151
|
Yao Q, Fischer KP, Tyrrell DL, Gutfreund KS. Molecular cloning, expression and characterization of Pekin duck programmed death-1. Gene 2019; 702:182-193. [PMID: 30910561 DOI: 10.1016/j.gene.2019.03.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/06/2019] [Accepted: 03/18/2019] [Indexed: 11/17/2022]
Abstract
Programmed death-1 (PD-1) has a pivotal role in the attenuation of adaptive immune responses and peripheral tolerance. Here we describe the identification of the Pekin duck programmed death-1 orthologue (duPD-1). The duPD-1 cDNA encodes a 283-amino acid polypeptide that has an amino acid identity of 70%, 32% and 31% with chicken, murine and human PD-1, respectively. The duck PD-1 gene shares five conserved exons with chicken, murine and human PD-1 genes. A cluster of putative regulatory elements within the conserved region B (CR-B) of the basal promotor is conserved. Homology modeling was most compatible with the two β-sheet IgV domain structure of murine PD-1. Contact residues, shown to be critical for binding of the respective human and murine PD-1 ligands are mostly conserved between avian and mammalian species, whereas residues that define the cytoplasmic immunoreceptor tyrosine-based inhibitory motif (ITIM) and immunoreceptor tyrosine-based switch motif (ITSM) are highly conserved across higher vertebrates and frog. Constitutive expression of duPD-1 transcripts was predominantly found in lymphocyte-rich tissues, and mitogen-stimulation of duck peripheral blood mononuclear cells transiently increased duPD-1 mRNA expression. A soluble duPD-1 protein was expressed and shown to engage the identified duck PD-1 ligands. Our observations show considerable evolutionary conservation between mammalian and avian PD-1 orthologues. This work will facilitate further investigation of the role of PD-1 signaling in adaptive immunity in the Pekin duck, a non-mammalian vertebrate and pathogen host with relevance for human and animal health.
Collapse
Affiliation(s)
- Qingxia Yao
- Dept. of Medicine, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karl P Fischer
- Dept. of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - D Lorne Tyrrell
- Dept. of Medicine, University of Alberta, Edmonton, AB, Canada; Dept. of Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Klaus S Gutfreund
- Dept. of Medicine, University of Alberta, Edmonton, AB, Canada; Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
152
|
West SM, Deng XA. Considering B7-CD28 as a family through sequence and structure. Exp Biol Med (Maywood) 2019; 244:1577-1583. [PMID: 31208204 DOI: 10.1177/1535370219855970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
With the emergence of immuno-oncology, new therapeutic agents that modulate immune activation and regulation are being used to treat cancer patients with durable response. It is well known that following T-cell receptor (TCR) activation, many co-receptors can augment or suppress the TCR signal, and therapeutically targeting these co-receptors has proven effective. The B7-CD28 family is comprised of such immune-regulatory receptors, and antibodies against its members programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) have revolutionized cancer treatment. These therapies promote an immune response against tumor cells, which demonstrated better long-term survival and tolerability compared to traditional cancer treatments. In this review we describe the history of the expanding B7-CD28 family, and by comparison of sequence and structure reveal that it is a non-traditional family. The family has grown to include proteins that share low sequence identity, generally grouped by regulation of immune response, which utilize the common immunoglobulin fold. This low level of commonality has provided additional challenges to the drug discovery process as the mechanisms and therapeutic potency between family members can vary greatly. Impact statement Immunotherapy as a field has dramatically expanded in the last decade in the area of oncology with efficacy demonstrated by PD-1, PD-L1, and CTLA-4 blockade. With all three “checkpoint blockade” receptors being in the B7-CD28 family, there has been increased interest in targeting other members in this family due to redundancy in immune regulation, i.e., the combination of therapeutic agents targeting multiple co-inhibitory receptors may yield additional antitumor efficacy. Therefore significant resources are being dedicated to developing additional B7-CD28 treatment options.
Collapse
Affiliation(s)
- Sean M West
- Bristol-Myers Squibb (BMS), Redwood City, CA 94063, USA
| | - Xiaodi A Deng
- Bristol-Myers Squibb (BMS), Redwood City, CA 94063, USA
| |
Collapse
|
153
|
Lin Y, Cui C, Su M, Tian X, Huang Y, Zhao J, Lai L. Skint8, a Novel B7 Family-Related Molecule, Negatively Regulates T Cell Responses. THE JOURNAL OF IMMUNOLOGY 2019; 203:400-407. [PMID: 31189570 DOI: 10.4049/jimmunol.1800639] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 05/17/2019] [Indexed: 01/18/2023]
Abstract
Immune responses are tightly controlled by T cell costimulatory and coinhibitory molecules. In this study, we identify Skint8 as a new member of the T cell coinhibitory group, whose extracellular domains share significant homology with existing B7 family members. Skint8 mRNA is expressed in resting and activated B cells, monocytes, and CD4 T cells. The Skint8 putative receptor is expressed on activated CD4 and CD8 T cells, B cells, monocytes and dendritic cells. Recombinant Skint8-IgG Fc fusion protein inhibits T cell proliferation, activation, and cytokine production in vitro. In vivo administration of Skint8-IgG Fc reduces T cell activation and alleviates experimental autoimmune encephalomyelitis in mice. The findings broaden our understanding of the regulation of immune responses and may have implications for treating immune-related diseases.
Collapse
Affiliation(s)
- Yujun Lin
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269.,Department of Oncology, Fuzhou Pulmonary Hospital, Fujian 350008, China; and
| | - Cheng Cui
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269
| | - Xiaohong Tian
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269
| | - Yuanmao Huang
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269
| | - Jin Zhao
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT 06269; .,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT 06269
| |
Collapse
|
154
|
Abstract
Immunotherapy has significantly advanced the field of oncology in recent decades. Understanding normal immunosurveillance, as well as the ways in which tumor cells have evolved to evade it, has provided the knowledge for development of drugs that allow one's own immune system to target and destroy malignant cells (immunotherapy). Cutaneous malignancies are particularly sensitive to this class of drugs. In a very sensitive anatomic region such as the periocular tissue, where surgical excision may come with significant morbidity, this technology has had a strong impact in the successful treatment of historically challenging tumors.
Collapse
Affiliation(s)
- Larissa A Habib
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Natalie Wolkow
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Suzanne K Freitag
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| | - Michael K Yoon
- a Ophthalmic Plastic Surgery , Massachusetts Eye and Ear Infirmary , Boston , MA , USA.,b Department of Ophthalmology , Harvard Medical School , Boston , MA , USA
| |
Collapse
|
155
|
The role of TNF-α in chordoma progression and inflammatory pathways. Cell Oncol (Dordr) 2019; 42:663-677. [PMID: 31175552 DOI: 10.1007/s13402-019-00454-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
PURPOSE Chordomas are highly therapy-resistant primary bone tumors that exhibit high relapse rates and may induce local destruction. Here, we evaluated the effects of tumor necrosis factor-alpha (TNF-α) on chordoma progression and clinical outcome. METHODS Chordoma cells were treated with TNF-α after which its short- and long-term effects were evaluated. Functional assays, qRT-PCR and microarray-based expression analyses were carried out to assess the effect of TNF-α on chemo-resistance, epithelial to mesenchymal transition (EMT), migration, invasion and cancer stem cell-like properties. Finally, relationships between TNF-α expression and clinicopathological features were assessed in a chordoma patient cohort. RESULTS We found that TNF-α treatment increased the migration and invasion of chordoma cells. Also, NF-κB activation was observed along with increased EMT marker expression. In addition, enhanced tumor sphere formation and soft agar colony formation were observed, concomitantly with increased chemo-resistance and CD338 marker expression. The TNF-α and TNFR1 expression levels were found to be significantly correlated with LIF, PD-L1 and Ki67 expression levels, tumor volume and a short survival time in patients. In addition, a high neutrophil to lymphocyte ratio was found to be associated with recurrence and a decreased overall survival. CONCLUSIONS From our data we conclude that TNF-α may serve as a prognostic marker for chordoma progression and that tumor-promoting inflammation may be a major factor in chordoma tumor progression.
Collapse
|
156
|
Abstract
Cancer immunotherapy (CIT) has transformed cancer treatment. In particular, immunotherapies targeting the programmed death ligand 1 (PD-L1)/programmed death 1 pathway have demonstrated durable clinical benefit in some patients. However, CIT combinations may create a more favorable environment in which to maximize the potential of the immune system to eliminate cancer. Here we describe 3 key mechanisms related to vascular endothelial growth factor (VEGF)-mediated immunosuppression: inhibition of dendritic cell maturation, reduction of T-cell tumor infiltration, and promotion of inhibitory cells in the tumor microenvironment; supporting data are also described. In addition, we discuss immunomodulatory properties observed within tumors following bevacizumab treatment. Combining anti-PD-L1 and anti-VEGF therapies has shown synergy and positive outcomes in phases I to III studies, particularly in settings where high VEGF levels are known to play an important role in tumor growth. We also review data from key studies supporting combination of bevacizumab and CIT, with a focus on PD-L1/programmed death 1 inhibitors.
Collapse
|
157
|
Roviello G, D'Angelo A, Generali D, Pittacolo M, Ganzinelli M, Iezzi G, Manzini ND, Sobhani N. Avelumab in gastric cancer. Immunotherapy 2019; 11:759-768. [PMID: 31060469 DOI: 10.2217/imt-2019-0011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 04/11/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is the fifth most common malignancy and the third cause of cancer-related deaths worldwide. Currently, surgery and chemotherapy remain the main therapeutic options and the prognosis of the disease is still poor in the metastatic setting. Avelumab is a human IgG1 antibody directed against PD-L1 approved for Merkel cell carcinoma and urothelial carcinoma that could be useful also for the treatment of GC. This review describes the chemical structure, the pharmacologic properties and the current knowledge of the efficacy of avelumab in the treatment of GC from the data available on the first and later phase clinical trials. The ongoing studies testing this drug either alone or in combination with other drugs are also described.
