both nivolumab and pembrolizumab serum concentrations of treated cancer patients. METHODS Optimal capture antigen and detection antibody concentrations were selected based on titrations. Nivolumab calibration standards ranging from 0.2 to 300 ng/mL were tested in duplicate. Accuracy was assessed in 2 recovery experiments. Intra- and interassay variations were assessed on 3 different days by 2 independent technicians. The developed ELISA was also set up for pembrolizumab calibration curves. Cross-reactivity of nivolumab measurements with ipilimumab was assessed. Of one nivolumab treated patient, serum concentrations in follow up samples were measured and presented. RESULTS Nivolumab calibration standards of 0.20-25 ng/mL were used. Nivolumab trough concentrations after 1 cycle in 8 patients ranged from 17.3 to 31.1 mcg/mL. The range of accuracy was 84%-105%, whereas intra- and interassay variations showed a coefficient of variation of 5.5% and 10.1%, respectively. No cross-reactivity with ipilimumab was detected. Pembrolizumab trough concentrations (n = 8) ranged from 9.1 to 19.7 mcg/mL after 1 infusion. CONCLUSIONS The in-house-developed ELISA provides the opportunity to measure both nivolumab and pembrolizumab serum concentrations. This may help identify possible ER relationships in treated cancer patients and may potentially lead to dose adjustments in the future.
Collapse
|
43
|
Davda J, Declerck P, Hu-Lieskovan S, Hickling TP, Jacobs IA, Chou J, Salek-Ardakani S, Kraynov E. Immunogenicity of immunomodulatory, antibody-based, oncology therapeutics. J Immunother Cancer 2019; 7:105. [PMID: 30992085 PMCID: PMC6466770 DOI: 10.1186/s40425-019-0586-0] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 04/01/2019] [Indexed: 12/18/2022] Open
Abstract
The increasing use of multiple immunomodulatory (IMD) agents for cancer therapies (e.g. antibodies targeting immune checkpoints, bispecific antibodies, and chimeric antigen receptor [CAR]-T cells), is raising questions on their potential immunogenicity and effects on treatment. In this review, we outline the mechanisms of action (MOA) of approved, antibody-based IMD agents, potentially related to their immunogenicity, and discuss the reported incidence of anti-drug antibodies (ADA) as well as their clinical relevance in patients with cancer. In addition, we discuss the impact of the administration route and potential strategies to reduce the incidence of ADA and manage treated patients. Analysis of published reports indicated that the risk of immunogenicity did not appear to correlate with the MOA of anti-programmed death 1 (PD-1)/PD-ligand 1 monoclonal antibodies nor to substantially affect treatment with most of these agents in the majority of patients evaluated to date. Treatment with B-cell depleting agents appears associated with a low risk of immunogenicity. No significant difference in ADA incidence was found between the intravenous and subcutaneous administration routes for a panel of non-oncology IMD antibodies. Additionally, while the data suggest a higher likelihood of immunogenicity for antibodies with T-cell or antigen-presenting cell (APC) targets versus B-cell targets, it is possible to have targets expressed on APCs or T cells and still have a low incidence of immunogenicity.
Collapse
Affiliation(s)
| | | | | | | | - Ira A Jacobs
- Pfizer, 219 East 42nd Street, New York, NY, 10017-5755, USA.
