1
|
Cho Y, Ahn S, Kim KM. PD-L1 as a Biomarker in Gastric Cancer Immunotherapy. J Gastric Cancer 2025; 25:177-191. [PMID: 39822174 PMCID: PMC11739645 DOI: 10.5230/jgc.2025.25.e4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 11/29/2024] [Indexed: 01/19/2025] Open
Abstract
Combining chemotherapy with immune checkpoint inhibitors (ICIs) that target the programmed death-1 (PD-1) protein has been shown to be a clinically effective first-line treatment for human epidermal growth factor receptor 2 (HER2)-negative and -positive advanced or metastatic gastric cancer (GC). Currently, PD-1 inhibitors combined with chemotherapy are the standard treatment for patients with HER2-negative/positive locally advanced or metastatic GC. Programmed death-ligand 1 (PD-L1) expression, as assessed using immunohistochemistry (IHC), is a crucial biomarker for predicting response to anti-PD-1/PD-L1 agents in various solid tumors, including GC. In GC, the PD-L1 IHC test serves as a companion or complementary diagnostic test for immunotherapy, and an accurate interpretation of PD-L1 status is essential for selecting patients who may benefit from immunotherapy. However, PD-L1 IHC testing presents several challenges that limit its reliability as a biomarker for immunotherapy. In this review, we provide an overview of the current practices of immunotherapy and PD-L1 testing in GC. In addition, we discuss the clinical challenges associated with PD-L1 testing and its future use as a biomarker for immunotherapy. Finally, we present prospective biomarkers currently under investigation as alternative predictors of immunotherapy response in GC.
Collapse
Affiliation(s)
- Yunjoo Cho
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
2
|
Harel E, Hewer E, La Rosa S, Brouland JP, Pitteloud N, Santoni F, Brunner M, Daniel RT, Messerer M, Cossu G. PD-L1 expression in PitNETs: Correlations with the 2022 WHO classification. BRAIN & SPINE 2024; 5:104171. [PMID: 39845357 PMCID: PMC11751414 DOI: 10.1016/j.bas.2024.104171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 12/15/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025]
Abstract
Introduction and research question: Prognostic factors to predict the behavior of pituitary neuroendocrine tumors (PitNET) are scarce. PD-L1 expression was associated with prognosis in other neuroendocrine neoplasms and we analyzed PD-L1 expression in PitNET, according to the 2022 WHO classification. Material and methods A retrospective analysis was performed. Immunohistochemistry was used to define PD-L1 expression, which was quantified as TPS (tumor proportion score). The primary outcome was to assess the correlation between PD-L1 expression and transcription factors (TF), namely T-pit, Pit-1, SF-1 and GATA-3. As secondary outcomes, we evaluated the association between PD-L1 expression and proliferation indexes. Results Eighty-eight patients were included. The largest group belonged to the SF-1-lineage (48%), followed by tumors of the Pit-1 lineage (32%) and T-pit lineage (17%). PD-L1 expression was associated with Pit-1 expression (p < 0.001) and with the somatotroph, lactotroph and mammosomatotroph subgroups. A TPS ⩾35% showed a 100% sensitivity for the mammosomatotroph subtype, while the optimal cut-off point was 20% for somatotroph and 15% for lactotroph tumors. PD-L1 expression was negatively associated with SF-1 and GATA3 expression(p < 0.001), with an optimal cut-point ≤5%. No association was found between PD-L1 expression and immunohistochemical proliferative factors but PD-L1 expression was associated with female sex and a younger age at diagnosis. Conclusion PD-L1 expression was associated with PIT-1 lineage, while it was downregulated in SF-1-lineage tumors. No correlation was found with proliferative factors. The role of PD-L1 expression in determining the biological behavior of PitNET remains debated and larger studies are necessary to further confirm these findings.
Collapse
Affiliation(s)
- Ethan Harel
- Division of Neurosurgery, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Ekkehard Hewer
- Department of Laboratory Medicine and Pathology, Institute of Pathology, University of Lausanne, Lausanne, Switzerland
| | - Stefano La Rosa
- Unit of Pathology, Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | - Jean Philippe Brouland
- Department of Laboratory Medicine and Pathology, Institute of Pathology, University of Lausanne, Lausanne, Switzerland
| | - Nelly Pitteloud
- Service of Endocrinology Diabetes and Metabolism, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Federico Santoni
- Service of Endocrinology Diabetes and Metabolism, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Maxime Brunner
- Service of Endocrinology Diabetes and Metabolism, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Roy Thomas Daniel
- Division of Neurosurgery, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Mahmoud Messerer
- Division of Neurosurgery, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| | - Giulia Cossu
- Division of Neurosurgery, Department of Clinical Neurosciences, University Hospital of Lausanne and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
3
|
Ugolini F, Tinunin L, Nozzoli F, Simi S, Di Gangi D, Baroni G, Antonini P, Szumera-Ciećkiewicz A, Massi D. Brightfield Multiplex Immunohistochemistry Assay for PD-L1 Evaluation in Challenging Melanoma Samples. Appl Immunohistochem Mol Morphol 2024; 32:389-394. [PMID: 39166464 PMCID: PMC11371108 DOI: 10.1097/pai.0000000000001219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/16/2024] [Indexed: 08/23/2024]
Abstract
Targeting the PD1/PD-L1 immune checkpoint pathway has rapidly become a therapeutic strategy for melanoma patients. Indeed, the quantification of PD-L1 expression by immunohistochemistry (IHC) in melanoma samples is already required, in some contexts, to allow access to anti-PD-1/PD-L1 immunotherapy. Despite a rising demand for PD-L1 testing, paralleling increasing cumulative experience in its assessment and quantification, it is fair to recognize that PD-L1 evaluation in melanoma samples still presents some critical issues. The aim of this technical report is to develop and validate a multiplex double staining protocol for PD-L1/SOX10 in Ventana Benchmark Ultra for routine practice. Our results show that double labeling provides the necessary tools to identify PD-L1 + melanoma cells clearly. The simultaneous visualization of 2 different proteins targets allows the topographical relationship between the 2 labeling to be evaluated within the context of the tissue morphology. Future studies are needed to test this technique's real-world applicability and effectiveness in implementing interpathologist agreement.