Collapse
MESH Headings
- Antibodies, Monoclonal/pharmacokinetics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/metabolism
- Carcinoma, Merkel Cell/drug therapy
- Carcinoma, Merkel Cell/metabolism
- Carcinoma, Merkel Cell/pathology
- Carcinoma, Transitional Cell/drug therapy
- Carcinoma, Transitional Cell/metabolism
- Carcinoma, Transitional Cell/pathology
- Clinical Trials as Topic
- Humans
- Immunoglobulin G/therapeutic use
- Skin Neoplasms/drug therapy
- Skin Neoplasms/metabolism
- Skin Neoplasms/pathology
- Stomach Neoplasms/drug therapy
- Stomach Neoplasms/metabolism
- Stomach Neoplasms/pathology
- Treatment Outcome
Collapse
Affiliation(s)
- Giandomenico Roviello
- Department of Health Sciences, University of Florence, Viale Pieraccini, 6, 50139 Florence, Italy
| | - Alberto D'Angelo
- Department of Biology & Biochemistry, University of Bath, Bath BA2 7AY, United Kingdom
| | - Daniele Generali
- Breast Cancer Unit, ASST Cremona, Viale Concordia 1, 26100 Cremona, Italy
| | - Matteo Pittacolo
- Department of Orthopedics & Orthopedic Oncology, University of Padova, Italy
| | - Monica Ganzinelli
- Thoracic Unit, Oncology Department, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Nicolò de Manzini
- General Surgery Unit, Department of Medical, Surgical and Health Sciences Cattinara University Hospital, Azienda Sanitaria Universitaria Integrata di Trieste, Trieste, Italy
| | - Navid Sobhani
- Breast Cancer Unit, ASST Cremona, Viale Concordia 1, 26100 Cremona, Italy
| |
Collapse
|
158
|
Graves M, CelliMarchett G, van Zyl B, Tang D, Vilain RE, van der Westhuizen A, Bowden NA. Monitoring Patient Response to Pembrolizumab With Peripheral Blood Exhaustion Marker Profiles. Front Med (Lausanne) 2019; 6:113. [PMID: 31192212 PMCID: PMC6540682 DOI: 10.3389/fmed.2019.00113] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 05/07/2019] [Indexed: 11/13/2022] Open
Abstract
Exhausted T cells are effector T cells that are silenced due to continuous T cell receptor (TCR) stimulation from persistent antigens. Characteristics of exhaustion include the increased expression of multiple inhibitory receptors such as programme death-1[PD-1], lymphocyte activation gene 3 [LAG-3], T cell Ig and mucin domain [TIM-3], the loss of effector cytokine secretion and altered transcriptional profile. The PD-1/PD-L1 interaction induces functional exhaustion of tumor-reactive cytotoxic T cells and interferes with anti-tumor T cell immunity. T cell exhaustion has been observed in metastatic melanoma patients where the exhaustion of tumor specific T cells suggests that tumor clearance has been impeded and contributed to tumor immune escape. Checkpoint immunotherapies are antibodies designed to block the interaction between the inhibitory receptors expressed on T cells and their respective ligands. Therapies such as anti-PD-1 (Pembrolizumab and Nivolumab) block these inhibitory receptors and are associated with a significant improvement in overall survival and progression free survival. However, only 20-40% of metastatic melanoma patients experience long-term benefit. In a cohort of 16 metastatic melanoma patients receiving pembrolizumab, blood was serially collected before each infusion (mean 8.3; range 1-12 cycles). The presence of inhibitory markers LAG-3, TIM-3, and PD-1 on the surface of T cells was examined and assessed in relation to patient response to identify if inhibitory markers can be used to differentiate responders from non-responders for Pembrolizumab. We confirmed that across a range of cycles (range 1-26) of pembrolizumab, PD-1 expression was significantly higher on CD4+ T cells from non-responders compared to responders and TIM-3 expressed on the surface of CD8+ T cells was significantly higher in non-responders compared to responders. This longitudinal data confirms previous studies that assessed single timepoints. This study provides preliminary evidence that PD-1 and TIM-3 may be predictive of non-responders when assessed over multiple treatment cycles.
Collapse
Affiliation(s)
- Moira Graves
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | | | - Belinda van Zyl
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| | - Denise Tang
- School of Biomedical Science and Pharmacy, University of Newcastle, Newcastle, NSW, Australia
| | - Ricardo E Vilain
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Anatomical Pathology, Pathology NSW, Newcastle, NSW, Australia
| | - Andre van der Westhuizen
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia.,Department of Medical Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Nikola A Bowden
- School of Medicine and Public Health, Hunter Medical Research Institute, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
159
|
Fritz JM, Lenardo MJ. Development of immune checkpoint therapy for cancer. J Exp Med 2019; 216:1244-1254. [PMID: 31068379 PMCID: PMC6547853 DOI: 10.1084/jem.20182395] [Citation(s) in RCA: 131] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 02/22/2019] [Accepted: 04/17/2019] [Indexed: 12/14/2022] Open
Abstract
Fritz and Lenardo discuss the basic science and clinical discoveries of immune checkpoint blockade, which boosts antitumor immunity and increases survival of patients with cancer. Since the early 20th century, immunologists have investigated mechanisms that protect vertebrates from damaging immune responses against self-antigens by mature lymphocytes, i.e., peripheral tolerance. These mechanisms have been increasingly delineated at the molecular level, ultimately culminating in new therapeutics that have revolutionized clinical oncology. Here, we describe basic science and clinical discoveries that converge mainly on two molecules, CTLA-4 and PD-1, that were recognized with the 2018 Nobel Prize in Physiology or Medicine awarded to James Allison and Tasuku Honjo. We discuss their investigations and those of many others in the field that contravene tolerance through checkpoint inhibition to boost immune killing of malignant cells. We also discuss the mechanisms underlying each therapy, the efficacy achieved, and the complications of therapy. Finally, we hint at research questions for the future that could widen the success of cancer immunotherapy.
Collapse
Affiliation(s)
- Jill M Fritz
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, and Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Michael J Lenardo
- Molecular Development of the Immune System Section, Laboratory of Immune System Biology, and Clinical Genomics Program, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|
160
|
Zhou H, Fu X, Li Q, Niu T. Safety and Efficacy of Anti-PD-1 Monoclonal Antibodies in Patients With Relapsed or Refractory Lymphoma: A Meta-Analysis of Prospective Clinic Trails. Front Pharmacol 2019; 10:387. [PMID: 31118893 PMCID: PMC6504777 DOI: 10.3389/fphar.2019.00387] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Accepted: 03/28/2019] [Indexed: 02/05/2023] Open
Abstract
Background: Immune checkpoint inhibition therapy with monoclonal antibody against programmed cell death protein 1 (PD-1), including nivolumab and pembrolizumab, has demonstrated powerful clinical efficacy in the treatment of advanced cancers. However, there is no evidence-based systematic review on the safety and efficacy of anti-PD-1 antibody in treating lymphoma. Methods: To evaluate the safety and efficacy of nivolumab/pembrolizumab, we analyzed clinical trials from PUBMED, EMBASE, and The Cochrane Library. For safety analysis, the incidence and exhibition of any grade and grade ≥3 adverse events (AEs) were evaluated. Overall response rate (ORR), 6-month progression-free survival (PFS) and 6-month overall survival (OS) were calculated for efficacy analysis. Results: Overall ten studies and 718 patients (114 non-Hodgkin lymphomas, 604 Hodgkin lymphomas) were enrolled, including 4 phase I studies and 6 phase II studies. The pooled incidences of any grade and grade ≥3 adverse events (AEs) were 74 and 24%, respectively. Drug-related deaths occurred in two patients. The most common any grade AEs were fatigue (14.91%), rash (14.8%), hypothyroidism (13.77%), platelet count decreased (13.54%), pyrexia (13%). The most common grade ≥3 AEs were neutropenia (4.79%), pneumonitis (3.58%), rash (3.38%), and leukopenia (3.31%). Fatigue (p = 0.0072) and rash (p = 0.0078) in any grade AEs were less observed in patients treated with pembrolizumab than nivolumab. The pooled ORR, PFS rate and OS rate were 58, 73, and 96%, respectively. The ORR in patients with Hodgkin lymphomas (HL) was higher than patients with non-Hodgkin lymphomas (NHL) (69.08 vs. 30.77%, p < 0.0001). However, there was no significant difference of efficacy between nivolumab and pembrolizumab. Conclusions: Nivolumab and pembrolizumab have promising outcomes with tolerable AEs and drug-related deaths in patients with relapsed or refractory lymphoma. Pembrolizumab caused less any grade AEs like fatigue and rash than nivolumab. Patients with HL got better response than NHL.
Collapse
Affiliation(s)
- Hui Zhou
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoyan Fu
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Qian Li
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, China
| | - Ting Niu
- Department of Hematology and Research Laboratory of Hematology, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
161
|
Tian X, Lin Y, Cui C, Su M, Lai L. BTNL2-Ig Protein Attenuates Type 1 Diabetes in Non-Obese Diabetic (NOD) Mice. Adv Healthc Mater 2019; 8:e1800987. [PMID: 30884196 DOI: 10.1002/adhm.201800987] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Indexed: 12/23/2022]
Abstract
Type 1 diabetes (T1D) is a T cell-mediated autoimmune disease in which insulin-producing β-cells are destroyed. Although butyrophilin-like 2 (BTNL2) has been shown to be a negative T cell regulator in vitro, its ability to inhibit T cell responses in vivo has not been determined. In this study, the effect of a recombinant BTNL2-IgG2a Fc (rBTNL2-Ig) fusion protein on T1D development in vivo is determined. It is shown here that in vivo administration of rBTNL2-Ig ameliorates T1D in non-obese diabetic (NOD) mice. This is associated with the ability of rBTNL2-Ig to inhibit the proliferation, activation, and inflammatory cytokine production from autoreactive T cells in vivo. In addition, rBTNL2-Ig treatment increases the generation of regulatory T cells. The results suggest that targeting the BTNL2 pathway has the potential to be used in the prevention and treatment of patients with T1D.
Collapse
Affiliation(s)
- Xiaohong Tian
- Department of Allied Health SciencesUniversity of Connecticut Storrs CT 06269 USA
- Department of Tissue engineeringSchool of Fundamental ScienceChina Medical University Shenyang Liaoning 110122 China
| | - Yujun Lin
- Department of Allied Health SciencesUniversity of Connecticut Storrs CT 06269 USA
| | - Cheng Cui
- Department of Allied Health SciencesUniversity of Connecticut Storrs CT 06269 USA
| | - Min Su
- Department of Allied Health SciencesUniversity of Connecticut Storrs CT 06269 USA
| | - Laijun Lai
- Department of Allied Health SciencesUniversity of Connecticut Storrs CT 06269 USA
- University of Connecticut Stem Cell InstituteUniversity of Connecticut Storrs CT 06269 USA
| |
Collapse
|
162
|
PD-L1 Expression is Associated With Poor Prognosis in Renal Cell Carcinoma. Appl Immunohistochem Mol Morphol 2019; 28:213-220. [DOI: 10.1097/pai.0000000000000766] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
163
|
Bastiaannet E, Battisti N, Loh KP, de Glas N, Soto-Perez-de-Celis E, Baldini C, Kapiteijn E, Lichtman S. Immunotherapy and targeted therapies in older patients with advanced melanoma; Young International Society of Geriatric Oncology review paper. J Geriatr Oncol 2019; 10:389-397. [PMID: 30025821 PMCID: PMC8074511 DOI: 10.1016/j.jgo.2018.06.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 06/19/2018] [Accepted: 06/19/2018] [Indexed: 12/17/2022]
Abstract
Malignant melanoma is an aggressive cancer associated with a poor prognosis in patients with metastatic disease. As in many other cancers, the incidence of melanoma rises with age; and combined with the longer life expectancy, this led to an increasing prevalence of melanoma in the older population. Recently, immune checkpoint inhibitors significantly improved the treatment of melanoma given their efficacy and tolerability profile. Two major classes of agents include the anti-cytotoxic T lymphocyte-associated protein 4 (CTLA-4) inhibitors, such as ipilimumab, and the anti-programmed death-ligand 1 (PD-1) inhibitors, such as nivolumab and pembrolizumab. Treatment of metastatic disease with immune checkpoint inhibitors demonstrated improved efficacy and better safety profiles compared to cytotoxic drugs and appears to be an attractive treatment option. Nevertheless, there is a need for tools designed to better predict which older patients will benefit from its use and who will experience toxicities related to the treatment. Current data do not show a major increase in toxicity rates in older patients. However, patients above 75 are often under-represented and those who are included are not representative of the general population of older patients, thereby also stressing the need for real-life data. Ongoing research is aiming at maximizing the potential treatment efficacy and developing novel immune-targeting modalities. Future studies should include older patients and assess geriatric domains in these older patients to better guide decision-making. This review discusses published clinical trials and where known, the efficacy and toxicity in older patients. Moreover, the clinical implications and future perspectives are discussed, with current recommendations for older patients, management of toxicities, and a proposal for an initial approach to the treatment of older patients with metastatic melanoma.
