Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Nature ; 614(7948): 539-547, 2023 02.
Article in English | MEDLINE | ID: mdl-36725933

ABSTRACT

Antibody responses during infection and vaccination typically undergo affinity maturation to achieve high-affinity binding for efficient neutralization of pathogens1,2. Similarly, high affinity is routinely the goal for therapeutic antibody generation. However, in contrast to naturally occurring or direct-targeting therapeutic antibodies, immunomodulatory antibodies, which are designed to modulate receptor signalling, have not been widely examined for their affinity-function relationship. Here we examine three separate immunologically important receptors spanning two receptor superfamilies: CD40, 4-1BB and PD-1. We show that low rather than high affinity delivers greater activity through increased clustering. This approach delivered higher immune cell activation, in vivo T cell expansion and antitumour activity in the case of CD40. Moreover, an inert anti-4-1BB monoclonal antibody was transformed into an agonist. Low-affinity variants of the clinically important antagonistic anti-PD-1 monoclonal antibody nivolumab also mediated more potent signalling and affected T cell activation. These findings reveal a new paradigm for augmenting agonism across diverse receptor families and shed light on the mechanism of antibody-mediated receptor signalling. Such affinity engineering offers a rational, efficient and highly tuneable solution to deliver antibody-mediated receptor activity across a range of potencies suitable for translation to the treatment of human disease.


Subject(s)
Antibodies, Monoclonal , Antibody Affinity , Immunomodulation , Humans , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , CD40 Antigens/drug effects , CD40 Antigens/immunology , Immunomodulation/drug effects , Immunomodulation/immunology , Nivolumab/immunology , Nivolumab/pharmacology
2.
BMC Pulm Med ; 21(1): 373, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34781910

ABSTRACT

BACKGROUND: Nivolumab is used for the treatment of malignant pleural mesothelioma (MPM). However, immune-related adverse events (irAEs) occur in patients treated with nivolumab. Several studies have reported the correlation between irAEs and therapeutic effects of immune checkpoint inhibitor, but none have reported the correlation in MPM. Here we report a retrospective study which shows the correlation between irAEs and therapeutic effects of nivolumab in patients with MPM. METHODS: This study included patients treated with nivolumab at Tokushima University Hospital from February 2009 to September 2021. We retrospectively reviewed the medical records to evaluate the several clinical factors, such as the presence or absence of irAEs, their severities, progression-free survival (PFS), overall survival (OS) or objective response to the treatment. RESULTS: Eleven patients received treatment with nivolumab. Objective response rate was 18.2% and the disease control rate was 90.9%. Median PFS was 6.8 months (95% confidence interval, 1.3 to 11.9 months) and median OS was 15.2 months (95% confidence interval, 8.9 to 21.5 months). IrAEs occurred in eight patients (72.7%), and grade ≥ 2 irAEs occurred in six patients (54.5%). PFS and OS were significantly longer in the grade ≥ 2 irAEs group than in grade < 2 irAEs group (median PFS 13.6 vs. 3.8 months, p = 0.0093; median OS not reached vs. 8.6 months, p = 0.0108). CONCLUSIONS: This is the first study to report the correlation between irAEs and therapeutic effects in patients with MPM. Because the presence of irAEs may be associated with a favorable clinical outcome, early detection and appropriate management of irAEs will increase the therapeutic benefits to patients.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Immune System Diseases/chemically induced , Mesothelioma, Malignant/drug therapy , Nivolumab/adverse effects , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , Female , Humans , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/mortality , Middle Aged , Nivolumab/immunology , Nivolumab/therapeutic use , Retrospective Studies , Survival Analysis , Treatment Outcome
3.
AAPS J ; 23(6): 109, 2021 10 04.
Article in English | MEDLINE | ID: mdl-34608545