| | | | | | | |
Collapse
|
44
|
Safety and activity of ibrutinib in combination with nivolumab in patients with relapsed non-Hodgkin lymphoma or chronic lymphocytic leukaemia: a phase 1/2a study. LANCET HAEMATOLOGY 2019; 6:e67-e78. [DOI: 10.1016/s2352-3026(18)30217-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 11/15/2018] [Accepted: 11/16/2018] [Indexed: 12/14/2022]
|
45
|
Cusato J, Genova C, Tomasello C, Carrega P, Ottonello S, Pietra G, Mingari MC, Cossu I, Rijavec E, Leggieri A, Di Perri G, Dal Bello MG, Coco S, Boccardo S, Ferlazzo G, Grossi F, D'Avolio A. Influence of Vitamin D in Advanced Non-Small Cell Lung Cancer Patients Treated with Nivolumab. Cancers (Basel) 2019; 11:cancers11010125. [PMID: 30669662 PMCID: PMC6357025 DOI: 10.3390/cancers11010125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 01/12/2019] [Accepted: 01/16/2019] [Indexed: 01/08/2023] Open
Abstract
Nivolumab is one of the most commonly used monoclonal antibodies for advanced non-small cell lung cancer treatment, to the extent that the presence of its anti-antibody is considered a negative prognostic factor. Vitamin D (VD) modulates expression of the genes involved in drug metabolism and elimination. Immune system regulation and immunodeficiency is frequent in non-small cell lung cancer patients. To date, no data have been reported about the relationship between nivolumab and VD. The aim of this study was to quantify plasma 25-hydroxyVD (25-VD) and 1,25-VD, nivolumab, and its anti-antibody before starting treatment (baseline) and at 15, 45 and 60 days of therapy. VD-pathway-associated gene single nucleotide polymorphisms (SNPs) were also evaluated. Molecules were quantified through enzyme-linked immunosorbent assay, and SNPs through real-time PCR. Forty-five patients were enrolled. Median nivolumab concentrations were 12.5 μg/mL, 22.3 μg/mL and 27.1 μg/mL at 15, 45 and 60 days respectively. No anti-nivolumab antibodies were found. Correlations were observed between nivolumab concentrations and 25-VD levels. Nivolumab concentrations were affected by VD-pathway-related gene SNPs. VDBP AC/CC genotype and baseline 25-VD < 10 ng/mL predicted a nivolumab concentration cut-off value of <18.7 μg/mL at 15 days, which was associated with tumor progression. This is the first study showing VD marker predictors of nivolumab concentrations in a real-life context of non-small cell lung cancer treatment.
Collapse
Affiliation(s)
- Jessica Cusato
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy.
| | - Carlo Genova
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Cristina Tomasello
- S.C. Farmacie Ospedaliere-Ospedale M.Vittoria-ASL Città di Torino, 10143 Turin, Italy.
| | - Paolo Carrega
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, 98125 Messina, Italy.
- Cell Factory Center, University of Messina, 98125 Messina, Italy.
| | - Selene Ottonello
- Department of Experimental Medicine (DiMES), University of Genoa, 16132 Genoa, Italy.
- Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genoa, Italy.
| | - Gabriella Pietra
- Department of Experimental Medicine (DiMES), University of Genoa, 16132 Genoa, Italy.
- Immunology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Maria Cristina Mingari
- Department of Experimental Medicine (DiMES), University of Genoa, 16132 Genoa, Italy.
- Center of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genoa, Italy.
- Immunology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Irene Cossu
- Giannina Gaslini Institute, Via Gerolamo Gaslini, 5, 16147 Genova, Italy.
| | - Erika Rijavec
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Anna Leggieri
- S.C. Farmacie Ospedaliere-Ospedale M.Vittoria-ASL Città di Torino, 10143 Turin, Italy.
| | - Giovanni Di Perri
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy.
| | | | - Simona Coco
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Simona Boccardo
- Lung Cancer Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Guido Ferlazzo
- Laboratory of Immunology and Biotherapy, Department of Human Pathology, University of Messina, 98125 Messina, Italy.
- Cell Factory Center, University of Messina, 98125 Messina, Italy.
- Division of Clinical Pathology, University Hospital Policlinico G. Martino, 98125 Messina, Italy.
| | - Francesco Grossi
- Medical Oncology Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy.
| | - Antonio D'Avolio
- Department of Medical Sciences, University of Turin, Amedeo di Savoia Hospital, 10149 Turin, Italy.