Collapse
Affiliation(s)
- Esther Bastiaannet
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands; Department of Surgery, Leiden University Medical Center, Leiden, The Netherlands.
| | - Nicolò Battisti
- Department of Medicine, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - Kah Poh Loh
- Division of Hematology/Oncology, James P. Wilmot Cancer Institute, University of Rochester Medical Center, USA
| | - Nienke de Glas
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Enrique Soto-Perez-de-Celis
- Cancer Care in the Elderly Clinic, Department of Geriatrics, Instituto Nacional de Ciencias Medicas y Nutricion Salvador Zubiran, Mexico City, Mexico
| | - Capucine Baldini
- Drug Development Department (DITEP), Gustave Roussy Cancer Campus, Villejuif Cedex F-94805, France
| | - Ellen Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, The Netherlands
| | - Stuart Lichtman
- Department of Medical Oncology, Memorial Sloan Kettering Cancer Center, NY, USA
| |
Collapse
|
164
|
Wartewig T, Ruland J. PD-1 Tumor Suppressor Signaling in T Cell Lymphomas. Trends Immunol 2019; 40:403-414. [PMID: 30979616 DOI: 10.1016/j.it.2019.03.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/11/2019] [Accepted: 03/11/2019] [Indexed: 12/15/2022]
Abstract
The inhibitory receptor PD-1 is critical to balancing antigen-induced T cell activation; its inhibition is currently being explored to enhance antitumor T cell immunity with certain successful outcomes. However, PD-1 has also emerged as a central tumor suppressor in T cell lymphomas, where the tumor cell originates from a T cell itself. These aggressive cancers are frequently characterized by oncogenic mutations in T cell receptor (TCR) signaling pathways. PD-1 activity within malignant T cells can negatively regulate the PI3K/AKT and PKCθ/NF-κB tumor survival pathways and PD-1 is frequently inactivated in this human malignancy. This review summarizes current insights into oncogenic T cell signaling, discusses tumor-suppressive functions and mechanisms of PD-1 in T cell lymphomagenesis, and addresses potential unwanted effects caused by PD-1 checkpoint inhibition.
Collapse
Affiliation(s)
- Tim Wartewig
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), Munich, Germany
| | - Jürgen Ruland
- Institute of Clinical Chemistry and Pathobiochemistry, School of Medicine, Technical University of Munich, Munich, Germany; Center for Translational Cancer Research (TranslaTUM), Munich, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany; German Center for Infection Research (DZIF), partner site, Munich, Germany.
| |
Collapse
|
165
|
Furusawa E, Ohno T, Nagai S, Noda T, Komiyama T, Kobayashi K, Hamamoto H, Miyashin M, Yokozeki H, Azuma M. Silencing of PD-L2/B7-DC by Topical Application of Small Interfering RNA Inhibits Elicitation of Contact Hypersensitivity. J Invest Dermatol 2019; 139:2164-2173.e1. [PMID: 30978356 DOI: 10.1016/j.jid.2019.02.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Revised: 02/26/2019] [Accepted: 02/27/2019] [Indexed: 11/25/2022]
Abstract
PD-L2 is a ligand for the immune checkpoint receptor PD-1; however, its regulatory function is unclear. We previously reported that silencing of CD86 in cutaneous dendritic cells by topical application of small interfering RNA (siRNA) inhibits the elicitation of contact hypersensitivity (CHS). Here, we investigated the effects of topical application of PD-L2 siRNA on allergic skin disease. PD-L2 was induced in dendritic cells concurrently with the elevation of major histocompatibility complex class II and CD86 expression. Topical application of PD-L2 siRNA inhibited the elicitation of CHS by suppressing early proinflammatory cytokine expression and migration of hapten-carrying dendritic cells into lymph nodes. Local injection of neutralizing anti-PD-L2 mAb inhibited CHS to the same extent. PD-L2 siRNA treatment inhibited CHS in PD-1/PD-L1 double knockout mice and in the sensitized T-cell-transferred skin. These results suggest that the effects of PD-L2 silencing are independent of PD-1 but dependent on local memory T cells. Most of the inhibitory effects of PD-L2 and CD86 silencing on CHS were comparable, but PD-L2 siRNA treatment did not inhibit atopic disease-like manifestations and T helper type 2 responses in NC/Nga mice. Our results suggest that PD-L2 in cutaneous dendritic cells acts as a costimulator rather than a regulator. Local PD-L2 silencing by topical application of siRNA represents a therapeutic approach for contact allergy.
Collapse
Affiliation(s)
- Emi Furusawa
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan; Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tatsukuni Ohno
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigenori Nagai
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Taisei Noda
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takuya Komiyama
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | - Michiyo Miyashin
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroo Yokozeki
- Department of Dermatology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Miyuki Azuma
- Department of Molecular Immunology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| |
Collapse
|
166
|
Xu-Monette ZY, Xiao M, Au Q, Padmanabhan R, Xu B, Hoe N, Rodríguez-Perales S, Torres-Ruiz R, Manyam GC, Visco C, Miao Y, Tan X, Zhang H, Tzankov A, Wang J, Dybkær K, Tam W, You H, Bhagat G, Hsi ED, Ponzoni M, Ferreri AJM, Møller MB, Piris MA, van Krieken JH, Winter JN, Westin JR, Pham LV, Medeiros LJ, Rassidakis GZ, Li Y, Freeman GJ, Young KH. Immune Profiling and Quantitative Analysis Decipher the Clinical Role of Immune-Checkpoint Expression in the Tumor Immune Microenvironment of DLBCL. Cancer Immunol Res 2019; 7:644-657. [PMID: 30745366 DOI: 10.1158/2326-6066.cir-18-0439] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 09/23/2018] [Accepted: 02/05/2019] [Indexed: 11/16/2022]
Abstract
PD-1/L1 and CTLA-4 blockade immunotherapies have been approved for 13 types of cancers and are being studied in diffuse large B-cell lymphoma (DLBCL), the most common aggressive B-cell lymphoma. However, whether both PD-1 and CTLA-4 checkpoints are active and clinically significant in DLBCL is unknown. Whether PD-1 ligands expressed by tumor cells or by the microenvironment of DLBCL are critical for the PD-1 immune checkpoint is unclear. We performed immunophenotypic profiling for 405 patients with de novo DLBCL using a MultiOmyx immunofluorescence platform and simultaneously quantitated expression/coexpression of 13 immune markers to identify prognostic determinants. In both training and validation cohorts, results demonstrated a central role of the tumor immune microenvironment, and when its functionality was impaired by deficiency in tumor-infiltrating T cells and/or natural killer cells, high PD-1 expression (but not CTLA-4) on CD8+ T cells, or PD-L1 expression on T cells and macrophages, patients had significantly poorer survival after rituximab-CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) immunochemotherapy. In contrast, tumor-cell PD-L2 expression was associated with superior survival, as well as PD-L1+CD20+ cells proximal (indicates interaction) to PD-1 + CD8+ T cells in patients with low PD-1 + percentage of CD8+ T cells. Gene-expression profiling results suggested the reversibility of T-cell exhaustion in PD-1+/PD-L1+ patients with unfavorable prognosis and implication of LILRA/B, IDO1, CHI3L1, and SOD2 upregulation in the microenvironment dysfunction with PD-L1 expression. This study comprehensively characterized the DLBCL immune landscape, deciphered the differential roles of various checkpoint components in rituximab-CHOP resistance in DLBCL patients, and suggests targets for PD-1/PD-L1 blockade and combination immunotherapies.
Collapse
Affiliation(s)
- Ziju Y Xu-Monette
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Min Xiao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Qingyan Au
- NeoGenomics Laboratories, Inc., Aliso Viejo, California.
| | | | - Bing Xu
- Department of Hematology, The First Affiliated Hospital of Xiamen University, Fujian, China.
| | - Nicholas Hoe
- NeoGenomics Laboratories, Inc., Aliso Viejo, California
| | - Sandra Rodríguez-Perales
- Molecular Cytogenetics Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
| | - Raul Torres-Ruiz
- Molecular Cytogenetics Unit, Human Cancer Genetics Programme, Spanish National Cancer Research Centre (CNIO), Madrid, Spain
- Josep Carreras Leukemia Research Institute, Department of Biomedicine, School of Medicine, University of Barcelona, Barcelona, Spain
| | - Ganiraju C Manyam
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Yi Miao
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Xiaohong Tan
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Hongwei Zhang
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Alexandar Tzankov
- Institute of Pathology, University Hospital of Basel, Basel, Switzerland
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | | - Wayne Tam
- Weill Cornell Medicine, Cornell University, New York, New York
| | - Hua You
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Govind Bhagat
- New York Presbyterian Hospital/Columbia University Medical Center, New York, New York
| | | | | | | | | | - Miguel A Piris
- Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - J Han van Krieken
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jane N Winter
- Feinberg School of Medicine, Northwestern University, Chicago, Illinois
| | - Jason R Westin
- Department of Lymphoma and Myeloma, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lan V Pham
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - L Jeffrey Medeiros
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - George Z Rassidakis
- Department of Oncology and Pathology, Karolinska Institutet and Karolinska University Hospital, Stockholm, Sweden
| | - Yong Li
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio
| | - Gordon J Freeman
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Ken H Young
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Graduate School of Biomedical Sciences, The University of Texas Health Science Center at Houston, Houston, Texas
| |
Collapse
|
167
|
Yu H, Tian Y, Wang Y, Mineishi S, Zhang Y. Dendritic Cell Regulation of Graft-Vs.-Host Disease: Immunostimulation and Tolerance. Front Immunol 2019; 10:93. [PMID: 30774630 PMCID: PMC6367268 DOI: 10.3389/fimmu.2019.00093] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Graft-vs.-host disease (GVHD) remains a significant cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Significant progresses have been made in defining the dichotomous role of dendritic cells (DCs) in the development of GVHD. Host-derived DCs are important to elicit allogeneic T cell responses, whereas certain donor-types of DCs derived from newly engrafted hematopoietic stem/progenitor cells (HSPCs) can amply this graft-vs.-host reaction. In contrast, some DCs also play non-redundant roles in mediating immune tolerance. They induce apoptotic deletion of host-reactive donor T cells while promoting expansion and function of regulatory T cells (Treg). Unfortunately, this tolerogenic effect of DCs is impaired during GVHD. Severe GVHD in patients subject to allo-HSCT is associated with significantly decreased number of circulating peripheral blood DCs during engraftment. Existing studies reveal that GVHD causes delayed reconstitution of donor DCs from engrafted HSPCs, impairs the antigen presentation function of newly generated DCs and reduces the capacity of DCs to regulate Treg. The present review will discuss the importance of DCs in alloimmunity and the mechanism underlying DC reconstitution after allo-HSCT.