ABSTRACT

Monoclonal antibodies (mAbs) are a leading class of biotherapeutics. In oncology, patients often fail on early lines of biologic therapy to a specific target. Some patients may then enroll in a new clinical trial with a mAb specific for the same target. Therefore, immunoassays designed to quantify the current mAb therapy or assess immunogenicity to the drug may be susceptible to cross-reactivity or interference with residual prior biologics. The impact of two approved anti-PD-1 mAbs, pembrolizumab and nivolumab, was tested in several immunoassays for cemiplimab, another approved anti-PD-1 mAb. The methods included a target-capture drug concentration assay, a bridging anti-drug antibody (ADA) assay and a competitive ligand-binding neutralizing antibody (NAb) assay. We also tested bioanalytical strategies to mitigate cross-reactivity or interference in these assays from other anti-PD-1 biologics. Both pembrolizumab and nivolumab cross-reacted in the cemiplimab drug concentration assay. This was mitigated by addition of antibodies specific to pembrolizumab or nivolumab. ADA specific for pembrolizumab and nivolumab did not interfere in the cemiplimab ADA assay. However, pembrolizumab and nivolumab generated a false-positive response in a target-capture NAb assay. Our results demonstrate that similar exogenous pre-existing anti-PD-1 mAbs (biotherapeutics) such as pembrolizumab and nivolumab are detected and accurately quantified in the cemiplimab drug concentration assay. However, once steady state is achieved for the new therapy, prior biologics would likely not be detected. Cross-reactivity and interference in immunoassays from previous treatment with class-specific biotherapeutic(s) pose significant bioanalytical challenges, especially in immuno-oncology.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , Antineoplastic Agents, Immunological/immunology , Nivolumab/immunology , Antibodies, Monoclonal, Humanized/blood , Antibodies, Neutralizing/immunology , Antineoplastic Agents, Immunological/blood , Binding, Competitive , Cross Reactions , Humans , Immune Checkpoint Inhibitors/blood , Immune Checkpoint Inhibitors/immunology , Immunoassay/methods , Nivolumab/blood
4.
Phys Chem Chem Phys ; 23(37): 21207-21217, 2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34533552

ABSTRACT

We investigate the interaction between the programmed cell death protein 1 (PD-1) and the programmed cell death ligand 1 (PD-L1), as well as the immuno-oncological drugs pembrolizumab (PEM), and nivolumab (NIV), through quantum chemistry methods based on the Density Functional Theory (DFT) and the molecular fractionation with conjugate caps (MFCC) scheme, in order to map their hot-spot regions. Our results showed that the total interaction energy order of the three complexes is in good agreement with the experimental binding affinity order: PD-1/PEM > PD-1/NIV > PD-1/PD-L1. Besides, a detailed investigation revealed the energetically most relevant residue-residue pairs-interaction for each complex. Our computational results give a better understanding of the interaction mechanism between the protein PD-1 and its ligands (natural and inhibitors), unleashing the immune surveillance to destroy the cancer cells by decreasing their immune evasion. They are also an efficient alternative towards the development of new small-molecules and antibody-based drugs, pointing out to new treatments for cancer therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/immunology , B7-H1 Antigen/metabolism , Nivolumab/immunology , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/chemistry , Density Functional Theory , Humans , Ligands , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/immunology , Protein Binding , Protein Structure, Tertiary , Thermodynamics
5.
J Immunother ; 44(4): 164-166, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33721881

ABSTRACT

Immune checkpoint inhibitors by blocking specific inhibitory pathways induce T-cell-mediated tumor lysis. However, many remain to be elucidated about their effect on human humoral immunity. We evaluated the effect of the PD1 inhibitor nivolumab on humoral immunity by following the levels of antibodies against hepatitis B surface antigen (anti-HBs) in a hemodialysis patient successfully vaccinated against hepatitis B virus 5 years ago and now starting nivolumab for renal cell carcinoma lung metastases. Anti-HBs kinetics after administration of an extra vaccine dose were also evaluated. Nivolumab increased anti-HBs and facilitated a further increase following an additional vaccine dose. The observed time frame indicates that nivolumab boosts humoral immune response by affecting long-lived plasma cells and at least memory B cells. This may protect cancer patients from pathogens encountered in the past or against which vaccination has been performed and provide information for the emerging immune checkpoint inhibitors administration concept against chronic infectious diseases.