Collapse
Affiliation(s)
| | - Luca Tinunin
- Section of Pathology, Department of Health Sciences
| | | | - Sara Simi
- Section of Pathology, Department of Health Sciences
| | | | | | - Pietro Antonini
- Section of Pathology, Department of Health Sciences
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, Verona, Italy
| | - Anna Szumera-Ciećkiewicz
- Department of Pathology, Maria Sklodowska-Curie National Research Institute of Oncology
- Maria Sklodowska-Curie National Research Institute of Oncology, Biobank, Warsaw, Poland
| | | |
Collapse
|
4
|
Ufimtseva EG, Gileva MS, Kostenko RV, Kozlov VV, Gulyaeva LF. Development of Ex Vivo Analysis for Examining Cell Composition, Immunological Landscape, Tumor and Immune Related Markers in Non-Small-Cell Lung Cancer. Cancers (Basel) 2024; 16:2886. [PMID: 39199657 PMCID: PMC11352364 DOI: 10.3390/cancers16162886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/11/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
NSCLC is a very aggressive solid tumor, with a poor prognosis due to post-surgical recurrence. Analysis of the specific tumor and immune signatures of NSCLC samples is a critical step in prognostic evaluation and management decisions for patients after surgery. Routine histological assays have some limitations. Therefore, new diagnostic tools with the capability to quickly recognize NSCLC subtypes and correctly identify various markers are needed. We developed a technique for ex vivo isolation of cancer and immune cells from surgical tumor and lung tissue samples of patients with NSCLC (adenocarcinomas and squamous cell carcinomas) and their examination on ex vivo cell preparations and, parallelly, on histological sections after Romanovsky-Giemsa and immunofluorescent/immunochemical staining for cancer-specific and immune-related markers. As a result, PD-L1 expression was detected for some patients only by ex vivo analysis. Immune cell profiling in the tumor microenvironment revealed significant differences in the immunological landscapes between the patients' tumors, with smokers' macrophages with simultaneous expression of pro- and anti-inflammatory cytokines, neutrophils, and eosinophils being the dominant populations. The proposed ex vivo analysis may be used as an additional diagnostic tool for quick examination of cancer and immune cells in whole tumor samples and to avoid false negatives in histological assays.
Collapse
Affiliation(s)
- Elena G. Ufimtseva
- Federal Research Center of Fundamental and Translational Medicine, 2 Timakova Street, 630060 Novosibirsk, Russia;
| | - Margarita S. Gileva
- V. Zelman Institute for the Medicine and Psychology, Novosibirsk State University, 1 Pirogova Street, 630090 Novosibirsk, Russia;
| | - Ruslan V. Kostenko
- Novosibirsk Regional Clinical Oncology Dispensary, 2 Plakhotny Street, 630108 Novosibirsk, Russia; (R.V.K.); (V.V.K.)
| | - Vadim V. Kozlov
- Novosibirsk Regional Clinical Oncology Dispensary, 2 Plakhotny Street, 630108 Novosibirsk, Russia; (R.V.K.); (V.V.K.)
- Faculty of General Medicine, Novosibirsk State Medical University, 52 Krasny Prospect, 630091 Novosibirsk, Russia
| | - Lyudmila F. Gulyaeva
- Federal Research Center of Fundamental and Translational Medicine, 2 Timakova Street, 630060 Novosibirsk, Russia;
- V. Zelman Institute for the Medicine and Psychology, Novosibirsk State University, 1 Pirogova Street, 630090 Novosibirsk, Russia;
| |
Collapse
|
5
|
Ubiali A, Cesar Conti L, Dall’Ara P, De Maria R, Aresu L, Moretti P, Sini F, Riondato F, Stefanello D, Comazzi S, Martini V. Exploring the dynamics of Programmed Death-Ligand 1 in canine lymphoma: unraveling mRNA amount, surface membrane expression and plasmatic levels. Front Vet Sci 2024; 11:1412227. [PMID: 39132435 PMCID: PMC11310028 DOI: 10.3389/fvets.2024.1412227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 07/15/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Programmed Death-Ligand 1 is a well-known immune checkpoint molecule. Recent studies evaluated its expression in different canine cancer types through different laboratory techniques. The present study aims to evaluate the surface membrane protein expression (mPD-L1) by means of flow cytometry (FC) in different canine lymphoma immunophenotypes. Furthermore, in a subset of cases, mRNA and plasmatic soluble protein (sPD-L1) have been assessed in the same patient, and correlations among results from the three analyses investigated. Methods Samples obtained for diagnostic purpose from untreated dogs with a confirmed lymphoma immunophenotype were included: surface protein was assessed via FC and quantified with median fluorescence index ratio (MFI ratio), gene expression was evaluated by real time quantitative polymerase chain reaction (RT-qPCR) and plasmatic concentration of soluble protein (sPD-L1) measured with ELISA. Statistical analyses were performed to investigate any difference among FC immunophenotypes, updated Kiel cytological classes, and in the presence of blood infiltration. Results Considering FC, most B-cell lymphomas (BCL) were positive, with higher MFI ratios than other subtypes (81%, median MFI ratio among positive samples = 1.50, IQR 1.21-2.03, range 1.01-3.47). Aggressive T-cell lymphomas had a lower percentage of positive samples (56%) and showed low expression (median MFI ratio in positive samples = 1.14, IQR 1.07-1.32, range 1.02-2.19), while T-zone lymphomas (TZL) were frequently positive (80%) but with low expression (median MFI ratio in positive samples = 1.19, IQR 1.03-1.46, range 1.02-6.03). Cellular transcript and sPD-L1 were detected in all samples, without differences among immunophenotypes. No correlation between results from different techniques was detected, but sPD-L1 resulted significantly increased in FC-negative lymphomas (p = 0.023). Discussion PD-L1 molecule is involved in canine lymphoma pathogenesis, with differences among immunophenotypes detected by FC. Specifically, BCL have the highest expression and aggressive T-cell lymphomas the lowest, whereas TZL need further investigations.