- Interdepartmental Center for Clinical and Experimental Pharmacology (CIFACS), University of Turin, 10149 Turin, Italy.
| |
Collapse
|
46
|
Basak EA, Koolen SLW, Hurkmans DP, Schreurs MWJ, Bins S, Oomen-de Hoop E, Wijkhuijs AJM, Besten ID, Sleijfer S, Debets R, van der Veldt AAM, Aerts JGJV, Mathijssen RHJ. Correlation between nivolumab exposure and treatment outcomes in non-small-cell lung cancer. Eur J Cancer 2019; 109:12-20. [PMID: 30654225 DOI: 10.1016/j.ejca.2018.12.008] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/02/2018] [Accepted: 12/05/2018] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Nivolumab treatment is subject to large interpatient variability in both efficacy and toxicity, which may partly be explained by differences in nivolumab exposure. Exposure-response relationships in regular healthcare have not been extensively investigated for nivolumab. Therefore, we aimed to identify possible exposure-response relationships in nivolumab-treated patients with non-small-cell lung cancer (NSCLC). METHODS Patients with NSCLC who started second-line nivolumab therapy (3 mg/kg Q2W) between May 5th 2016 and August 1st 2017, and from whom serial blood samples, toxicity data and outcome data were prospectively collected, were included. Follow-up was carried out until November 1st 2017. Patients were classified according to the best overall response (BOR) based on the Response Evaluation Criteria in Solid Tumours, v1.1, and toxicities according to the Common Terminology Criteria for Adverse Events. Nivolumab trough concentrations were measured after 2, 4 and 10 weeks of treatment, excluding dose delays, and calculated geometric means were tested versus BOR or toxicity using analysis of variance and an independent samples t-test, respectively. Overall survival (OS) and progression-free survival were compared between high and low trough concentration groups. RESULTS Seventy-six patients were evaluable for analyses. Responders (n = 15) had higher mean trough concentrations than patients with progression (n = 33): 47% higher after 2 weeks (p = 0.001), 53% higher after 4 weeks (p = 0.008) and 73% higher after 10 weeks (p = 0.002). Higher trough concentrations were associated with longer OS (p = 0.001). CONCLUSIONS This study shows that patients with NSCLC with a response to nivolumab had a higher nivolumab exposure than patients with progression, indicating a potential exposure-response relationship. Further clinical research should focus on clarifying these exposure-response relationships.
Collapse
Affiliation(s)
- Edwin A Basak
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands.
| | - Stijn L W Koolen
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands; Dept. of Hospital Pharmacy, Erasmus MC, University Medical Centre, Rotterdam, the Netherlands
| | - Daan P Hurkmans
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Marco W J Schreurs
- Dept. of Immunology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Sander Bins
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Esther Oomen-de Hoop
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Ilse den Besten
- Dept. of Pulmonology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Stefan Sleijfer
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Reno Debets
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | | | - Joachim G J V Aerts
- Dept. of Pulmonology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Ron H J Mathijssen
- Dept. of Medical Oncology, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| |
Collapse
|
47
|
Bitoun S, Nocturne G, Ly B, Krzysiek R, Roques P, Pruvost A, Paoletti A, Pascaud J, Dönnes P, Florence K, Gleizes A, Hincelin-Mery A, Allez M, Hacein-Bey-Abina S, Mackay F, Pallardy M, Le Grand R, Mariette X. Methotrexate and BAFF interaction prevents immunization against TNF inhibitors. Ann Rheum Dis 2018; 77:1463-1470. [PMID: 29936438 DOI: 10.1136/annrheumdis-2018-213403] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 06/06/2018] [Accepted: 06/08/2018] [Indexed: 11/03/2022]
Abstract
OBJECTIVES TNF inhibitors (TNFi) can induce anti-drug antibodies (ADA) in patients with autoimmune diseases (AID) leading to clinical resistance. We explored a new way of using methotrexate (MTX) to decrease this risk of immunisation. METHODS We treated BAFF transgenic (BAFFtg) mice, a model of AID in which immunisation against biologic drugs is high, with different TNFi. We investigated the effect of a single course of MTX during the first exposure to TNFi. Wild-type (WT) and BAFFtg mice were compared for B-Cell surface markers involved in MTX-related purinergic metabolism, adenosine production and regulatory B-cells (Bregs).We translated the study to macaques and patients with rheumatoid arthritis from the ABIRISK cohort to determine if there was an interaction between serum BAFF levels and MTX that prevented immuniation. RESULTS In BAFFtg but not in WT mice or macaques, a single course of MTX prevented immunisation against TNFi and maintained drug concentration for over 52 weeks. BAFFtg mice B-cells expressed more CD73 and CD39 compared to WT mice. MTX induced adenosine release from B cells and increased Bregs and precursors. Use of CD73 blocking antibodies reversed MTX-induced tolerance. In patients from the ABIRISK cohort treated with TNFi for chronic inflammatory diseases, high BAFF serum level correlated with absence of ADA to TNFi only in patients cotreated with MTX but not in patients on TNFi monotherapy. CONCLUSION MTX and BAFF interact in mice where CD73, adenosine and regulatory B cells were identified as key actors in this phenomenon. MTX and BAFF also interact in patients to prevent ADA formation.