Subject(s)
Immune Checkpoint Inhibitors/immunology , Immunity, Humoral/immunology , Kidney Neoplasms/immunology , Kidney Neoplasms/therapy , Programmed Cell Death 1 Receptor/immunology , Hepatitis B/immunology , Hepatitis B Antibodies/immunology , Hepatitis B Surface Antigens/metabolism , Hepatitis B Vaccines/immunology , Humans , Immunotherapy/methods , Male , Memory B Cells/immunology , Middle Aged , Nivolumab/immunology , Renal Dialysis/methods
6.
Cancer Res ; 81(8): 2184-2194, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33408117

ABSTRACT

Both tumor-infiltrating lymphocytes (TIL) and PD-1+ peripheral blood lymphocytes (PBL) are enriched for tumor-reactive clones recognizing known and unknown tumor antigens. However, the relationship between the T-cell receptor-ß (TCRß) repertoires of the TILs and T cells expanded from paired PD-1+ PBLs, and whether T cells expanded from PD-1+ PBLs can be used to treat patients with cancer as TIL substitutes remain unclear. Here, we established a highly efficient protocol to prepare polyclonal T cells from PD-1+ PBLs. A functional T-cell assay and tetramer staining revealed that cells from PD-1+ PBLs were relatively enriched for tumor-reactive T cells. Furthermore, deep TCRß sequencing data revealed that an average of 11.29% (1.32%-29.06%; P = 0.015; n = 8) tumor-resident clonotypes were found in T cells expanded from paired PD-1+ PBLs, and the mean accumulated frequency of TIL clones found in T cells expanded from PD-1+ PBLs was 35.11% (7.23%-78.02%; P = 0.017; n = 8). Moreover, treatment of four patients, who failed multiline therapy and developed acquired resistance to anti-PD-1, with autologous T cells expanded from PD-1+ PBLs combined with anti-PD-1 antibody elicited objective responses from three of them. These results indicate that T cells expanded from PD-1+ PBLs share more clones with paired TILs and could be used to treat patients with cancer as TIL substitutes. SIGNIFICANCE: This study harnesses the tumor reactivity of PD-1+ PBLs, developing a method to expand T cells from these clones as a potential therapeutic strategy and TIL substitute in patients with cancer.See related commentary by Ladle, p. 1940.


Subject(s)
Carcinoma, Renal Cell/therapy , Cell Proliferation , Immunotherapy, Adoptive/methods , Kidney Neoplasms/therapy , Melanoma/therapy , Programmed Cell Death 1 Receptor , Receptors, Antigen, T-Cell, alpha-beta , T-Lymphocytes/transplantation , Adult , Antigens, Neoplasm/immunology , Cell Separation , Clone Cells/cytology , Clone Cells/immunology , Combined Modality Therapy/methods , Drug Resistance, Neoplasm/immunology , Female , High-Throughput Nucleotide Sequencing , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunomagnetic Separation , Lymphocytes/chemistry , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes, Tumor-Infiltrating/chemistry , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Melanoma/genetics , Middle Aged , Nivolumab/immunology , Receptors, Antigen, T-Cell, alpha-beta/genetics , T-Lymphocytes/chemistry , T-Lymphocytes/cytology , T-Lymphocytes/immunology
7.
Chem Commun (Camb) ; 57(3): 367-370, 2021 Jan 14.
Article in English | MEDLINE | ID: mdl-33325465

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.


Subject(s)
Antibodies, Monoclonal/analysis , Nivolumab/immunology , Transistors, Electronic , Antibodies, Monoclonal/immunology , Electrolytes/chemistry , Humans
8.
Proteins ; 89(2): 141-148, 2021 02.
Article in English | MEDLINE | ID: mdl-32862461

ABSTRACT

Activation of T cells triggers the expression of regulatory molecules like the programmed cell death 1 (PD1) protein. The association of PD1 with the natural ligands PDL1 and PDL2 induces an inhibitory signal that prevents T cells from proliferating and exerting effector functions. However, little is known about how the binding of the ligands induce the PD1 inhibitory signal over T cells effector functions. Here, we explore the dynamics of PD1 free, and in complex with different PDL1 variants as well as the therapeutic antibodies nivolumab and pembrolizumab in order to assess the conformational changes in PD1 related to the signaling process. Our simulations suggest a pre-conformational selection mechanism for the binding of the different PDL1 variants, while an induced-fit model fits better for the molecular recognition process of the therapeutic antibodies. A deep analysis of the changes on PD1 movement upon the binding to different ligands revealed that as larger is the difference in the conformation adopted by loop C'D with respect to the complex with PDL1 is higher the ligand ability to reduce the PD1 inhibitory signaling. This behavior suggests that targeting specific conformations of this loop can be useful for designing therapies able to recover T cells effector functions.