Collapse
Affiliation(s)
- Alessandra Ubiali
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Luiza Cesar Conti
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Paola Dall’Ara
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Raffaella De Maria
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Luca Aresu
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Pierangelo Moretti
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Federica Sini
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Fulvio Riondato
- Department of Veterinary Sciences, University of Turin, Grugliasco, TO, Italy
| | - Damiano Stefanello
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Stefano Comazzi
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| | - Valeria Martini
- Department of Veterinary Medicine and Animal Sciences, University of Milan, Lodi, Italy
| |
Collapse
|
6
|
Giuliano A, Pimentel PAB, Horta RS. Checkpoint Inhibitors in Dogs: Are We There Yet? Cancers (Basel) 2024; 16:2003. [PMID: 38893123 PMCID: PMC11171034 DOI: 10.3390/cancers16112003] [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: 04/11/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICI) have revolutionised cancer treatment in people. Immune checkpoints are important regulators of the body's reaction to immunological stimuli. The most studied immune checkpoint molecules are programmed death (PD-1) with its ligand (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) with its ligands CD80 (B7-1) and CD86 (B7-2). Certain tumours can evade immunosurveillance by activating these immunological checkpoint targets. These proteins are often upregulated in cancer cells and tumour-infiltrating lymphocytes, allowing cancer cells to evade immune surveillance and promote tumour growth. By blocking inhibitory checkpoints, ICI can help restore the immune system to effectively fight cancer. Several studies have investigated the expression of these and other immune checkpoints in human cancers and have shown their potential as therapeutic targets. In recent years, there has been growing interest in studying the expression of immune checkpoints in dogs with cancer, and a few small clinical trials with ICI have already been performed on these species. Emerging studies in veterinary oncology are centred around developing and validating canine-targeted antibodies. Among ICIs, anti-PD-1 and anti-PD-L1 treatments stand out as the most promising, mirroring the success in human medicine over the past decade. Nevertheless, the efficacy of caninized antibodies remains suboptimal, especially for canine oral melanoma. To enhance the utilisation of ICIs, the identification of predictive biomarkers for treatment response and the thorough screening of individual tumours are crucial. Such endeavours hold promise for advancing personalised medicine within veterinary practice, thereby improving treatment outcomes. This article aims to review the current research literature about the expression of immune checkpoints in canine cancer and the current results of ICI treatment in dogs.
Collapse
Affiliation(s)
- Antonio Giuliano
- Department of Veterinary Clinical Science, Jockey Club College of Veterinary Medicine, City University of Hong Kong, Hong Kong, China
- Veterinary Medical Centre, City University of Hong Kong, Hong Kong, China
| | - Pedro A. B. Pimentel
- Department of Veterinary Medicine and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| | - Rodrigo S. Horta
- Department of Veterinary Medicine and Surgery, Veterinary School, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte 31270-901, MG, Brazil;
| |
Collapse
|
7
|
Muscatello LV, Gobbo F, Avallone G, Innao M, Benazzi C, D'Annunzio G, Romaniello D, Orioles M, Lauriola M, Sarli G. PDL1 immunohistochemistry in canine neoplasms: Validation of commercial antibodies, standardization of evaluation, and scoring systems. Vet Pathol 2024; 61:393-401. [PMID: 37920996 DOI: 10.1177/03009858231209410] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Immuno-oncology research has brought to light the paradoxical role of immune cells in the induction and elimination of cancer. Programmed cell death protein 1 (PD1), expressed by tumor-infiltrating lymphocytes, and programmed cell death ligand 1 (PDL1), expressed by tumor cells, are immune checkpoint proteins that regulate the antitumor adaptive immune response. This study aimed to validate commercially available PDL1 antibodies in canine tissue and then, applying standardized methods and scoring systems used in human pathology, evaluate PDL1 immunopositivity in different types of canine tumors. To demonstrate cross-reactivity, a monoclonal antibody (22C3) and polyclonal antibody (cod. A1645) were tested by western blot. Cross-reactivity in canine tissue cell extracts was observed for both antibodies; however, the polyclonal antibody (cod. A1645) demonstrated higher signal specificity. Canine tumor histotypes were selected based on the human counterparts known to express PDL1. Immunohistochemistry was performed on 168 tumors with the polyclonal anti-PDL1 antibody. Only membranous labeling was considered positive. PDL1 labeling was detected both in neoplastic and infiltrating immune cells. The following tumors were immunopositive: melanomas (17 of 17; 100%), renal cell carcinomas (4 of 17; 24%), squamous cell carcinomas (3 of 17; 18%), lymphomas (2 of 14; 14%), urothelial carcinomas (2 of 18; 11%), pulmonary carcinomas (2 of 20; 10%), and mammary carcinomas (1 of 31; 3%). Gastric (0 of 10; 0%) and intestinal carcinomas (0 of 24; 0%) were negative. The findings of this study suggest that PDL1 is expressed in some canine tumors, with high prevalence in melanomas.
Collapse
Affiliation(s)
| | | | | | | | | | - Giulia D'Annunzio
- University of Bologna, Bologna, Italy
- Experimental Zooprophylactic Institute of Lombardia and Emilia-Romagna, Brescia, Italy
| | | | | | | | | |
Collapse
|
8
|
Ahn S, Kwak Y, Kwon GY, Kim KM, Kim M, Kim H, Park YS, Oh HJ, Lee K, Lee SH, Lee HS. Interpretation of PD-L1 expression in gastric cancer: summary of a consensus meeting of Korean gastrointestinal pathologists. J Pathol Transl Med 2024; 58:103-116. [PMID: 38653580 PMCID: PMC11106610 DOI: 10.4132/jptm.2024.03.15] [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: 02/22/2024] [Revised: 03/14/2024] [Accepted: 03/14/2024] [Indexed: 04/25/2024] Open
Abstract
Nivolumab plus chemotherapy in the first-line setting has demonstrated clinical efficacy in patients with human epidermal growth factor receptor 2-negative advanced or metastatic gastric cancer, and is currently indicated as a standard treatment. Programmed death-ligand 1 (PD-L1) expression is an important biomarker for predicting response to anti-programmed death 1/PD-L1 agents in several solid tumors, including gastric cancer. In the CheckMate-649 trial, significant clinical improvements were observed in patients with PD-L1 combined positive score (CPS) ≥ 5, determined using the 28-8 pharmDx assay. Accordingly, an accurate interpretation of PD-L1 CPS, especially at a cutoff of 5, is important. The CPS method evaluates both immune and tumor cells and provides a comprehensive assessment of PD-L1 expression in the tumor microenvironment of gastric cancer. However, CPS evaluation has several limitations, one of which is poor interobserver concordance among pathologists. Despite these limitations, clinical indications relying on PD-L1 CPS are increasing. In response, Korean gastrointestinal pathologists held a consensus meeting for the interpretation of PD-L1 CPS in gastric cancer. Eleven pathologists reviewed 20 PD-L1 slides with a CPS cutoff close to 5, stained with the 28-8 pharmDx assay, and determined the consensus scores. The issues observed in discrepant cases were discussed. In this review, we present cases of gastric cancer with consensus PD-L1 CPS. In addition, we briefly touch upon current practices and clinical issues associated with assays used for the assessment of PD-L1 expression in gastric cancer.