Collapse
Affiliation(s)
- Samuel Bitoun
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Gaetane Nocturne
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Bineta Ly
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Roman Krzysiek
- Clinical Immunology Laboratory, Hôpitaux Universitaires Paris-Sud, Hôpital Kremlin Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Immunoregulation, Chemokines and Viral Persistence, INSERM, Université Paris-Sud, Clamart, France
| | - Pierre Roques
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure CEA-Université Paris-Sud-INSERM U1184, Fontenay-Aux-Roses, France
| | - Alain Pruvost
- Service de Pharmacologie et Immunoanalyse (SPI), Plateforme SMArt-MS, CEA, INRA, Université Paris-Sud, Gif-sur-Yvette Cedex, France
| | - Audrey Paoletti
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| | - Juliette Pascaud
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| | | | - Kimberly Florence
- Immunogenicity and Clinical Immunology, GlaxoSmithKline, King of Prussia, Pennsylvania, USA
| | - Aude Gleizes
- Clinical Immunology Laboratory, Hôpitaux Universitaires Paris-Sud, Hôpital Kremlin Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,Inflammation, Chimiokines et Immunopathologie, INSERM, Fac de pharmacie-Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Agnes Hincelin-Mery
- Translational Medecine and Clinical Pharmacology Department, Sanofi, Chilly Mazarin, France
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, AP-HP, Université Paris-Diderot, Paris, France
| | - Salima Hacein-Bey-Abina
- Clinical Immunology Laboratory, Hôpitaux Universitaires Paris-Sud, Hôpital Kremlin Bicêtre, Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,UTCBS, CNRS UMR 8258, INSERM U1022, Faculté de Pharmacie de Paris, Université Paris Descartes, Paris, France
| | - Fabienne Mackay
- Department of Immunology, Monash University Central Clinical School, Alfred Medical Research and Education Precinct (AMREP), Melbourne, Victoria, Australia
| | - Marc Pallardy
- LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France.,Inflammation, Chimiokines et Immunopathologie, INSERM, Fac de pharmacie-Université Paris-Sud, Université Paris-Saclay, Châtenay-Malabry, France
| | - Roger Le Grand
- Immunology of Viral Infections and Autoimmune Diseases, IDMIT Infrastructure CEA-Université Paris-Sud-INSERM U1184, Fontenay-Aux-Roses, France
| | - Xavier Mariette
- Rheumatology Department, Université Paris-Sud-CEA-INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, Hôpitaux Universitaires Paris-Sud-Assistance Publique-Hôpitaux de Paris (AP-HP), Le Kremlin Bicêtre, France.,LabEX LERMIT, Université Paris-Sud, Le Kremlin Bicêtre, France
| |
Collapse
|
48
|
Passey C, Suryawanshi S, Sanghavi K, Gupta M. Reporting, Visualization, and Modeling of Immunogenicity Data to Assess Its Impact on Pharmacokinetics, Efficacy, and Safety of Monoclonal Antibodies. AAPS JOURNAL 2018; 20:35. [PMID: 29484520 DOI: 10.1208/s12248-018-0194-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/17/2018] [Indexed: 12/21/2022]
Abstract
The rapidly increasing number of therapeutic biologics in development has led to a growing recognition of the need for improvements in immunogenicity assessment. Published data are often inadequate to assess the impact of an antidrug antibody (ADA) on pharmacokinetics, safety, and efficacy, and enable a fully informed decision about patient management in the event of ADA development. The recent introduction of detailed regulatory guidance for industry should help address many past inadequacies in immunogenicity assessment. Nonetheless, careful analysis of gathered data and clear reporting of results are critical to a full understanding of the clinical relevance of ADAs, but have not been widely considered in published literature to date. Here, we review visualization and modeling of immunogenicity data. We present several relatively simple visualization techniques that can provide preliminary information about the kinetics and magnitude of ADA responses, and their impact on pharmacokinetics and clinical endpoints for a given therapeutic protein. We focus on individual sample- and patient-level data, which can be used to build a picture of any trends, thereby guiding analysis of the overall study population. We also discuss methods for modeling ADA data to investigate the impact of immunogenicity on pharmacokinetics, efficacy, and safety.