Subject(s)
Antibodies, Monoclonal, Humanized/chemistry , B7-H1 Antigen/chemistry , Nivolumab/chemistry , Programmed Cell Death 1 Receptor/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/metabolism , Antineoplastic Agents, Immunological , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Binding Sites , Gene Expression , Humans , Ligands , Molecular Dynamics Simulation , Nivolumab/immunology , Nivolumab/metabolism , Principal Component Analysis , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Signal Transduction
9.
Protein Expr Purif ; 177: 105766, 2021 01.
Article in English | MEDLINE | ID: mdl-32987122

ABSTRACT

Activated T-cells express Programmed cell Death protein 1 (PD-1), a key immune checkpoint receptor. PD-1 functions primarily in peripheral tissues, where T cells may encounter tumor-derived immunosuppressive ligands. Monoclonal antibodies that disrupt the interaction between T-cell derived PD-1 and immunosuppressive ligands, such as PD-L1, have revolutionized approaches to cancer therapy. For instance, Nivolumab is a monoclonal Ab that targets human PD-1 and has played an important role in immune checkpoint therapy. Herein we report the purification and initial characterization of a ~27 kDa single chain variable fragment (scFv) of Nivolumab that targets human PD-1 and blocks binding by PD-L1. The possibility that the anti-PD-1 scFv can serve as both an anti-tumor agent and as an anti-viral agent is discussed. IMPORTANCE: The clinical significance of anti-PD-1 antibodies for treatment of a range of solid tumors is well documented (reviewed in [1-4]). In this report, we describe the results of studies that establish that an anti-PD-1 scFv purified from E. coli binds tightly to human PD-1. Furthermore, we demonstrate that upon binding, the anti-PD-1 scFv disrupts the interaction between PD-1 and PD-L1. Thus, the properties of this scFv, including its small size, stability and affinity for human PD-1, suggest that it has the potential to be a useful reagent in subsequent immunotherapeutic, diagnostic and anti-viral applications.


Subject(s)
B7-H1 Antigen/chemistry , Nivolumab/chemistry , Programmed Cell Death 1 Receptor/chemistry , Single-Chain Antibodies/chemistry , Amino Acid Sequence , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Binding Sites , Cloning, Molecular , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Models, Molecular , Nivolumab/genetics , Nivolumab/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , T-Lymphocytes/chemistry , T-Lymphocytes/immunology
10.
BMC Bioinformatics ; 21(Suppl 17): 557, 2020 Dec 14.
Article in English | MEDLINE | ID: mdl-33308148

ABSTRACT

BACKGROUND: The immune checkpoint receptor programmed cell death protein I (PD-1) has been identified as a key target in immunotherapy. PD-1 reduces the risk of autoimmunity by inducing apoptosis in antigen-specific T cells upon interaction with programmed cell death protein ligand I (PD-L1). Various cancer types overexpress PD-L1 to evade the immune system by inducing apoptosis in tumor-specific CD8+ T cells. The clinically used blocking antibody nivolumab binds to PD-1 and inhibits the immunosuppressive interaction with PD-L1. Even though PD-1 is already used as a drug target, the exact mechanism of the receptor is still a matter of debate. For instance, it is hypothesized that the signal transduction is based on an active conformation of PD-1. RESULTS: Here we present the results of the first molecular dynamics simulations of PD-1 with a complete extracellular domain with a focus on the role of the BC-loop of PD-1 upon binding PD-L1 or nivolumab. We could demonstrate that the BC-loop can form three conformations. Nivolumab binds to the BC-loop according to the conformational selection model whereas PD-L1 induces allosterically a conformational change of the BC-loop. CONCLUSION: Due to the structural differences of the BC-loop, a signal transduction based on active conformation cannot be ruled out. These findings will have an impact on drug design and will help to refine immunotherapy blocking antibodies.