Collapse
Affiliation(s)
- Hongshuang Yu
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Yuanyuan Tian
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Ying Wang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States
| | - Shin Mineishi
- Department of Medicine, Pennsylvania State University, Hershey, PA, United States
| | - Yi Zhang
- Fels Institute for Cancer Research and Molecular Biology, Temple University, Philadelphia, PA, United States,Department of Microbiology & Immunology, Temple University, Philadelphia, PA, United States,*Correspondence: Yi Zhang
| |
Collapse
|
168
|
Wing JB, Tay C, Sakaguchi S. Control of Regulatory T Cells by Co-signal Molecules. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:179-210. [DOI: 10.1007/978-981-32-9717-3_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
169
|
Signal Transduction Via Co-stimulatory and Co-inhibitory Receptors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:85-133. [PMID: 31758532 DOI: 10.1007/978-981-32-9717-3_4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
T-cell receptor (TCR)-mediated antigen-specific stimulation is essential for initiating T-cell activation. However, signaling through the TCR alone is not sufficient for inducing an effective response. In addition to TCR-mediated signaling, signaling through antigen-independent co-stimulatory or co-inhibitory receptors is critically important not only for the full activation and functional differentiation of T cells but also for the termination and suppression of T-cell responses. Many studies have investigated the signaling pathways underlying the function of each molecular component. Co-stimulatory and co-inhibitory receptors have no kinase activity, but their cytoplasmic region contains unique functional motifs and potential phosphorylation sites. Engagement of co-stimulatory receptors leads to recruitment of specific binding partners, such as adaptor molecules, kinases, and phosphatases, via recognition of a specific motif. Consequently, each co-stimulatory receptor transduces a unique pattern of signaling pathways. This review focuses on our current understanding of the intracellular signaling pathways provided by co-stimulatory and co-inhibitory molecules, including B7:CD28 family members, immunoglobulin, and members of the tumor necrosis factor receptor superfamily.
Collapse
|
170
|
Sun P, Jin Q, Nie S, Jia S, Li Y, Li X, Guo F. Unlike PD-L1, PD-1 Is Downregulated on Partial Immune Cells in Type 2 Diabetes. J Diabetes Res 2019; 2019:5035261. [PMID: 31008114 PMCID: PMC6441514 DOI: 10.1155/2019/5035261] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 09/21/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION Type 2 diabetes is a worldwide disease which is associated with chronic, low-grade inflammation. The PD-1/PD-L1 pathway has been reported to be a negative regulatory element in immune homeostasis and to be involved in many diseases. MATERIALS AND METHODS Peripheral blood mononuclear cells (PBMCs) were obtained from type 2 diabetes patients (n = 23) and healthy donors (n = 20). The PD-L1 and PD-1 expressions on corresponding immune cells were evaluated by flow cytometry. RESULTS The PD-L1 expression on corresponding immune cells has no significant difference between these two groups. We showed the downregulated PD-1 expression in type 2 diabetes patients. The correlation analysis indicated that the PD-1 on NK cells has a positive correlation with insulin and diabetes duration. And an inverse correlation has been shown between the PD-1 expression on monocytes and BMI (body mass index). CONCLUSIONS The results in this article suggest that PD-1, unlike PD-L1, might participate in the progression of type 2 diabetes. This investigation will provide evidence for the potential immune therapy for T2D.
Collapse
Affiliation(s)
- Peng Sun
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qingyan Jin
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Nie
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shijie Jia
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yuanyuan Li
- Operation Room of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xiaoxue Li
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Fang Guo
- Department of Intervention Oncology, Shandong Cancer Hospital and Institute, Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
171
|
Co-signal Molecules in T-Cell Activation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1189:3-23. [DOI: 10.1007/978-981-32-9717-3_1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
172
|
Immune checkpoint blockade and its combination therapy with small-molecule inhibitors for cancer treatment. Biochim Biophys Acta Rev Cancer 2018; 1871:199-224. [PMID: 30605718 DOI: 10.1016/j.bbcan.2018.12.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2018] [Revised: 12/13/2018] [Accepted: 12/14/2018] [Indexed: 02/05/2023]
Abstract
Initially understood for its physiological maintenance of self-tolerance, the immune checkpoint molecule has recently been recognized as a promising anti-cancer target. There has been considerable interest in the biology and the action mechanism of the immune checkpoint therapy, and their incorporation with other therapeutic regimens. Recently the small-molecule inhibitor (SMI) has been identified as an attractive combination partner for immune checkpoint inhibitors (ICIs) and is becoming a novel direction for the field of combination drug design. In this review, we provide a systematic discussion of the biology and function of major immune checkpoint molecules, and their interactions with corresponding targeting agents. With both preclinical studies and clinical trials, we especially highlight the ICI + SMI combination, with its recent advances as well as its application challenges.
Collapse
|
173
|
Phung CD, Nguyen HT, Tran TH, Choi HG, Yong CS, Kim JO. Rational combination immunotherapeutic approaches for effective cancer treatment. J Control Release 2018; 294:114-130. [PMID: 30553850 DOI: 10.1016/j.jconrel.2018.12.020] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 12/18/2022]
Abstract
Immunotherapy is an important mode of cancer treatment. Over the past decades, immunotherapy has improved the clinical outcome for cancer patients. However, in many cases, mutations in cancer cells, lack of selectivity, insufficiency of tumor-reactive T cells, and host immunosuppression limit the clinical benefit of immunotherapy. Combination approaches in immunotherapy may overcome these obstacles. Accumulating evidence demonstrates that combination immunotherapy is the future of cancer treatment. However, designing safe and rational combinations of immunotherapy with other treatment modalities is critical. This review will discuss the optimal immunotherapy-based combinations mainly with respect to the mechanisms of action of individual therapeutic agents that target multiple steps in evasion and progression of tumor.
Collapse
Affiliation(s)
- Cao Dai Phung
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Hanh Thuy Nguyen
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Tuan Hiep Tran
- Department for Management of Science and Technology Development, Ton Duc Thang University, Ho Chi Minh City, Viet Nam; Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Viet Nam
| | - Han-Gon Choi
- College of Pharmacy, Institute of Pharmaceutical Science and Technology, Hanyang University, 55, Hanyangdaehak-ro, Sangnok-gu, Ansan 426-791, Republic of Korea
| | - Chul Soon Yong
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea
| | - Jong Oh Kim
- College of Pharmacy, Yeungnam University, 280 Deahak-ro, Gyeongsan 38541, Republic of Korea.
| |
Collapse
|
174
|
Liu S, Kong P, Wang X, Yang L, Jiang C, He W, Quan Q, Huang J, Xie Q, Xia X, Zhang B, Xia L. Tumor microenvironment classification based on T-cell infiltration and PD-L1 in patients with mismatch repair-proficient and -deficient colorectal cancer. Oncol Lett 2018; 17:2335-2343. [PMID: 30675299 PMCID: PMC6341814 DOI: 10.3892/ol.2018.9826] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 11/21/2018] [Indexed: 12/12/2022] Open
Abstract
The classification of tumor microenvironments according to the presence or absence of tumor infiltrating lymphocytes (TILs) and programmed death ligand-1 (PD-L1) expression has been used to predict the efficacy of immune checkpoint inhibitor antibodies in several cancer types, not including colorectal cancer (CRC). The current study investigated the TIL/PD-L1 status of patients with CRC, particularly patients who presented as mismatch repair-proficient (pMMR) and mismatch repair-deficient (dMMR). A total of 243 patients with CRC were enrolled and defined as pMMR (121 patients) or dMMR (122 patients). Using Pearson's χ2 test and multivariable multinomial logistic regression analysis, the associations between MMR status, TIL presence and PD-L1 expression were investigated, in addition to the association between TIL/PD-L1 status and clinicopathological features. The results demonstrated that the dMMR group more frequently exhibited TIL+ (85/122 vs. 61/121) and PD-L1+ (49/122 vs. 32/121) phenotypes compared with the pMMR group. PD-L1+ expression was identified in 42.4% of TIL+ cases in the dMMR group, while only 18.0% of TIL+ cases were PD-L1+ in the pMMR group. High programmed death-1 expression and dMMR status were revealed as two independent risk factors for TIL+ PD-L1+ status. In conclusion, compared with the pMMR group, the dMMR group was more likely to present with a TIL+ PD-L1+ status, which suggests that a TIL+ PD-L1+ tumor microenvironment may partly contribute to the improved response of dMMR patients to anti-PD-1/L1 therapy.
Collapse
Affiliation(s)
- Shousheng Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Pengfei Kong
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaopai Wang
- Department of Pathology, Guangzhou First People's Hospital, Guangzhou Medical University, Guangzhou, Guangdong 510080, P.R. China
| | - Lin Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Chang Jiang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Wenzhuo He
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Qi Quan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Jinsheng Huang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Qiankun Xie
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Xiaojun Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Bei Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| | - Liangping Xia
- State Key Laboratory of Oncology in South China, Collaborative Innovation Centre for Cancer Medicine, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China.,Department of The VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong 510060, P.R. China
| |
Collapse
|
175
|
Cui C, Su M, Lin Y, Lai L. A CD300c-Fc Fusion Protein Inhibits T Cell Immunity. Front Immunol 2018; 9:2657. [PMID: 30498497 PMCID: PMC6249344 DOI: 10.3389/fimmu.2018.02657] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/29/2018] [Indexed: 11/13/2022] Open
Abstract
T cell responses are fine-tuned by co-stimulatory and co-inhibitory molecules. Among the T cell regulators, the B7 family members are of central importance. The recent success in targeting the B7 family molecules for the treatment of immune-related diseases has attracted intense interest in identifying additional B7-related molecules. In this study, we describe CD300c as a novel T cell co-inhibitory molecule that shares significant sequence homology with existing B7 family members. CD300c protein is expressed on professional antigen-presenting cells (APC), including B cells, monocytes, macrophages, and dendritic cells (DCs). The putative CD300c counter-receptor is expressed on CD4 and CD8 T cells, and the expression levels are upregulated upon activation. Soluble human and mouse CD300c-Fc fusion proteins significantly inhibit the proliferation, activation, and cytokine production by CD4 and CD8 T cells in vitro. Administration of CD300c-Fc protein attenuates graft-vs.-host disease (GVHD) in mice. Our results suggest that therapeutic interaction with the CD300c inhibitory pathway may represent a new strategy to modulate T cell-mediated immunity for the treatment of GVHD and autoimmune disease.