Collapse
Affiliation(s)
- Soomin Ahn
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yoonjin Kwak
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| | - Gui Young Kwon
- Seoul Clinical Laboratories, Department of Pathology, Yongin, Korea
| | - Kyoung-Mee Kim
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Moonsik Kim
- Department of Pathology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu, Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Young Soo Park
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Hyeon Jeong Oh
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyoungyul Lee
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | - Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Seung Lee
- Department of Pathology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
9
|
Maekawa N, Konnai S, Hosoya K, Kim S, Kinoshita R, Deguchi T, Owaki R, Tachibana Y, Yokokawa M, Takeuchi H, Kagawa Y, Takagi S, Ohta H, Kato Y, Yamamoto S, Yamamoto K, Suzuki Y, Okagawa T, Murata S, Ohashi K. Safety and clinical efficacy of an anti-PD-L1 antibody (c4G12) in dogs with advanced malignant tumours. PLoS One 2023; 18:e0291727. [PMID: 37792729 PMCID: PMC10550157 DOI: 10.1371/journal.pone.0291727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/02/2023] [Indexed: 10/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been developed for canine tumour treatment, and pilot clinical studies have demonstrated their antitumour efficacy in dogs with oral malignant melanoma (OMM). Although ICIs have been approved for various human malignancies, their clinical benefits in other tumour types remain to be elucidated in dogs. Here, we conducted a clinical study of c4G12, a canine chimeric anti-PD-L1 antibody, to assess its safety and efficacy in dogs with various advanced malignant tumours (n = 12) at the Veterinary Teaching Hospital of Hokkaido University from 2018 to 2023. Dogs with digit or foot pad malignant melanoma (n = 4), osteosarcoma (n = 2), hemangiosarcoma (n = 1), transitional cell carcinoma (n = 1), nasal adenocarcinoma (n = 1), B-cell lymphoma (n = 1), or undifferentiated sarcoma (n = 2) were treated with 2 or 5 mg/kg c4G12 every 2 weeks. Treatment-related adverse events of any grade were observed in eight dogs (66.7%), including elevated aspartate aminotransferase (grade 3) in one dog (8.3%) and thrombocytopenia (grade 4) in another dog (8.3%). Among dogs with target disease at baseline (n = 8), as defined by the response evaluation criteria for solid tumours in dogs (cRECIST), one dog with nasal adenocarcinoma and another with osteosarcoma experienced a partial response (PR), with an objective response rate of 25.0% (2 PR out of 8 dogs; 95% confidence interval: 3.2-65.1%). These results suggest that c4G12 is safe and tolerable and shows antitumor effects in dogs with malignant tumours other than OMM. Further clinical studies are warranted to identify the tumour types that are most likely to benefit from c4G12 treatment.
Collapse
Affiliation(s)
- Naoya Maekawa
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Department of Disease Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
| | - Kenji Hosoya
- Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Sangho Kim
- Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Ryohei Kinoshita
- Cancer Research Unit, One Health Research Center, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Tatsuya Deguchi
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
- Department of Companion Animal Clinical Sciences, Companion Animal Internal Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Ryo Owaki
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Yurika Tachibana
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Madoka Yokokawa
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
| | - Hiroto Takeuchi
- Faculty of Veterinary Medicine, Department of Disease Control, Hokkaido University, Sapporo, Japan
| | | | - Satoshi Takagi
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
- Department of Veterinary Surgery 1, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Hiroshi Ohta
- Faculty of Veterinary Medicine, Veterinary Teaching Hospital, Hokkaido University, Sapporo, Japan
- Department of Companion Animal Clinical Sciences, Companion Animal Internal Medicine, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Satoshi Yamamoto
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Fuso Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Keiichi Yamamoto
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Fuso Pharmaceutical Industries, Ltd., Osaka, Japan
| | - Yasuhiko Suzuki
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
| | - Shiro Murata
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Department of Disease Control, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Faculty of Veterinary Medicine, Department of Advanced Pharmaceutics, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, Department of Disease Control, Hokkaido University, Sapporo, Japan
- Faculty of Veterinary Medicine, International Affairs Office, Hokkaido University, Sapporo, Japan
| |
Collapse
|
10
|
Kuusisalo S, Tikkanen A, Lappi‐Blanco E, Väisänen T, Knuuttila A, Tiainen S, Ahvonen J, Iivanainen S, Koivunen JP. The prognostic and predictive roles of plasma C-reactive protein and PD-L1 in non-small cell lung cancer. Cancer Med 2023; 12:16087-16097. [PMID: 37329173 PMCID: PMC10469721 DOI: 10.1002/cam4.6262] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 05/16/2023] [Accepted: 06/06/2023] [Indexed: 06/18/2023] Open
Abstract
BACKGROUND Anti-PD-(L)1 agents have revolutionized the treatment paradigms of non-small cell lung cancer (NSCLC), while predictive biomarkers are limited. It has been previously shown that systemic inflammation, indicated by elevated C-reactive protein (CRP) level, is associated with a poor prognosis in anti-PD-(L)1 treated. The aim of the study was to analyze the prognostic and predictive value of CRP in addition to traditional prognostic and predictive markers and tumor PD-L1 score. METHODS We identified all NSCLC patients (n = 329) who had undergone PD-L1 tumor proportion score (TPS) analysis at Oulu University Hospital 2015-22. CRP levels, treatment history, immune checkpoint inhibitor (ICI) therapy details, and survival were collected. The patients were categorized based on CRP levels (≤10 vs. >10) and PD-L1 TPS scores (<50 vs. ≥50). RESULTS In the whole cohort (n = 329), CRP level of ≤10 mg/L was associated with improved survival in univariate (HR 0.30, Cl 95% 0.22-0.41) and multivariate analyzes (HR 0.44, CI 95% 0.28-0.68). With ICI treated (n = 70), both CRP of ≤10 and PD-L1 TPS of ≥50 were associated with improved progression-free survival (PFS) in univariate (HR 0.51, CI 95% 0.27-0.96; HR 0.54, CI 95% 0.28-1.02) and multivariate (HR 0.48, CI 95% 0.26-0.90; HR 0.50, CI 95% 0.26-0.95) analyzes. The combination (PD-L1 TPS ≥50 and CRP >10) carried a high negative predictive value with a median PFS of 4.11 months (CI 95% 0.00-9.63), which was similar to patients with low PD-L1 (4.11 months, CI 95% 2.61-5.60). CONCLUSIONS Adding plasma CRP levels to PD-L1 TPS significantly increased the predictive value of sole PD-L1. Furthermore, patients with high CRP beard little benefit from anti-PD-(L)1 therapies independent of PD-L1 score. The study highlights the combined evaluation of plasma CRP and PD-L1 TPS as a negative predictive marker for ICI therapies.