Subject(s)
B7-H1 Antigen/chemistry , Molecular Dynamics Simulation , Nivolumab/immunology , Programmed Cell Death 1 Receptor/chemistry , Antibodies, Monoclonal/immunology , B7-H1 Antigen/metabolism , Cluster Analysis , Humans , Hydrogen Bonding , Ligands , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Protein Domains
11.
BMJ Case Rep ; 13(9)2020 Sep 09.
Article in English | MEDLINE | ID: mdl-32912886

ABSTRACT

Immune-related myositis is one of the rare immune-related adverse events whose underlying precise mechanisms are not fully understood. Here, we describe a case of immune-related myositis that developed after four cycles of combination therapy with nivolumab plus ipilimumab for the treatment of metastatic renal cell carcinoma. Negative results of autoimmune antibodies, including anti-acetylcholine receptor and anti-muscle-specific kinase antibodies suggested a T-cell-mediated mechanism. After recovery with steroid therapy, the patient resumed nivolumab monotherapy and survived without any evidence of disease progression or refractory of myositis. Differential diagnosis between T-cell-mediated and B-cell-mediated immune-related myositis and its impact on optimal management are discussed.


Subject(s)
Bone Neoplasms , Carcinoma, Renal Cell , Ipilimumab , Lung Neoplasms , Myositis , Nephrectomy/methods , Nivolumab , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/immunology , Bone Neoplasms/diagnosis , Bone Neoplasms/secondary , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/physiopathology , Carcinoma, Renal Cell/therapy , Diagnosis, Differential , Humans , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/immunology , Immunologic Tests/methods , Ipilimumab/adverse effects , Ipilimumab/immunology , Lung Neoplasms/diagnosis , Lung Neoplasms/secondary , Male , Middle Aged , Myositis/etiology , Myositis/immunology , Myositis/therapy , Neoplasm Staging , Nivolumab/administration & dosage , Nivolumab/adverse effects , Nivolumab/immunology , Patient Care Management/methods , Tomography, X-Ray Computed/methods
12.
Immunotherapy ; 12(14): 1041-1046, 2020 10.
Article in English | MEDLINE | ID: mdl-32808556

ABSTRACT

Aim: We describe a case of an advanced disease non-small-cell lung cancer patient with low PD-L1 expression, but high tumor mutation burden (35 muts/Mb) who developed immune-related hypothyroidism and achieved subsequent partial response, while on clinical trial (NCT03382912) with nivolumab and PEGylated IL-10 (Pegilodecakin, ARMO BioSciences/Eli Lilly and Company, IN, USA). Results/conclusion: Results suggest positive antitumor activity to combination IL-10/nivolumab despite low PD-L1 expression but in likely relationship to high tumor mutation burden and in association with immune-mediated thyroid dysfunction in this case.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Hypothyroidism/complications , Immunity/immunology , Interleukin-10/therapeutic use , Lung Neoplasms/drug therapy , Nivolumab/therapeutic use , Polyethylene Glycols/therapeutic use , Aged , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/complications , Lung Neoplasms/immunology , Male , Nivolumab/immunology , Treatment Outcome
13.
Immunotherapy ; 12(13): 957-964, 2020 09.
Article in English | MEDLINE | ID: mdl-32686549

ABSTRACT

Background: Immune checkpoint inhibitors targeting PD-1 and PD-L1 have noticeably improved the treatment landscape of advanced non-small-cell lung cancer, including lung squamous cell carcinoma (SCC). Although patients with immune checkpoint therapy can achieve long-term survival, acquired resistance has been recognized more frequently, while the underlying mechanisms are currently poorly understood. Materials and methods: Here, we report a patient with metastatic lung SCC treated with nivolumab as a first-line treatment for 28 months. Conclusion: The analysis of specimens prenivolumab and postnivolumab treatment suggests that genetic alterations in SOX2 and CDKN2A/CDKN2B and changes in the tumor microenvironment could be reasons for the acquired resistance to nivolumab observed in the lung SCC patient.