Collapse
Affiliation(s)
- Cheng Cui
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States.,Department of Physiology, College of Basic Medical Science, China Medical University, Shenyang, China
| | - Min Su
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Yujun Lin
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States
| | - Laijun Lai
- Department of Allied Health Sciences, University of Connecticut, Storrs, CT, United States.,University of Connecticut Stem Cell Institute, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
176
|
CRISPR/Cas9 for Cancer Therapy: Hopes and Challenges. Biomedicines 2018; 6:biomedicines6040105. [PMID: 30424477 PMCID: PMC6315587 DOI: 10.3390/biomedicines6040105] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/02/2018] [Accepted: 11/05/2018] [Indexed: 12/13/2022] Open
Abstract
Cancer is the second leading cause of death globally and remains a major economic and social burden. Although our understanding of cancer at the molecular level continues to improve, more effort is needed to develop new therapeutic tools and approaches exploiting these advances. Because of its high efficiency and accuracy, the CRISPR-Cas9 genome editing technique has recently emerged as a potentially powerful tool in the arsenal of cancer therapy. Among its many applications, CRISPR-Cas9 has shown an unprecedented clinical potential to discover novel targets for cancer therapy and to dissect chemical-genetic interactions, providing insight into how tumours respond to drug treatment. Moreover, CRISPR-Cas9 can be employed to rapidly engineer immune cells and oncolytic viruses for cancer immunotherapeutic applications. Perhaps more importantly, the ability of CRISPR-Cas9 to accurately edit genes, not only in cell culture models and model organisms but also in humans, allows its use in therapeutic explorations. In this review, we discuss important considerations for the use of CRISPR/Cas9 in therapeutic settings and major challenges that will need to be addressed prior to its clinical translation for a complex and polygenic disease such as cancer.
Collapse
|
177
|
Guo PD, Sun ZW, Lai HJ, Yang J, Wu PP, Guo YD, Sun J. Clinicopathological analysis of PD-L2 expression in colorectal cancer. Onco Targets Ther 2018; 11:7635-7642. [PMID: 30464512 PMCID: PMC6219115 DOI: 10.2147/ott.s177329] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background (PD-L2), a ligand of programmed cell death protein 1 (PD-1), is an inhibitory receptor of T cells and activated B cells. Many studies have focused on PD-L1, another ligand of PD-1, and the prognostic significance of PD-L1 has been reported in many tumors. However, the expression of PD-L2 in relation to clinical outcomes has not been fully investigated in cancer patients. Patients and methods In this study, we investigated the expression of PD-L2 via immunohistochemistry (IHC) in the pathological specimens of 348 patients treated for colorectal cancer (CRC). Results Strong PD-L2 expression was found in the cancer tissues from 41% of the CRC patients who also had a high TNM stage and carcinoembryonic antigen (CEA) concentration. We also carried out functional studies in vitro, which showed that PD-L2 did not influence the growth of the CRC cell line HCT116, but increased cell invasion. Conclusion Collectively, these findings suggest that PD-L2 may be a potential therapeutic target for CRC.
Collapse
Affiliation(s)
- Peng-Da Guo
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| | - Zhong-Wen Sun
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| | - Hui-Jun Lai
- Department of Ultrasound, Suzhou Traditional Chinese Medicine, Suzhou, Jiangsu 215007, China
| | - Jie Yang
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| | - Ping-Ping Wu
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| | - Yun-Di Guo
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| | - Jing Sun
- Institute of Medical Technology, Suzhou Vocational Health College, Suzhou, Jiangsu 215009, China,
| |
Collapse
|
178
|
Peled M, Dragovich MA, Adam K, Strazza M, Tocheva AS, Vega IE, Mor A. EF Hand Domain Family Member D2 Is Required for T Cell Cytotoxicity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2018; 201:2824-2831. [PMID: 30275048 PMCID: PMC6200634 DOI: 10.4049/jimmunol.1800839] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 12/29/2022]
Abstract
Programmed cell death 1 (PD-1) is a major coinhibitory receptor and a member of the immunological synapse (IS). To uncover proteins that regulate PD-1 recruitment to the IS, we searched for cytoskeleton-related proteins that also interact with PD-1 using affinity purification mass spectrometry. Among these proteins, EF hand domain family member D2 (EFHD2), a calcium binding adaptor protein, was functionally and mechanistically analyzed for its contribution to PD-1 signaling. EFHD2 was required for PD-1 to inhibit cytokine secretion, proliferation, and adhesion of human T cells. Interestingly, EFHD2 was also required for human T cell-mediated cytotoxicity and for mounting an antitumor immune response in a syngeneic murine tumor model. Mechanistically, EFHD2 contributed to IS stability, lytic vesicles trafficking, and granzyme B secretion. Altogether, EFHD2 is an important regulator of T cell cytotoxicity and further studies should evaluate its role in T cell-mediated inflammation.
Collapse
Affiliation(s)
- Michael Peled
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
- Pulmonary Department, The Chaim Sheba Medical Center, Ramat Gan 52620, Israel
| | - Matthew A Dragovich
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Kieran Adam
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Marianne Strazza
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Anna S Tocheva
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016
| | - Irving E Vega
- Department of Translational Science and Molecular Medicine, College of Human Medicine, Michigan State University, Grand Rapids, MI 49503; and
| | - Adam Mor
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY 10016;
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| |
Collapse
|
179
|
Concha-Benavente F, Kansy B, Moskovitz J, Moy J, Chandran U, Ferris RL. PD-L1 Mediates Dysfunction in Activated PD-1 + NK Cells in Head and Neck Cancer Patients. Cancer Immunol Res 2018; 6:1548-1560. [PMID: 30282672 DOI: 10.1158/2326-6066.cir-18-0062] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/25/2018] [Accepted: 09/27/2018] [Indexed: 01/02/2023]
Abstract
Inhibitory immune-checkpoint receptors (ICRs), including programmed death 1 (PD-1), have been characterized as exhaustion markers on T cells that infiltrate the tumor microenvironment (TME) of many cancer types, including head and neck cancer (HNC). However, expression and function of ICRs, including PD-1, on natural killer (NK) cells remains less defined. NK cells are innate immune effector cells that lyse epidermal growth factor receptor-overexpressing HNC cells via cetuximab-mediated antibody-dependent cytotoxicity. Cetuximab is clinically effective but only in 10% to 15% of patients. Therefore, it is necessary to investigate how immunomodulation with cetuximab or PD-1 blockade might enhance NK cell responses in the TME and improve monoclonal antibody therapeutic efficacy. We observed that expression of PD-1 on NK cells marks an activated phenotype, which was suppressed only after binding programmed death ligand-1 (PD-L1). HNC patients who exhibit higher circulating PD-1+ NK cells associate with better clinical outcome, and these cells are enriched in the TME. Cetuximab-mediated NK cell activation increased PD-1 expression on NK cells in vitro, which was confirmed in vivo in a prospective neoadjuvant cetuximab trial. In contrast, PD-L1 ligation of PD-1+ NK cells diminished their activation status, whereas PD-1 blockade increased cetuximab-mediated NK cell activation and cytotoxicity, but only against HNC targets with high PD-L1 expression. Therefore, blocking the PD-1-PD-L1 axis may be a useful strategy to reverse immune evasion of HNC tumors with high PD-L1 expression during cetuximab therapy by reversing NK cell dysfunction.
Collapse
Affiliation(s)
- Fernando Concha-Benavente
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
| | - Benjamin Kansy
- Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Jessica Moskovitz
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jennifer Moy
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Uma Chandran
- Department of Biomedical informatics, University of Pittsburgh, Pennsylvania
| | - Robert L Ferris
- Department of Otolaryngology, University of Pittsburgh, Pittsburgh, Pennsylvania.
- University of Pittsburgh Hillman Cancer Center, Pittsburgh, Pennsylvania
- Department of Immunology, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
180
|
Koppolu V, Rekha Vasigala VK. Checkpoint immunotherapy by nivolumab for treatment of metastatic melanoma. J Cancer Res Ther 2018; 14:1167-1175. [PMID: 30488824 DOI: 10.4103/jcrt.jcrt_1290_16] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Clinical management of metastatic melanoma suffered historically from a lack of effective targeted and immunotherapies due to short-lived clinical responses. Recent advances in our understanding of tumor-immune signaling pathways, discovery of immunosuppressive checkpoints, and subsequent development of antibodies that target these checkpoints reverses the situation to some extent. Two antibodies ipilimumab and nivolumab gained Food and Drug administration approval for the treatment of metastatic melanoma and target two major immunosuppressive checkpoints cytotoxic T lymphocyte antigen and programmed cell death protein 1 (PD-1), respectively. Nivolumab binds to PD-1, prevents PD-1 interaction with ligand Programmed death ligand 1 (PD-L1), and thus releases the T-cell exhaustion events (such as T cell apoptosis, decrease in T cell proliferation, etc.) leading to buildup of potent tumor-specific immune response. Successful Phase I-III results with remarkable antitumor activity and safety led to approval of nivolumab against ipilimumab refractory metastatic melanoma. Nivolumab therapy is exciting in that it not only provides substantial benefit but also provides durable responses. This review focuses on the evolution of immunotherapy leading to nivolumab approval and its potential in treating melanoma either alone or in combination with other therapies.
Collapse
Affiliation(s)
- Veerendra Koppolu
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Veneela Krishna Rekha Vasigala
- Department of General Medicine, Rangaraya Medical College, NTR University of Health Sciences, Vijayawada, Andhra Pradesh, India
| |
Collapse
|
181
|
Webb ES, Liu P, Baleeiro R, Lemoine NR, Yuan M, Wang Y. Immune checkpoint inhibitors in cancer therapy. J Biomed Res 2018; 32:317-326. [PMID: 28866656 PMCID: PMC6163118 DOI: 10.7555/jbr.31.20160168] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 03/23/2017] [Indexed: 12/14/2022] Open
Abstract
In recent years immune checkpoint inhibitors have garnered attention as being one of the most promising types of immunotherapy on the horizon. There has been particular focus on the immune checkpoint molecules, cytotoxic T-lymphocyte antigen-4 (CTLA-4) and programmed cell death protein 1 (PD-1) which have been shown to have potent immunomodulatory effects through their function as negative regulators of T cell activation. CTLA-4, through engagement with its ligands B7-1 (CD80) and B7-2 (CD86), plays a pivotal role in attenuating the activation of naïve and memory T cells. In contrast, PD-1 is primarily involved in modulating T cell activity in peripheral tissues via its interaction with PD-L1 and PD-L2. The discovery of these negative regulators of the immune response was crucial in the development of checkpoint inhibitors. This shifted the focus from developing therapies that targeted activation of the host immune system against cancer to checkpoint inhibitors, which aimed to mediate tumor cell destruction through the removal of coinhibitory signals blocking anti-tumor T cell responses.