Collapse
Affiliation(s)
- Saara Kuusisalo
- Department of Medical Oncology and Radiotherapy and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Antti Tikkanen
- Department of Medical Oncology and Radiotherapy and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Elisa Lappi‐Blanco
- Department of Pathology, Oulu University Hospital and Department of Pathology, Cancer and Translational Medicine Research UnitUniversity of OuluOuluFinland
| | - Timo Väisänen
- Department of Pathology, Oulu University Hospital and Department of Pathology, Cancer and Translational Medicine Research UnitUniversity of OuluOuluFinland
| | - Aija Knuuttila
- Department of Pulmonary Medicine, Heart and Lung Center and Cancer CenterHelsinki University Hospital and University of HelsinkiHelsinkiFinland
| | - Satu Tiainen
- Cancer CenterKuopio University HospitalKuopioFinland
| | - Jarkko Ahvonen
- Faculty of Medicine and Health Technology, Tampere University and Tays Cancer Center, Department of OncologyTampere University HospitalTampereFinland
| | - Sanna Iivanainen
- Department of Medical Oncology and Radiotherapy and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| | - Jussi P. Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center OuluOulu University Hospital and University of OuluOuluFinland
| |
Collapse
|
11
|
Talavera Guillén NC, Barboza de Nardi A, Noleto de Paiva F, Dias QC, Pinheiro Fantinatti A, Fávaro WJ. Clinical Implications of Immune Checkpoints and the RANK/RANK-L Signaling Pathway in High-Grade Canine Mast Cell Tumors. Animals (Basel) 2023; 13:1888. [PMID: 37370399 DOI: 10.3390/ani13121888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/13/2023] [Accepted: 05/22/2023] [Indexed: 06/29/2023] Open
Abstract
Mast cell tumors (MCTs) are the most common malignant cutaneous tumors in dogs, and they present extremely variable biological behavior. The interaction between RANK, RANK-L, and immune checkpoints is frequently detected in the tumor microenvironment, and, together, they participate in every stage of cancer development. Thus, the aim of this study was to characterize the molecular profiles of PD-L1, CTLA-4, RANK/RANK-L signaling pathway, and IFN-γ in primary tumors and lymph node metastases. Formalin-fixed, paraffin-embedded slides of MCTs and metastatic lymph nodes of ten dogs were submitted to immunohistochemical investigations. The results demonstrated that the tumor microenvironment of the high-grade mast cell tumors showed moderate or intense immunolabeling of all proteins, and the lymph node metastases also showed moderate or intense immunolabeling of checkpoint proteins. In addition, MCTs larger than 3 cm were associated with intensified PD-L1 (p = 0.03) in metastatic lymph nodes and RANK-L (p = 0.049) immunoreactivity in the tumor. Furthermore, dogs with a survival time of less than 6 months showed higher PD-L1 immunoreactivity (p = 0.042). In conclusion, high-grade MCT is associated with an immunosuppressive microenvironment that exhibits elevated RANK/RANK-L signaling and enhanced immune checkpoint immunoreactivity, potentially facilitating intratumorally immune escape. These biomarkers show promise as clinical indicators of disease progression and might response to immunotherapy in dogs with high-grade MCTs, thus emphasizing their importance for guiding treatment decisions and improving outcomes.
Collapse
Affiliation(s)
- Noelia C Talavera Guillén
- Department of Veterinary Clinics and Surgery, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Andrigo Barboza de Nardi
- Department of Veterinary Clinics and Surgery, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Felipe Noleto de Paiva
- Department of Veterinary Clinics and Surgery, São Paulo State University (UNESP), Jaboticabal 14884-900, Brazil
| | - Queila Cristina Dias
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
| | | | - Wagner José Fávaro
- Department of Structural and Functional Biology, University of Campinas (UNICAMP), Campinas 13083-970, Brazil
| |
Collapse
|
12
|
Fitzsimmons TS, Singh N, Walker TDJ, Newton C, Evans DGR, Crosbie EJ, Ryan NAJ. Immune checkpoint inhibitors efficacy across solid cancers and the utility of PD-L1 as a biomarker of response: a systematic review and meta-analysis. Front Med (Lausanne) 2023; 10:1192762. [PMID: 37250628 PMCID: PMC10219231 DOI: 10.3389/fmed.2023.1192762] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/20/2023] [Indexed: 05/31/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICPI) are a tumor agnostic treatment. However, trials of their use have been site specific. Here we summarize the trial data and explore the utility of programmed death-ligand 1 (PD-L1) expression as a biomarker to direct their pan-cancer use. Method A systematic review of literature, following PRISMA guidelines, was performed. Medline, Embase, Cochrane CENTRAL, NHS Health and Technology, and Web of Science were searched from their conception to June 2022 limited to the English language. The search terms and method were devised by a specialist medical librarian. Studies were limited to adults with solid cancers (excluding melanomas) treated with ICPIs. Only phase III randomized control trials (RCT) were included. The primary outcome was overall survival and secondary outcomes were progression free survival, PD-L1 expression, quality of life outcomes and adverse event data. Where present in eligible clinical trials, hazard ratios (HR), risk ratios (RR), standard error (SE) and 95% confidence intervals (CI) were extracted or calculated. Heterogeneity across studies was described with the use of an I2 score (Low: 25, 50%: moderate, 75% low heterogeneity). HR pools inverse variance methods were adopted by Random Effects (RE). Means were standardized across any heterogenous scale limits. Results In total 46,510 participants were included in the meta-analysis. Overall, meta-analysis favored the use of ICPIs with an overall survival (OS) HR of 0.74 (95% CI 0.71 to 0.78). Lung cancers showed the most benefit in OS [HR 0.72 (95% 0.66-0.78)] followed by head and neck cancers [HR 0.75 (95% CI 0.66-0.84)] and gastroesophageal junction cancers [HR 0.75 (95% CI 0.61-0.92)]. ICPIs seem to be efficacious at both primary presentation and recurrence [OS HR 0.73 (95% CI 0.68-0.77)] vs. [OS HR 0.79 (95% CI 0.72 to 0.87)] respectively. Interestingly, subgroup analysis comparing studies in which most cancers demonstrated PD-L1 expression