Subject(s)
Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/pathology , Drug Resistance/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lymphatic Metastasis/diagnostic imaging , Nivolumab/therapeutic use , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Squamous Cell/immunology , Humans , Lung Neoplasms/immunology , Lymph Nodes/diagnostic imaging , Lymph Nodes/immunology , Lymphatic Metastasis/immunology , Male , Middle Aged , Nivolumab/immunology , Positron Emission Tomography Computed Tomography/methods
14.
J Oncol Pharm Pract ; 26(5): 1289-1294, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32423324

ABSTRACT

Outbreak of the new type coronavirus infection, known as coronavirus infection 2019 (COVID-19), has begun in December 2019, in Wuhan, China. As of today, 3 April 2020, 972,640 people affected and 50,325 people died from Severe Acute Respiratory Syndrome-Coronavirus 2. There is not any standard treatment for coronavirus infection 2019; however, there are promising data for hydroxychloroquine and some anti-retroviral drugs. Programmed death-1 (PD-1)/programmed death ligand-1 (PDL-1) pathway is an important target for the cancer immunotherapy. However, there is a robust pre-clinical and clinical data regarding inhibitor effect of this pathway on the acute or chronic viral infections. Thus, blockade of this pathway may lead to an anti-viral effect and decrease viral load. Here, we report the clinical course of coronavirus infection 2019 infection of a patient in whom older aged, having multiple co-morbidities, and taking nivolumab for metastatic malignant melanoma. In contrast to her older age, comorbidities, and cancer diagnosis, she was in a good condition, and there was also no pneumonia finding. We think that this good clinical course of coronavirus infection 2019 infection may be related to blockade of PD-1/PDL-1 pathway with nivolumab. It is impossible to say that blockade of PD-1/PDL-1pathway is a treatment option for COVID-19; however, we want to share our experience.


Subject(s)
Azithromycin/administration & dosage , Betacoronavirus , Coronavirus Infections , Lung Neoplasms , Melanoma , Nivolumab/administration & dosage , Oseltamivir/administration & dosage , Pandemics , Pneumonia, Viral , Aged , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Agents, Immunological/immunology , Antiviral Agents/administration & dosage , Betacoronavirus/drug effects , Betacoronavirus/physiology , COVID-19 , Coronavirus Infections/diagnosis , Coronavirus Infections/epidemiology , Coronavirus Infections/physiopathology , Coronavirus Infections/virology , Female , Humans , Hydroxychloroquine/administration & dosage , Lung Neoplasms/drug therapy , Lung Neoplasms/secondary , Melanoma/drug therapy , Melanoma/epidemiology , Melanoma/pathology , Multiple Chronic Conditions/epidemiology , Nivolumab/immunology , Pneumonia, Viral/diagnosis , Pneumonia, Viral/epidemiology , Pneumonia, Viral/physiopathology , Pneumonia, Viral/virology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , SARS-CoV-2 , Tomography, X-Ray Computed/methods , Treatment Outcome
15.
Biochem Biophys Res Commun ; 527(1): 213-217, 2020 06 18.
Article in English | MEDLINE | ID: mdl-32446369

ABSTRACT

Monoclonal antibodies, including immune-checkpoint inhibitors, are becoming popular in treatments of many cancers and connective tissue diseases. However, little is known about how long the antibodies combine with antigens on targeted cells or how this duration of binding associates with therapeutic efficacy or potential adverse events. Here, we show the principle and the results of a feasible method for measuring the antibodies' occupancy on the targeted cells using two different detecting antibodies in conjunction with different fluorochromes. Nivolumab occupancy was measured using two detecting antibodies, MIH4 and EH12.2, which are commercially available in vitro (programmed cell death-1 [PD-1] expressing the cell line MIT9 and human T cells) and in T cells from patients treated with nivolumab. Our method has potential for use as a simple and feasible monitoring system in the clinical setting.