Collapse
Affiliation(s)
- Eika S. Webb
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Peng Liu
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Renato Baleeiro
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Nicholas R. Lemoine
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
- . Sino-British Research Centre for Molecular Oncology, National Center for International Research in Cell and Gene Therapy, Zhengzhou University, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| | - Ming Yuan
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
| | - Yaohe Wang
- . Center for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, EC1M 6BQ, UK
- . Sino-British Research Centre for Molecular Oncology, National Center for International Research in Cell and Gene Therapy, Zhengzhou University, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, Henan 450002, China
| |
Collapse
|
182
|
Wyss J, Dislich B, Koelzer VH, Galván JA, Dawson H, Hädrich M, Inderbitzin D, Lugli A, Zlobec I, Berger MD. Stromal PD-1/PD-L1 Expression Predicts Outcome in Colon Cancer Patients. Clin Colorectal Cancer 2018; 18:e20-e38. [PMID: 30389315 DOI: 10.1016/j.clcc.2018.09.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Revised: 06/19/2018] [Accepted: 09/17/2018] [Indexed: 02/08/2023]
Abstract
INTRODUCTION The programmed cell death 1 (PD-1)/programmed cell death ligand 1 (PD-L1) axis plays an important role in controlling immune suppression by down-regulating T effector cell activities, enabling tumor cells to escape from the host's antitumor immunsurveillance. While only a small part of colon cancer cells express PD-L1, we sought to evaluate the differential impact of stromal and epithelial PD-L1 expression of primary tumors and liver metastasis on overall survival (OS) in colon cancer patients. PATIENTS AND METHODS Using a next-generation tissue microarray approach, we assessed both epithelial and stromal PD-L1 expression levels in primary tumors (n = 279) and corresponding liver metastases (n = 14) of colon cancer patients. PD-L1 positivity was graded according to the percentage (0.1%-1%, > 1%, > 5%, > 50%) of tumor cells with membranous PD-L1 expression or as the percentage of positive stroma cells and associated inflammatory infiltrates. We also assessed the interplay between stromal PD-1/PD-L1 and both intratumoral and stromal CD8 count and their impact on outcome. The primary end point was OS. RESULTS Stromal PD-L1 and PD-1 expression were both associated with less aggressive tumor behavior in colon cancer patients, which translated into better OS and disease-free survival, respectively. Conversely, PD-L1 staining in the tumor cells was less frequent than stromal staining and was associated with features of aggressive tumor biology, although without impact on outcome. Interestingly, the PD-L1 staining pattern remained similar between primary tumors and corresponding liver metastases. Stromal PD-1 expression correlated significantly with stromal PD-L1 staining and both intratumoral and stromal CD8 expression. CONCLUSION Stromal PD-1/PD-L1 expression might serve as a prognostic marker in colon cancer patients.
Collapse
Affiliation(s)
- Jacqueline Wyss
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Bastian Dislich
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Viktor H Koelzer
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland; Molecular and Population Genetics Laboratory, University of Oxford, Oxford, UK
| | - José A Galván
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Heather Dawson
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Marion Hädrich
- Departments of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Daniel Inderbitzin
- Departments of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Surgery, Bürgerspital Solothurn, Solothurn, Switzerland
| | - Alessandro Lugli
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Inti Zlobec
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - Martin D Berger
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
183
|
Yang J, Hu L. Immunomodulators targeting the PD-1/PD-L1 protein-protein interaction: From antibodies to small molecules. Med Res Rev 2018; 39:265-301. [PMID: 30215856 DOI: 10.1002/med.21530] [Citation(s) in RCA: 140] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 07/18/2018] [Accepted: 07/23/2018] [Indexed: 02/06/2023]
Abstract
Cancer immunotherapy has made great strides in the recent decade, especially in the area of immune checkpoint blockade. The outstanding efficacy, prolonged durability of effect, and rapid assimilation of anti-PD-1 and anti-PD-L1 monoclonal antibodies in clinical practice have been nothing short of a medical breakthrough in the treatment of numerous malignancies. The major advantages of these therapeutic antibodies over their small molecule counterparts have been their high binding affinity and target specificity. However, antibodies do have their flaws including immune-related toxicities, inadequate pharmacokinetics and tumor penetration, and high cost burden to manufacturers and consumers. These limitations hinder broader clinical applications of the antibodies and have heightened interests in developing the alternative small molecule platform that includes peptidomimetics and peptides to target the PD-1/PD-L1 immune checkpoint system. The progress on these small molecule alternatives has been relatively slow compared to that of the antibodies. Fortunately, recent structural studies of the interactions among PD-1, PD-L1, and their respective antibodies have revealed key hotspots on PD-1 and PD-L1 that may facilitate drug discovery efforts for small molecule immunotherapeutics. This review is intended to discuss key concepts in immuno-oncology, describe the successes and shortcomings of PD-1/PD-L1 antibody-based therapies, and to highlight the recent development of small molecule inhibitors of the PD-1/PD-L1 protein-protein interaction.
Collapse
Affiliation(s)
- Jeffrey Yang
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey
| | - Longqin Hu
- Department of Medicinal Chemistry, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey.,Cancer Pharmacology Program, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey
| |
Collapse
|
184
|
Dewanjee S, Bhattacharjee N. MicroRNA: A new generation therapeutic target in diabetic nephropathy. Biochem Pharmacol 2018; 155:32-47. [DOI: 10.1016/j.bcp.2018.06.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/20/2018] [Indexed: 12/11/2022]
|
185
|
LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 Pathway in Tumor Therapy. THE JOURNAL OF IMMUNOLOGY 2018; 200:375-383. [PMID: 29311378 DOI: 10.4049/jimmunol.1701044] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/05/2017] [Indexed: 12/13/2022]
Abstract
The programmed death 1 (PD-1) pathway delivers inhibitory signals that function as a brake for immune responses. This pathway limits the initiation and duration of immune responses, thereby protecting tissues from immune-mediated damage and autoimmune diseases. However, the PD-1 pathway also inhibits immune responses to tumors. The critical role of PD-1 in preventing antitumor immunity is demonstrated by the transformative effects of PD-1 pathway blockade in a broad range of cancers with the hallmark of durability of response. Despite this success, most patients do not respond to PD-1 monotherapy, and some patients experience adverse events. In this review, we discuss the functions of the PD-1 pathway and its translation to cancer immunotherapy. We also consider current challenges and opportunities for PD-1 cancer immunotherapy, including mechanisms of response and resistance, identification of biomarkers of response to PD-1 therapy, characterization and treatment of PD-1 therapy-related adverse events, and development of safe and effective combination therapies.
Collapse
Affiliation(s)
- Martin W LaFleur
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115.,Department of Pediatric Oncology, Dana Farber Cancer Institute, Boston, MA 02115
| | - Yuki Muroyama
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231.,Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032
| | - Charles G Drake
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY 10032; .,Herbert Irving Comprehensive Cancer Center, Columbia University Medical Center, New York, NY 10032; and
| | - Arlene H Sharpe
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115; .,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115
| |
Collapse
|
186
|
Wei SC, Duffy CR, Allison JP. Fundamental Mechanisms of Immune Checkpoint Blockade Therapy. Cancer Discov 2018; 8:1069-1086. [PMID: 30115704 DOI: 10.1158/2159-8290.cd-18-0367] [Citation(s) in RCA: 2150] [Impact Index Per Article: 307.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 06/04/2018] [Accepted: 07/11/2018] [Indexed: 02/06/2023]
Abstract
Immune checkpoint blockade is able to induce durable responses across multiple types of cancer, which has enabled the oncology community to begin to envision potentially curative therapeutic approaches. However, the remarkable responses to immunotherapies are currently limited to a minority of patients and indications, highlighting the need for more effective and novel approaches. Indeed, an extraordinary amount of preclinical and clinical investigation is exploring the therapeutic potential of negative and positive costimulatory molecules. Insights into the underlying biological mechanisms and functions of these molecules have, however, lagged significantly behind. Such understanding will be essential for the rational design of next-generation immunotherapies. Here, we review the current state of our understanding of T-cell costimulatory mechanisms and checkpoint blockade, primarily of CTLA4 and PD-1, and highlight conceptual gaps in knowledge.Significance: This review provides an overview of immune checkpoint blockade therapy from a basic biology and immunologic perspective for the cancer research community. Cancer Discov; 8(9); 1069-86. ©2018 AACR.
Collapse
Affiliation(s)
- Spencer C Wei
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Colm R Duffy
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas. .,Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
187
|
Kabacaoglu D, Ciecielski KJ, Ruess DA, Algül H. Immune Checkpoint Inhibition for Pancreatic Ductal Adenocarcinoma: Current Limitations and Future Options. Front Immunol 2018; 9:1878. [PMID: 30158932 PMCID: PMC6104627 DOI: 10.3389/fimmu.2018.01878] [Citation(s) in RCA: 135] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 07/30/2018] [Indexed: 12/16/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), as the most frequent form of pancreatic malignancy, still is associated with a dismal prognosis. Due to its late detection, most patients are ineligible for surgery, and chemotherapeutic options are limited. Tumor heterogeneity and a characteristic structure with crosstalk between the cancer/malignant cells and an abundant tumor microenvironment (TME) make PDAC a very challenging puzzle to solve. Thus far, targeted therapies have failed to substantially improve the overall survival of PDAC patients. Immune checkpoint inhibition, as an emerging therapeutic option in cancer treatment, shows promising results in different solid tumor types and hematological malignancies. However, PDAC does not respond well to immune checkpoint inhibitors anti-programmed cell death protein 1 (PD-1) or anti-cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) alone or in combination. PDAC with its immune-privileged nature, starting from the early pre-neoplastic state, appears to escape from the antitumor immune response unlike other neoplastic entities. Different mechanisms how cancer cells achieve immune-privileged status have been hypothesized. Among them are decreased antigenicity and impaired immunogenicity via both cancer cell-intrinsic mechanisms and an augmented immunosuppressive TME. Here, we seek to shed light on the recent advances in both bench and bedside investigation of immunotherapeutic options for PDAC. Furthermore, we aim to compile recent data about how PDAC adopts immune escape mechanisms, and how these mechanisms might be exploited therapeutically in combination with immune checkpoint inhibitors, such as PD-1 or CTLA-4 antibodies.
Collapse
Affiliation(s)
| | | | | | - Hana Algül
- Internal Medicine II, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| |
Collapse
|
188
|
Meyers D, Bryan P, Banerji S, Morris D. Targeting the PD-1/PD-L1 axis for the treatment of non-small-cell lung cancer. Curr Oncol 2018; 25:e324-e334. [PMID: 30111979 PMCID: PMC6092051 DOI: 10.3747/co.25.3976] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Lung cancer is the leading cause of cancer-specific death among Canadians, with non-small-cell lung cancer (nsclc) being the most common histologic variant. Despite advances in the understanding of the molecular biology of nsclc, the survival rate for this malignancy is still poor. It is now understood that, to evade detection and immune clearance, nsclc tumours overexpress the immunosuppressive checkpoint protein programmed death ligand 1 (PD-L1). Inhibiting the PD-1/PD-L1 axis with monoclonal antibodies has significantly changed the treatment landscape in nsclc during the last 5 years. Despite evidence of clinical response in some patients, only approximately 20% of patients obtain any durable benefit, and many of the patients who do respond ultimately relapse with drug-resistant disease. The identification of patients who are most likely to benefit from such therapy is therefore important. In the present review, we cover the basics of the PD-1/PD-L1 axis and its clinical significance in nsclc, biomarkers that are predictive of treatment response, relevant clinical trials of PD-1/PD-L1 blockade completed to date, and proposed mechanisms of acquired therapeutic resistance.