vs. those studies in which a minority of cancer demonstrated PD-L1 expression reported similar effect of ICPI use on OS; oddly the data favored ICPI use in studies with a minority of PD-L1 expression. Specifically, studies with minority PD-L1 expression had an HR 0.73 (95% CI 0.68-0.78) vs. studies with majority PD-L1 expression HR 0.76 (95% CI 0.70-0.84). This was maintained even when studies exploring the same cancer site were directly compared. Subgroup analysis was performed comparing the impact on OS subdivided by the specific ICPI used. Where meta-analysis was performed, Nivolumab led to the greatest impact [HR 0.70 (95% CI 0.64-0.77)] with Avelumab failing to reach significance [HR 0.93 (95% CI 0.80-1.06)]. However, overall heterogenicity was high (I2 = 95%). Finally, the use of ICPIs led to an improved side effect profile when compared with standard chemotherapy [RR 0.85 (95% CI 0.73-0.98)]. Conclusion ICPIs improve survival outcomes in all cancer types. These effects are seen in the primary, recurrent, chemotherapy sensitive, chemotherapy resistant disease. These data support their use as a tumor agnostic therapy. Furthermore, they are well tolerated. However, PD-L1 as a biomarker for the targeting of ICPI use seems problematic. Other biomarkers such as mismatch repair or tumor mutational burden should be explored in randomized trials. In addition, there are still limited trials looking at ICPI use outside of lung cancer.
Collapse
Affiliation(s)
| | - Niharika Singh
- Clinical Medical School, University of Bristol, Bristol, United Kingdom
| | - Thomas D. J. Walker
- Division of Cancer Sciences, St Mary’s Hospital, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Claire Newton
- Department of Obstetrics and Gynaecology, St Michaels Hospital, Bristol, United Kingdom
| | - Dafydd G. R. Evans
- Division of Evolution and Genomic Medicine, St Mary’s Hospital, University of Manchester, Manchester, United Kingdom
| | - Emma J. Crosbie
- Division of Cancer Sciences, St Mary’s Hospital, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Neil A. J. Ryan
- The College of Medicine and Veterinary Medicine, University of Edinburgh, Edinburgh, United Kingdom
- Department of Gynaecology Oncology, Royal Infirmary of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
13
|
Krutzek F, Donat CK, Ullrich M, Zarschler K, Ludik MC, Feldmann A, Loureiro LR, Kopka K, Stadlbauer S. Design and Biological Evaluation of Small-Molecule PET-Tracers for Imaging of Programmed Death Ligand 1. Cancers (Basel) 2023; 15:cancers15092638. [PMID: 37174103 PMCID: PMC10177516 DOI: 10.3390/cancers15092638] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/18/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Noninvasive molecular imaging of the PD-1/PD-L1 immune checkpoint is of high clinical relevance for patient stratification and therapy monitoring in cancer patients. Here we report nine small-molecule PD-L1 radiotracers with solubilizing sulfonic acids and a linker-chelator system, designed by molecular docking experiments and synthesized according to a new, convergent synthetic strategy. Binding affinities were determined both in cellular saturation and real-time binding assay (LigandTracer), revealing dissociation constants in the single digit nanomolar range. Incubation in human serum and liver microsomes proved in vitro stability of these compounds. Small animal PET/CT imaging, in mice bearing PD-L1 overexpressing and PD-L1 negative tumors, showed moderate to low uptake. All compounds were cleared primarily through the hepatobiliary excretion route and showed a long circulation time. The latter was attributed to strong blood albumin binding effects, discovered during our binding experiments. Taken together, these compounds are a promising starting point for further development of a new class of PD-L1 targeting radiotracers.
Collapse
Affiliation(s)
- Fabian Krutzek
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Cornelius K Donat
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Martin Ullrich
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Kristof Zarschler
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Marie-Charlotte Ludik
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Anja Feldmann
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Liliana R Loureiro
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
| | - Klaus Kopka
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
- German Cancer Consortium (DKTK), Partner Site Dresden, Fetscherstraße 74, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT) Dresden, University Hospital Carl Gustav Carus, Fetscherstraße 74, 01307 Dresden, Germany
| | - Sven Stadlbauer
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Bautzner Landstraße 400, 01328 Dresden, Germany
- School of Science, Faculty of Chemistry and Food Chemistry, Technische Universität Dresden, Mommsenstraße 4, 01069 Dresden, Germany
| |
Collapse
|
14
|
Wang T, Sun S, Zeng X, Li J. ICI-based therapies: A new strategy for oral potentially malignant disorders. Oral Oncol 2023; 140:106388. [PMID: 37054586 DOI: 10.1016/j.oraloncology.2023.106388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 03/27/2023] [Indexed: 04/15/2023]
Abstract
Oral potentially malignant disorders (OPMDs) are linked with an escalated risk of developing cancers, particularly oral squamous cell carcinoma (OSCC). Since prevailing therapies cannot effectively forestall the exacerbation and recurrence of OPMDs, halting their malignant progression is paramount. The immune checkpoint serves as a cardinal regulator of the immune response and the primary cause of adaptive immunological resistance. Although the exact mechanism remains elusive, elevated expression of multiple immune checkpoints in OPMDs and OSCC relative to healthy oral mucosa has been ascertained. This review delves into the immunosuppressive microenvironment of OPMDs, the expression of diverse immune checkpoints such as programmed death receptor-1 (PD-1) and programmed death receptor-1 ligand (PD-L1) in OPMDs, and the potential application of corresponding inhibitors. In addition, synergistic strategies incorporating combined immune checkpoint inhibitors, such as cGAS-STING, costimulatory molecules, cancer vaccines, and hydrogels, are discussed to gain a more comprehensive understanding of the role and application of immune checkpoint inhibitors (ICIs) in oral carcinogenesis.