Subject(s)
Immune Checkpoint Inhibitors/immunology , Nivolumab/immunology , T-Lymphocytes/immunology , Cell Line , Humans , Immune Checkpoint Inhibitors/analysis , Immune Checkpoint Inhibitors/pharmacology , Nivolumab/analysis , Nivolumab/pharmacology , T-Lymphocytes/drug effects
16.
Anal Chem ; 92(13): 9086-9094, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32441507

ABSTRACT

Programmed cell death-1 (PD-1), an antigen co-receptor on cell surfaces, is one of the conspicuous immune checkpoints. Nivolumab, a monoclonal antibody therapeutic approved by the FDA, binds to PD-1 and efficiently blocks its pathways. In this study, an integrated approach was developed to map the epitope/paratope of PD-1/nivolumab. The approach includes hydrogen-deuterium exchange mass spectrometry (HDX-MS) followed by electron-transfer dissociation (ETD), chemical cross-linking, and molecular docking. HDX-ETD offers some binding-site characterization with amino acid resolution. Chemical cross-linking provides complementary information on one additional epitope (i.e., the BC-loop) and a potential paratope at the N-terminus of the heavy chain. Furthermore, cross-linking identifies another loop region (i.e., the C'D-loop) that undergoes a remote conformational change. The distance restraints derived from the cross-links enable building high-confidence models of PD-1/nivolumab, evaluated with respect to a resolved crystal structure. This integrated strategy is an opportunity to characterize comprehensively other antigen-antibody interactions, to enable the understanding of binding mechanisms, and to design future antibody therapeutics.


Subject(s)
Deuterium Exchange Measurement , Epitope Mapping/methods , Epitopes/analysis , Nivolumab/immunology , Programmed Cell Death 1 Receptor/immunology , Amino Acid Sequence , Antigen-Antibody Complex/chemistry , Binding Sites , Chromatography, High Pressure Liquid , Epitopes/chemistry , Epitopes/immunology , Humans , Molecular Docking Simulation , Nivolumab/metabolism , Programmed Cell Death 1 Receptor/chemistry , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Protein Structure, Secondary , Tandem Mass Spectrometry
17.
Expert Opin Biol Ther ; 20(7): 687-693, 2020 07.
Article in English | MEDLINE | ID: mdl-32249635

ABSTRACT

INTRODUCTION: Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths worldwide and up to recently, sorafenib was the only approved systemic treatment options. In recent years, many other treatment options have received approval for advanced HCC, including nivolumab, a Programmed-Death-1 (PD-1) inhibitor. AREAS COVERED: We review the current treatment landscape of HCC, an overview of the characteristics of nivolumab and current ongoing and completed trials of nivolumab. EXPERT OPINION: Nivolumab is generally well tolerated and has modest single-agent activity as a monotherapy in advanced HCC. Further studies are required to better explore its role as part of combination approaches, in adjuvant/neoadjuvant setting, as well as to understand the impact of sequencing of its use with other systemic therapy options.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Nivolumab/therapeutic use , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/metabolism , Clinical Trials as Topic , Drug Therapy, Combination , Humans , Ipilimumab/therapeutic use , Nivolumab/immunology , Nivolumab/metabolism , Programmed Cell Death 1 Receptor/immunology , Treatment Outcome
18.
J Oncol Pharm Pract ; 26(2): 459-461, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30909794

ABSTRACT

Immune checkpoint inhibitors have ushered in a new era in cancer management. Nivolumab is a human immunoglobulin G4 (IgG4) monoclonal antibody that selectively inhibits programmed cell death-1 (PD-1) activity by binding to the PD-1 receptor. This inhibits suppression of the T-cell activity, which can in turn cause increased killing of cancer cells. This alteration in the activity of the T cells can cause them to lose their ability to identify host cells and leads to immune-related adverse effects (irAE). Nivolumab-induced hepatotoxicity is rare and accounts for 3-6% of all irAE. We present a case of nivolumab-induced hepatitis. A woman who was treated for recurrent renal cell carcinoma presented with hepatitis. Workup for other causes was negative and the hepatitis was attributed to the administration of nivolumab. She was started on oral steroids followed which she initially improved. However, she later presented with massive upper gastrointestinal bleeding secondary to gastroduodenal ulcers and subsequently developed acute tubular necrosis and passed from the complications. Immune checkpoint inhibitors have proven to be a promising approach in the management of a wide array of neoplasms by immunomodulation. As these agents are becoming standard of therapy in the management of cancers, a heightened vigilance in the diagnosis of irAE is warranted. With heightened vigilance, early recognition can lead to decreased mortality and morbidity.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Hepatitis/diagnosis , Hepatitis/etiology , Immunotherapy/adverse effects , Nivolumab/adverse effects , Aged , Antineoplastic Agents, Immunological/immunology , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/drug therapy , Carcinoma, Renal Cell/immunology , Drug-Related Side Effects and Adverse Reactions/diagnosis , Drug-Related Side Effects and Adverse Reactions/drug therapy , Female , Hepatitis/immunology , Humans , Kidney Neoplasms/diagnosis , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/drug therapy , Nivolumab/immunology , Nivolumab/therapeutic use
19.
J Oncol Pharm Pract ; 26(2): 445-458, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31631812