Collapse
Affiliation(s)
- D.E. Meyers
- Department of Oncology, University of Calgary, and
- Tom Baker Cancer Centre, Calgary, AB
| | - P.M. Bryan
- Department of Oncology, University of Calgary, and
| | - S. Banerji
- Department of Internal Medicine, Max Rady College of Medicine, University of Manitoba, and
- Department of Medical Oncology, CancerCare Manitoba, Winnipeg, MB
| | - D.G. Morris
- Department of Oncology, University of Calgary, and
- Tom Baker Cancer Centre, Calgary, AB
| |
Collapse
|
189
|
Chen CY, Hutzen B, Wedekind MF, Cripe TP. Oncolytic virus and PD-1/PD-L1 blockade combination therapy. Oncolytic Virother 2018; 7:65-77. [PMID: 30105219 PMCID: PMC6074764 DOI: 10.2147/ov.s145532] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Oncolytic viruses are lytic for many types of cancers but are attenuated or replication-defective in normal tissues. Aside from tumor lysis, oncolytic viruses can induce host immune responses against cancer cells and may thus be viewed as a form of immunotherapy. Although recent successes with checkpoint inhibitors have shown that enhancing antitumor immunity can be effective, the dynamic nature of the immunosuppressive tumor microenvironment presents significant hurdles to the broader application of these therapies. Targeting one immune-suppressive pathway may not be sufficient to eliminate tumors. Here we focus on the development of the combination of oncolytic virotherapy with checkpoint inhibitors designed to target the programmed cell death protein 1 and programmed cell death ligand 1 signaling axis. We also discuss future directions for the clinical application of this novel combination therapy.
Collapse
Affiliation(s)
- Chun-Yu Chen
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital,
| | - Brian Hutzen
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital,
| | - Mary F Wedekind
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, .,Division of Hematology/Oncology/Blood and Marrow Transplantation, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA,
| | - Timothy P Cripe
- Department of Pediatrics, Center for Childhood Cancer and Blood Diseases, Nationwide Children's Hospital, .,Division of Hematology/Oncology/Blood and Marrow Transplantation, Nationwide Children's Hospital, The Ohio State University, Columbus, OH, USA,
| |
Collapse
|
190
|
Acúrcio RC, Scomparin A, Conniot J, Salvador JAR, Satchi-Fainaro R, Florindo HF, Guedes RC. Structure–Function Analysis of Immune Checkpoint Receptors to Guide Emerging Anticancer Immunotherapy. J Med Chem 2018; 61:10957-10975. [DOI: 10.1021/acs.jmedchem.8b00541] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Rita C. Acúrcio
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Anna Scomparin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - João Conniot
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Jorge A. R. Salvador
- Laboratory of Pharmaceutical Chemistry, Faculty of Pharmacy, and Centre for Neuroscience and Cell Biology, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 6997801 Tel Aviv, Israel
| | - Helena F. Florindo
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Rita C. Guedes
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| |
Collapse
|
191
|
Mushtaq MU, Papadas A, Pagenkopf A, Flietner E, Morrow Z, Chaudhary SG, Asimakopoulos F. Tumor matrix remodeling and novel immunotherapies: the promise of matrix-derived immune biomarkers. J Immunother Cancer 2018; 6:65. [PMID: 29970158 PMCID: PMC6029413 DOI: 10.1186/s40425-018-0376-0] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 06/15/2018] [Indexed: 02/08/2023] Open
Abstract
Recent advances in our understanding of the dynamics of cellular cross-talk have highlighted the significance of host-versus-tumor effect that can be harnessed with immune therapies. Tumors exploit immune checkpoints to evade adaptive immune responses. Cancer immunotherapy has witnessed a revolution in the past decade with the development of immune checkpoint inhibitors (ICIs), monoclonal antibodies against cytotoxic T lymphocyte antigen 4 (CTLA-4) and programmed cell death protein 1 (PD-1) or their ligands, such as PD1 ligand 1 (PD-L1). ICIs have been reported to have activity against a broad range of tumor types, in both solid organ and hematologic malignancy contexts. However, less than one-third of the patients achieve a durable and meaningful treatment response. Expression of immune checkpoint ligands (e.g., PD-L1), mutational burden and tumor-infiltrating lymphocytes are currently used as biomarkers for predicting response to ICIs. However, they do not reliably predict which patients will benefit from these therapies. There is dire need to discover novel biomarkers to predict treatment efficacy and to identify areas for development of combination strategies to improve response rates. Emerging evidence suggests key roles of tumor extracellular matrix (ECM) components and their proteolytic remodeling products in regulating each step of the cancer-immunity cycle. Here we review tumor matrix dynamics and matrix remodeling in context of anti-tumor immune responses and immunotherapy and propose the exploration of matrix-based biomarkers to identify candidates for immune therapy.
Collapse
Affiliation(s)
- Muhammad Umair Mushtaq
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Athanasios Papadas
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Adam Pagenkopf
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Evan Flietner
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Zachary Morrow
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Sibgha Gull Chaudhary
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA
| | - Fotis Asimakopoulos
- Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA. .,University of Wisconsin Carbone Cancer Center, 1111 Highland Avenue, WIMR 4031, Madison, WI, 53705, USA.
| |
Collapse
|
192
|
Czaja AJ. Under-Evaluated or Unassessed Pathogenic Pathways in Autoimmune Hepatitis and Implications for Future Management. Dig Dis Sci 2018; 63:1706-1725. [PMID: 29671161 DOI: 10.1007/s10620-018-5072-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 04/12/2018] [Indexed: 12/11/2022]
Abstract
Autoimmune hepatitis is a consequence of perturbations in homeostatic mechanisms that maintain self-tolerance but are incompletely understood. The goals of this review are to describe key pathogenic pathways that have been under-evaluated or unassessed in autoimmune hepatitis, describe insights that may shape future therapies, and encourage investigational efforts. The T cell immunoglobulin mucin proteins constitute a family that modulates immune tolerance by limiting the survival of immune effector cells, clearing apoptotic bodies, and expanding the population of granulocytic myeloid-derived suppressor cells. Galectins influence immune cell migration, activation, proliferation, and survival, and T cell exhaustion can be induced and exploited as a possible management strategy. The programmed cell death-1 protein and its ligands comprise an antigen-independent inhibitory axis that can limit the performance of activated T cells by altering their metabolism, and epigenetic changes can silence pro-inflammatory genes or de-repress anti-inflammatory genes that affect disease severity. Changes in the intestinal microbiota and permeability of the intestinal mucosal barrier can be causative or consequential events that affect the occurrence and phenotype of immune-mediated disease, and they may help explain the female propensity for autoimmune hepatitis. Perturbations within these homeostatic mechanisms have been implicated in experimental models and limited clinical experiences, and they have been favorably manipulated by monoclonal antibodies, recombinant molecules, pharmacological agents or dietary supplements. In conclusion, pathogenic mechanisms that have been implicated in other systemic immune-mediated and liver diseases but under-evaluated or unassessed in autoimmune hepatitis warrant consideration and rigorous evaluation.
Collapse
Affiliation(s)
- Albert J Czaja
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street S.W., Rochester, MN, 55905, USA.
| |
Collapse
|
193
|
Hsieh CC, Hsu HS, Li AFY, Chen YJ. Clinical relevance of PD-L1 and PD-L2 overexpression in patients with esophageal squamous cell carcinoma. J Thorac Dis 2018; 10:4433-4444. [PMID: 30174892 DOI: 10.21037/jtd.2018.06.167] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Even with the advance of diagnosis and the treatment, the 5-year survival rate for esophageal cancer patients is still poor. The checkpoint protein inhibition provides another choice to improve the survival. The expression of the programmed death ligand-1 (PD-L1) was reported but the clinical relevance remained inconsistent in esophageal cancer. Besides, there were few references about the other ligand, programed death ligand-2 (PD-L2). In this study, we evaluated the expressions of PD-L1 and PD-L2 in patients with esophageal squamous cell carcinoma (ESCC) and assessed their clinical relevance. Methods From 1996 to 2011, 150 patients undergone complete surgical resection for ESCC were enrolled. Clinical data were recorded. Expression of PD-L1 and PD-L2 on cytoplasm in paraffin embedded tumor samples were analyzed by immunohistochemistry staining and scored with a semi-quantitative method. Results Of the patients, 96 (64.0%) patients had PD-L1 overexpression and 63 (42.0%) had PD-L2 overexpression. There was a correlation between the expression of PD-L1 and PD-L2 (P<0.001). Patients without overexpression of PD-L1, pathological T1-2 and N0 status, pathological stage I-II and no post-operative adjuvant treatment had a better disease free survival (DFS). In multivariate analysis, PD-L1 expression and pathological stage were the independent prognostic factors for DFS. The expression of PD-L2 did not influence the DFS. Although not statistically significant, patients without overexpression of PD-L1 and PD-L2 seem to have a better overall survival (OS). Conclusions The overexpression of PD-L1 on cytoplasm, not PD-L2, is an independent prognostic factor for DFS in patients with ESCC undergone esophagectomy. However, there is a trend which suggested that patients without overexpression of PD-L1 and PD-L2 had a better OS.
Collapse
Affiliation(s)
- Chih-Cheng Hsieh
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei.,Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei
| | - Han-Shui Hsu
- Division of Thoracic Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei.,Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang-Ming University, Taipei
| | - Anna Fen-Yau Li
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei.,School of Medicine, National Yang-Ming University, Taipei
| | - Yann-Jang Chen
- Institute of Clinical Medicine, School of Medicine, National Yang-Ming University, Taipei.,Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei.,Department of Pediatrics, Renai Branch, Taipei City Hospital, Taipei
| |
Collapse
|
194
|
Czaja AJ. Emerging therapeutic biomarkers of autoimmune hepatitis and their impact on current and future management. Expert Rev Gastroenterol Hepatol 2018. [PMID: 29540068 DOI: 10.1080/17474124.2018.1453356] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autoimmune hepatitis lacks a quantifiable biomarker that is close to its pathogenic mechanisms and that accurately reflects inflammatory activity, correlates with treatment response, and ensures inactive disease before treatment withdrawal. Areas covered: Micro-ribonucleic acids, programmed death-1 protein and its ligands, macrophage migration inhibitory factor, soluble CD163, B cell activating factor, and metabolite patterns in blood were considered the leading candidates as therapeutic biomarkers after search of PubMed from August 1981 to August 2017 using the search words 'biomarkers of autoimmune hepatitis'. Expert commentary: Each of the candidate biomarkers is close to the putative pathogenic mechanisms of autoimmune hepatitis, and each has attributes that support its potential role as a surrogate marker of inflammatory activity that can be monitored during treatment. Future studies must demonstrate the superiority of each biomarker to conventional indices of inflammatory activity and validate their correlation with treatment response and outcome. A reliable therapeutic biomarker would facilitate the individualization of current management algorithms, ensure that pathogenic mechanisms were disrupted or eliminated prior to treatment withdrawal, and reduce the frequency of relapse or unnecessary protracted therapy. The biomarker might also prove to be a target of next-generation therapies.