Collapse
Affiliation(s)
- Tianqing Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Silu Sun
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Zeng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| | - Jing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
15
|
Lester DK, Burton C, Gardner A, Innamarato P, Kodumudi K, Liu Q, Adhikari E, Ming Q, Williamson DB, Frederick DT, Sharova T, White MG, Markowitz J, Cao B, Nguyen J, Johnson J, Beatty M, Mockabee-Macias A, Mercurio M, Watson G, Chen PL, McCarthy S, MoranSegura C, Messina J, Thomas KL, Darville L, Izumi V, Koomen JM, Pilon-Thomas SA, Ruffell B, Luca VC, Haltiwanger RS, Wang X, Wargo JA, Boland GM, Lau EK. Fucosylation of HLA-DRB1 regulates CD4 + T cell-mediated anti-melanoma immunity and enhances immunotherapy efficacy. NATURE CANCER 2023; 4:222-239. [PMID: 36690875 PMCID: PMC9970875 DOI: 10.1038/s43018-022-00506-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 12/14/2022] [Indexed: 01/24/2023]
Abstract
Immunotherapy efficacy is limited in melanoma, and combinations of immunotherapies with other modalities have yielded limited improvements but also adverse events requiring cessation of treatment. In addition to ineffective patient stratification, efficacy is impaired by paucity of intratumoral immune cells (itICs); thus, effective strategies to safely increase itICs are needed. We report that dietary administration of L-fucose induces fucosylation and cell surface enrichment of the major histocompatibility complex (MHC)-II protein HLA-DRB1 in melanoma cells, triggering CD4+ T cell-mediated increases in itICs and anti-tumor immunity, enhancing immune checkpoint blockade responses. Melanoma fucosylation and fucosylated HLA-DRB1 associate with intratumoral T cell abundance and anti-programmed cell death protein 1 (PD1) responder status in patient melanoma specimens, suggesting the potential use of melanoma fucosylation as a strategy for stratifying patients for immunotherapies. Our findings demonstrate that fucosylation is a key mediator of anti-tumor immunity and, importantly, suggest that L-fucose is a powerful agent for safely increasing itICs and immunotherapy efficacy in melanoma.
Collapse
Affiliation(s)
- Daniel K Lester
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chase Burton
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Alycia Gardner
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Patrick Innamarato
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Krithika Kodumudi
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Qian Liu
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Emma Adhikari
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Cancer Biology Ph.D. Program, University of South Florida, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Qianqian Ming
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Daniel B Williamson
- Complex Carbohydrate Research Center, the University of Georgia, Athens, GA, USA
| | | | - Tatyana Sharova
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Michael G White
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Markowitz
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Biwei Cao
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jonathan Nguyen
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joseph Johnson
- Department of Analytic Microscopy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Matthew Beatty
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Andrea Mockabee-Macias
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Matthew Mercurio
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Gregory Watson
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Pei-Ling Chen
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Susan McCarthy
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Carlos MoranSegura
- Advanced Analytical and Digital Laboratory, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jane Messina
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kerry L Thomas
- Department of Diagnostic Imaging, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lancia Darville
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Victoria Izumi
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - John M Koomen
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Shari A Pilon-Thomas
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Brian Ruffell
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Immunology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Vincent C Luca
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Robert S Haltiwanger
- Complex Carbohydrate Research Center, the University of Georgia, Athens, GA, USA
| | - Xuefeng Wang
- Department of Biostatistics and Bioinformatics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jennifer A Wargo
- Department of Surgical Oncology, MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, MD Anderson Cancer Center, Houston, TX, USA
| | - Genevieve M Boland
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Harvard and Massachusetts Institute of Technology, Massachusetts General Hospital, Boston, MA, USA
| | - Eric K Lau
- Department of Tumor Biology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
- Molecular Medicine Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
16
|
Maekawa N, Konnai S, Asano Y, Otsuka T, Aoki E, Takeuchi H, Kato Y, Kaneko MK, Yamada S, Kagawa Y, Nishimura M, Takagi S, Deguchi T, Ohta H, Nakagawa T, Suzuki Y, Okagawa T, Murata S, Ohashi K. Molecular characterization of feline immune checkpoint molecules and establishment of PD-L1 immunohistochemistry for feline tumors. PLoS One 2023; 18:e0281143. [PMID: 36701405 PMCID: PMC9879432 DOI: 10.1371/journal.pone.0281143] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/11/2023] [Indexed: 01/27/2023] Open
Abstract
Spontaneous tumors are a major cause of death in cats. Treatment of human tumors has progressed dramatically in the past decade, partly due to the success of immunotherapies using immune checkpoint inhibitors, such as anti-programmed death 1 (PD-1) and anti-PD-ligand 1 (PD-L1) antibodies. However, little is known about the PD-1 pathway and its association with tumor disease in cats. This study investigated the applicability of anti-PD-1/PD-L1 therapy in feline tumors. We first determined the complete coding sequence of feline PD-L1 and PD-L2, and found that the deduced amino acid sequences of feline PD-L1/PD-L2 share high sequence identities (66-83%) with orthologs in other mammalian species. We prepared recombinant feline PD-1, PD-L1, and PD-L2 proteins and confirmed receptor-ligand binding between PD-1 and PD-L1/PD-L2 using flow cytometry. Next, we established an anti-feline PD-L1 monoclonal antibody (clone CL1Mab-7) to analyze the expression of PD-L1. Flow cytometry using CL1Mab-7 revealed the cell surface expression of PD-L1 in a feline macrophage (Fcwf-4) and five mammary adenocarcinoma cell lines (FKNp, FMCm, FYMp, FONp, and FONm), and showed that PD-L1 expression was upregulated by interferon-γ stimulation. Finally, immunohistochemistry using CL1Mab-7 also showed PD-L1 expression in feline squamous cell carcinoma (5/5, 100%), mammary adenocarcinoma (4/5, 80%), fibrosarcoma (5/5, 100%), and renal cell carcinoma (2/2, 100%) tissues. Our results strongly encourage further investigations of the PD-1/PD-L1 pathway as a potential therapeutic target for feline tumors.