ABSTRACT

Renal cell carcinoma is a common malignancy of the genitourinary system and is the eight most common cancer type in the United States. The overall incidence of renal cell carcinoma appears to be increasing but death rates have been declining. Patients with poor risk, advanced disease have a two-year survival rate of approximately 7%. Prior to the advent of tyrosine kinase inhibitors, anti-vascular endothelial growth factor antibodies, mammalian target of rapamycin inhibitors, and checkpoint inhibitors, IFN-α and high-dose IL-2, were standard of care treatment options but, conversely, their use is now limited to select patients. Immunotherapies have played a significant role in the treatment of various cancers and have continued to expand. It is of utmost importance that practitioners include checkpoint inhibitors as treatment options for renal cell carcinoma as they mark a new era in the treatment of advanced or relapsed setting. Nivolumab, pembrolizumab, avelumab, ipilimumab, and atezolizumab all play a role in management of disease as either monotherapy or in combination with other agents. Ongoing clinical trials are ongoing to further assess the benefits of inducing cellular immunity in the treatment of renal cell carcinoma. In this article, the available data on immune checkpoint inhibitors for the treatment of advanced or relapsed renal cell carcinoma and their place in therapy are reviewed.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Carcinoma, Renal Cell/drug therapy , Immunotherapy/methods , Kidney Neoplasms/drug therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/immunology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Immunological/immunology , Carcinoma, Renal Cell/immunology , Humans , Ipilimumab/immunology , Ipilimumab/therapeutic use , Kidney Neoplasms/immunology , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Nivolumab/immunology , Nivolumab/therapeutic use
20.
Sci Rep ; 9(1): 15205, 2019 10 23.
Article in English | MEDLINE | ID: mdl-31645587

ABSTRACT

Immunotherapy has emerged as a promising and effective treatment for cancer. The frequently used immunotherapy agents are immune checkpoint inhibitors, such as antibodies specific to PD1, PD-L1, or CTLA-4. However, these drugs are highly expensive, and most people in the world cannot access the treatment. The development of recombinant protein production platforms that are cost-effective, scalable, and safe is needed. Plant platforms are attractive because of their low production cost, speed, scalability, lack of human and animal pathogens, and post-translational modifications that enable them to produce effective monoclonal antibodies. In this study, an anti-PD1 IgG4 monoclonal antibody (mAb) was transiently produced in Nicotiana benthamiana leaves. The plant-produced anti-PD1 mAb was compared to the commercial nivolumab produced in CHO cells. Our results showed that both antibodies have similar protein structures, and the N-glycans on the plant-produced antibody lacks plant-specific structures. The PD1 binding affinity of the plant-produced and commercial nivolumab, determined by two different techniques, that is, enzyme-linked immunosorbent assay (ELISA) and surface plasmon resonance (SPR), are also comparable. Plant-produced nivolumab binds to human PD1 protein with high affinity and specificity, blocks the PD-1/PD-L1 interaction, and enhances T cell function, comparable to commercial nivolumab. These results confirmed that plant-produced anti-PD1 antibody has the potential to be effective agent for cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Nicotiana/genetics , Programmed Cell Death 1 Receptor/immunology , Animals , Antibodies, Monoclonal/chemistry , Biotechnology , CHO Cells , Cloning, Molecular , Cricetulus , Gene Expression , Genetic Vectors/genetics , Humans , Nivolumab/chemistry , Nivolumab/genetics , Nivolumab/immunology , Protein Conformation , Nicotiana/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...