Collapse
Affiliation(s)
- Albert J Czaja
- a Division of Gastroenterology and Hepatology , Mayo Clinic College of Medicine and Science , Rochester , MN , USA
| |
Collapse
|
195
|
El-Murr T, Patel A, Sedlak C, D'Souza-Lobo B. Evaluating dendritic cells as an in vitro screening tool for immunotherapeutic formulations. J Immunol Methods 2018; 459:55-62. [PMID: 29800576 DOI: 10.1016/j.jim.2018.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Revised: 04/09/2018] [Accepted: 05/09/2018] [Indexed: 01/24/2023]
Abstract
Immunotherapy approaches targeting dendritic cells (DCs) are being studied as treatment options in cancer. This project focused on utilizing DCs as a valuable in vitro screening tool for efficacious microparticle formulations containing tumor associated antigens (TAAs) and adjuvants as immunotherapy alternatives. The innate immune system, including DCs, distinctly responds to the particulate matter and adjuvants in these formulations which stimulates the adaptive immune system to eliminate resident cancer cells. We formulated microparticles (MPs) co-loaded with TAAs along with the adjuvants, AddaVax™ and Imiquimod, and measured their effect on DCs in eliciting a cell-mediated immune response towards tumors. The MP zeta potential was measured as -24.0 mV and -26.5 mV for blank and TAA/adjuvant co-loaded microparticles, and the average particle size was 671.2 nm and 854.4 nm respectively. We determined that nitric oxide (NO) secretion was significantly higher in the adjuvant MP treated DCs group and was dose dependent with 1 mg/mL demonstrating the highest secretion levels. TNF-α release was highest in AddaVax™/TAA and Imiquimod/TAA MPs treated DCs, while IL-6 secretion was highest from Imiquimod/TAA MPs as well as from combined AddaVax™/TAA and Imiquimod/TAA MPs. Overall, the cell surface marker expressions of CD80, CD86, CD40, CD54, MHC-I and MHC-II levels were highest with combined AddaVax™/TAA and Imiquimod/TAA MPs. The results of our experiments suggest that a combination of adjuvants targeting different DC receptors loaded with TAA MPs creates an efficient delivery system to T-cells that could improve adaptive immune responses. Our studies also confirm that DCs are potent innate immune cells that can be used successfully as an in vitro tool to screen novel delivery formulations focused on immunotherapy.
Collapse
Affiliation(s)
- Theresa El-Murr
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Ankita Patel
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Carrie Sedlak
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States
| | - Bernadette D'Souza-Lobo
- McWhorter School of Pharmacy, Samford University, 800 Lakeshore Dr, Birmingham, AL 35229, United States.
| |
Collapse
|
196
|
Kim TK, Herbst RS, Chen L. Defining and Understanding Adaptive Resistance in Cancer Immunotherapy. Trends Immunol 2018; 39:624-631. [PMID: 29802087 DOI: 10.1016/j.it.2018.05.001] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/01/2018] [Accepted: 05/03/2018] [Indexed: 12/31/2022]
Abstract
Despite the unprecedented tumor regression and long-term survival benefit observed with anti-programmed death (PD) [anti-PD-1 or anti-B7-homolog 1 (B7-H1)] therapy in patients with advanced cancers, a large portion of patients do not benefit from such treatment and a fraction of responders relapse. Current efforts to overcome resistance and improve efficacy of anti-PD therapy require a clear understanding of resistance and should precede current avenues using random combinations with available treatment regimens. Here, we categorized three types of resistance, namely target-missing, primary, and acquired resistance. This categorization requires reliable, accurate tissue sampling and appropriate interpretation of results based on the four classifications of tumor immunity in the microenvironment (TIME). We believe that fundamental understanding of these complex tumor-immune interactions and of the cellular and molecular mechanisms underlying these types of true resistance is the key for targeting the right targets in combination with or beyond anti-PD therapy in the future.
Collapse
Affiliation(s)
- Tae Kon Kim
- Section of Hematology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Roy S Herbst
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lieping Chen
- Section of Medical Oncology, Department of Medicine, Yale University School of Medicine, New Haven, CT, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
197
|
Chae YK, Arya A, Iams W, Cruz MR, Chandra S, Choi J, Giles F. Current landscape and future of dual anti-CTLA4 and PD-1/PD-L1 blockade immunotherapy in cancer; lessons learned from clinical trials with melanoma and non-small cell lung cancer (NSCLC). J Immunother Cancer 2018; 6:39. [PMID: 29769148 PMCID: PMC5956851 DOI: 10.1186/s40425-018-0349-3] [Citation(s) in RCA: 308] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 05/02/2018] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy is among the most rapidly evolving treatment strategies in oncology. The therapeutic potential of immune-checkpoint inhibitors is exemplified by the recent hail of Food and Drug Administration (FDA) approvals for their use in various malignancies. Continued efforts to enhance outcomes with immunotherapy agents have led to the formulation of advanced treatment strategies. Recent evidence from pre-clinical studies evaluating immune-checkpoint inhibitors in various cancer cell-lines has suggested that combinatorial approaches may have superior survival outcomes compared to single-agent immunotherapy regimens. Preliminary trials assessing combination therapy with anti-PD-1/PD-L1 plus anti-CTLA-4 immune-checkpoint inhibitors have documented considerable advantages in survival indices over single-agent immunotherapy. The therapeutic potential of combinatorial approaches is highlighted by the recent FDA approval of nivolumab plus ipilimumab for patients with advanced melanoma. Presently, dual-immune checkpoint inhibition with anti-programmed death receptor-1/programmed cell death receptor- ligand-1 (anti-PD-1/PD-L1) plus anti-cytotoxic T lymphocyte associated antigen-4 (anti-CTLA-4) monoclonal antibodies (MoAbs) is being evaluated for a wide range of tumor histologies. Furthermore, several ongoing clinical trials are investigating combination checkpoint inhibition in association with traditional treatment modalities such as chemotherapy, surgery, and radiation. In this review, we summarize the current landscape of combination therapy with anti-PD-1/PD-L1 plus anti-CTLA-4 MoAbs for patients with melanoma and non-small cell lung cancer (NSCLC). We present a synopsis of the prospects for expanding the indications of dual immune-checkpoint inhibition therapy to a more diverse set of tumor histologies.
Collapse
Affiliation(s)
- Young Kwang Chae
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA. .,Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA. .,Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL, 60611, USA.
| | - Ayush Arya
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Wade Iams
- 0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Marcelo R. Cruz
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Sunandana Chandra
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Jaehyuk Choi
- 0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| | - Francis Giles
- Developmental Therapeutics Program of the Division of Hematology Oncology, Early Phase Clinical Trials Unit, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Robert H. Lurie Comprehensive Cancer Center of Northwestern University, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA ,0000 0001 2299 3507grid.16753.36Northwestern University Feinberg School of Medicine, 645 N. Michigan Avenue, Suite 1006, Chicago, IL 60611 USA
| |
Collapse
|
198
|
Smith-Cohn MA, Gill D, Voorhies BN, Agarwal N, Garrido-Laguna I. Case report: pembrolizumab-induced Type 1 diabetes in a patient with metastatic cholangiocarcinoma. Immunotherapy 2018; 9:797-804. [PMID: 28877632 DOI: 10.2217/imt-2017-0042] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Immune checkpoint inhibitors are novel cancer therapies associated with numerous autoimmune toxicities, some of which are only now being appreciated. CASE PRESENTATION A 67-year old female with metastatic cholangiocarcinoma and no prior history of diabetes was treated with leucovorin, fluorouracil, oxaliplatin and pembrolizumab. After eight cycles, she developed new onset type 1 diabetes mellitus with positive glutamic acid decarboxylase antibody titers. Conclusions: To our knowledge, this is the first reported case of PD-1 inhibitor associated Type 1 diabetes mellitus in a patient with cholangiocarcinoma and supports others' experiences that PD-1 inhibition can cause a spectrum of autoimmune adverse events that require clinical monitoring and periodic screenings.
Collapse
Affiliation(s)
- Matthew A Smith-Cohn
- Department of Neurology, University of Utah, Salt Lake City, 175 North Medical Drive East, UT 84132, USA
| | - David Gill
- Department of Hematology and Oncology, University of Utah School of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Benjamin N Voorhies
- Department of Hematology and Oncology, University of Utah School of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Neeraj Agarwal
- Department of Hematology and Oncology, University of Utah School of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| | - Ignacio Garrido-Laguna
- Department of Hematology and Oncology, University of Utah School of Medicine, Huntsman Cancer Institute, 2000 Circle of Hope, Salt Lake City, UT 84112, USA
| |
Collapse
|
199
|
Abstract
INTRODUCTION The prognosis of recurrent/metastatic (R/M) squamous cell carcinoma of the head and neck (HNSCC) after failure of first line chemotherapy is dismal. Until the publication of the results of CheckMate 141, not a single agent provided any survival benefit as a second line treatment for R/M HNSCC. Areas covered: A comprehensive review of the literature was conducted on the role of nivolumab in HNSCC. Expert commentary: Nivolumab is approved by the Food and Drug Administration for the treatment of patients based on the results of CheckMate 141 showing an overall survival benefit as compared to standard care (single agent docetaxel, methotrexate, or cetuximab). Of particular interest are immune-related adverse events which should be managed according to published guidelines.
Collapse
Affiliation(s)
- Pol Specenier
- a Department of Oncology , Antwerp University Hospital , Edegem , Belgium
| |
Collapse
|
200
|
Yao Q, Fischer KP, Tyrrell DL, Gutfreund KS. The Pekin duck programmed death ligand-2: cDNA cloning, genomic structure, molecular characterization and expression analysis. Biochem Biophys Rep 2018; 13:116-122. [PMID: 29556566 PMCID: PMC5857182 DOI: 10.1016/j.bbrep.2018.01.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2016] [Accepted: 01/22/2018] [Indexed: 11/27/2022] Open
Abstract
Programmed death-1 (PD-1), upon engagement by its ligands, programmed death ligand-1 (PD-L1) and programmed death ligand-2 (PD-L2), provides signals that attenuate adaptive immune responses. Here we describe the identification of the Pekin duck PD-L2 (duPD-L2) and its gene structure. The duPD-L2 cDNA encodes a 321 amino acid protein that has an amino acid identity of 76% and 35% with chicken and human PD-L2, respectively. Mapping of the duPD-L2 cDNA with duck genomic sequences revealed an exonic structure similar to that of the human Pdcd1lg2 gene. Homology modelling of the duPD-L2 protein was compatible with the murine PD-L2 ectodomain structure. Residues known to be important for PD-1 receptor binding of murine PD-L2 were mostly conserved in duPD-L2 within sheets A and G and partially conserved within sheets C and F. DuPD-L2 mRNA was constitutively expressed in all tissues examined with highest expression levels in lung, spleen, cloaca, bursa, cecal tonsil, duodenum and very low levels of expression in muscle, kidney and brain. Lipopolysaccharide treatment of adherent duck PBMC upregulated duPD-L2 mRNA expression. Our work shows evolutionary conservation of the PD-L2 ectodomain structure and residues important for PD-1 binding in vertebrates including fish. The information provided will be useful for further investigation of the role of duPD-L2 in the regulation of duck adaptive immunity and exploration of PD-1-targeted immunotherapies in the duck hepatitis B infection model.
Collapse
Affiliation(s)
- Qingxia Yao
- Depts. of Medicine, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Karl P Fischer
- Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - D Lorne Tyrrell
- Depts. of Medicine, University of Alberta, Edmonton, AB, Canada.,Medical Microbiology & Immunology, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| | - Klaus S Gutfreund
- Depts. of Medicine, University of Alberta, Edmonton, AB, Canada.,Li Ka Shing Institute of Virology, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|