Collapse
Affiliation(s)
- Chaitali Passey
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Satyendra Suryawanshi
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Kinjal Sanghavi
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA
| | - Manish Gupta
- Clinical Pharmacology & Pharmacometrics, Bristol-Myers Squibb, Princeton, New Jersey, USA.
| |
Collapse
|
49
|
Kverneland AH, Enevold C, Donia M, Bastholt L, Svane IM, Nielsen CH. Development of anti-drug antibodies is associated with shortened survival in patients with metastatic melanoma treated with ipilimumab. Oncoimmunology 2018; 7:e1424674. [PMID: 29721387 PMCID: PMC5927482 DOI: 10.1080/2162402x.2018.1424674] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 12/31/2017] [Accepted: 01/03/2018] [Indexed: 11/03/2022] Open
Abstract
Introduction: Checkpoint inhibitors, including the CTLA-4 blocking antibody ipilimumab, have become the new standard therapy for many metastatic cancers. Development of anti-drug antibodies (ADAs) after treatment with other biopharmaceuticals has been thoroughly described, but the induction of ADAs after treatment with checkpoint inhibitors has been inadequately investigated. In this retrospective study, we relate ipilimumab serum levels and anti-ipilimumab antibody levels to clinical outcomes in patients with metastatic melanoma (MM). Method: Serum samples from 31 patients with MM were analyzed for serum levels of ipilimumab and ADAs to ipilimumab at baseline, and before the 2nd and 4th infusion using an in-house bead-based assay. The results were correlated with progression-free survival (PFS) and overall survival (OS). Results: Low serum levels of ipilimumab before the 2nd infusion correlated significantly with a shorter OS (p = 0.01) and PFS (p = 0.02). Eight patients (26%) were ADA-positive at either timepoint. ADA positivity correlated significantly with a shorter OS (p = 0.03) with a hazard ratio (HR) of 3.0 (95% CI: 1.2-7.8). Four of 8 ADA-positive patients (50%) discontinued therapy before the 4th infusion due to disease progression, compared to three of 23 (13%) ADA-negative patients. Conclusion: We confirm that low serum levels of ipilimumab are associated with a shortened OS, and we show for the first time that ADAs to ipilimumab are associated with shorter OS in patients with MM.
Collapse
Affiliation(s)
- Anders H. Kverneland
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
- Center for Cancer Immune Therapy, Department of Oncology and Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Marco Donia
- Center for Cancer Immune Therapy, Department of Oncology and Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Lars Bastholt
- Department of Oncology, Odense University Hospital, Odense, Denmark
| | - Inge Marie Svane
- Center for Cancer Immune Therapy, Department of Oncology and Hematology, Copenhagen University Hospital, Herlev, Denmark
| | - Claus H. Nielsen
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
50
|
Abstract
The incidence of melanoma continues to rise worldwide. Prior to 2010, there had been no progress in the treatment of advanced melanoma in living memory. Since then, immunotherapy has become a standard of care in the treatment of advanced melanoma. Nivolumab is a fully human monoclonal antibody against PD-1, which is a negative regulatory checkpoint in the T cells. The clinical benefit of nivolumab as a single agent is well established, with response rates of ≥40%, durable responses and a favorable tolerability profile. The combination of nivolumab and ipilimumab has also become a standard of care and the role of nivolumab in the adjuvant setting for high-risk patients has been recently confirmed.
Collapse
Affiliation(s)
- Fabio Gomes
- The Christie NHS Foundation Trust, Manchester, UK
| | | | - Paul Lorigan
- The Christie NHS Foundation Trust, Manchester, UK
| |
Collapse
|