Collapse
Affiliation(s)
- Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- * E-mail:
| | - Yumie Asano
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Takumi Otsuka
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Eri Aoki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroto Takeuchi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K. Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shinji Yamada
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | - Satoshi Takagi
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Veterinary Surgery 1, School of Veterinary Medicine, Azabu University, Sagamihara, Japan
| | - Tatsuya Deguchi
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Companion Animal Internal Medicine, Department of Companion Animal Clinical Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Hiroshi Ohta
- Veterinary Teaching Hospital, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Companion Animal Internal Medicine, Department of Companion Animal Clinical Sciences, School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Bunkyo, Japan
| | - Yasuhiko Suzuki
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Global Station for Zoonosis Control, Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shiro Murata
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- International Affairs Office, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
17
|
Ameli F, Shajareh E, Mokhtari M, Kosari F. Expression of PD1 and PDL1 as immune-checkpoint inhibitors in mantle cell lymphoma. BMC Cancer 2022; 22:848. [PMID: 35922773 PMCID: PMC9351258 DOI: 10.1186/s12885-022-09803-x] [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: 04/23/2022] [Accepted: 06/22/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Mantle cell lymphoma (MCL) has remained incurable in most patients. The expression of PD-L1 as a prognostic and predictive marker has not been fully evaluated in MCL. The current study aimed to determine PD-1/PD-L1 expression in MCL specimens and its significance as an immune check point inhibitor. METHODS This retrospective study was conducted on the formalin-fixed paraffin-embedded blocks of 79 confirmed MCL patients based on immunohistochemistry (IHC). The IHC method was used to stain patient samples for PD1 and PDL1. Positive PD-1/PD-L1 expression was defined as moderate to strong or memberanous or memberanous/cytoplasmic staining in at least 5% of tumor and/or 20% of associated immune cells. Tumor aggressiveness was determined based on Ki67 and variant. RESULTS The mean age of the patients was 60.08 ± 10.78 years old. Majority of the patients were male. The prevalence of aggressive tumor was 25%. Positive PD1 and PDL1 expression were identified in 12 (15.0%) and 3 (3.8%) of tumor cells, respectively. PD1 and PDL1 were positive in zero (0%) and 7 (8.9%) of background cells, respectively. There was no significant difference in terms of study parameters between positive and negative groups for both PD1 and PDL1 proteins. PD1 tumor cell percentage was negatively correlated with age (r = -0.254, p = 0.046). CONCLUSION Our results suggest that neither PD-1 nor its ligands represent relevant targets for MCL treatment. Age may impact the efficiency of immune checkpoint inhibitors and could be related to the increased incidence of MCL with age.
Collapse
Affiliation(s)
- Fereshteh Ameli
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Science, Tehran, Iran
| | - Elham Shajareh
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Science, Tehran, Iran
| | - Maral Mokhtari
- Department of pathology, Shiraz University of Medical Science, Shiraz, Iran
| | - Farid Kosari
- Department of Pathology, Shariati Hospital, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
18
|
Bryant AK, Sankar K, Strohbehn GW, Zhao L, Daniel V, Elliott D, Ramnath N, Green MD. Prognostic and Predictive Role of PD-L1 Expression in Stage III Non-small Cell Lung Cancer Treated With Definitive Chemoradiation and Adjuvant Durvalumab. Int J Radiat Oncol Biol Phys 2022; 113:752-758. [PMID: 35450753 PMCID: PMC9246927 DOI: 10.1016/j.ijrobp.2022.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/07/2022] [Accepted: 03/13/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE It is unclear whether programmed death ligand 1 (PD-L1) expression is prognostic or predictive of immunotherapy benefit among patients with stage III non-small cell lung cancer (NSCLC) treated with definitive chemoradiation and adjuvant durvalumab. METHODS AND MATERIALS We determined pretreatment tumor PD-L1 expression for 312 patients with stage III NSCLC treated with definitive chemoradiation and at least 1 dose of adjuvant durvalumab between November 2017 and April 2021 across the national Veterans Health Administration. Progression-free survival (PFS) and overall survival (OS) in PD-L1 expression subgroups (<1%, 1%-49%, and 50%-100%) were compared with 994 patients with stage III NSCLC treated without adjuvant durvalumab from 2015 to 2016. RESULTS PD-L1 expression was <1%, 1% to 49%, and 50% to 100% in 109 (34.9%), 96 (30.7%), and 107 (34.3%) patients, respectively. Increasing PD-L1 expression was associated with longer PFS (adjusted hazard ratio [aHR], 0.84 per 25% absolute increase in expression; 95% confidence interval [CI], 0.75-0.94; P = .003) and OS (aHR, 0.86 per 25% absolute increase in expression; 95% CI, 0.74-0.99; P = .036). Compared with the no-durvalumab group, PFS was longer for PD-L1 50% to 100% (aHR, 0.44; 95% CI, 0.32-0.60; P < .001) and PD-L1 1% to 49% (aHR, 0.64; 95% CI, 0.47-0.86; P = .003) but not PD-L1 <1% (aHR, 0.84; 95% CI, 0.64-1.10; P = .19). Similar results were found for OS, with no significant difference between the no-durvalumab group and PD-L1 <1% (aHR, 0.81; 95% CI, 0.58-1.13; P = .22). CONCLUSIONS Increasing tumor PD-L1 expression is prognostic for PFS and OS among patients with stage III NSCLC treated with adjuvant durvalumab, and patients with PD-L1 expression <1% may have limited benefit from adjuvant durvalumab.
Collapse
Affiliation(s)
- Alex K Bryant
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Kamya Sankar
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Section of Hematology Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Garth W Strohbehn
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Section of Hematology Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan; Veterans Affairs Center for Clinical Management Research, Veterans Affairs Ann Arbor, Ann Arbor, Michigan
| | - Lili Zhao
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Department of Biostatistics, University of Michigan, Ann Arbor, Michigan
| | - Victoria Daniel
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan
| | - David Elliott
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan
| | - Nithya Ramnath
- Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Division of Hematology Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; Section of Hematology Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan.
| | - Michael D Green
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan; Rogel Cancer Center, University of Michigan, Ann Arbor, Michigan; Department of Radiation Oncology, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, Michigan.
| |
Collapse
|
19
|
Mackenzie NJ, Nicholls C, Templeton AR, Perera MPJ, Jeffery PL, Zimmermann K, Kulasinghe A, Kenna TJ, Vela I, Williams ED, Thomas PB. Modelling the tumor immune microenvironment for precision immunotherapy. CLINICAL & TRANSLATIONAL IMMUNOLOGY 2022; 11:e1400. [PMID: 35782339 PMCID: PMC9234475 DOI: 10.1002/cti2.1400] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/14/2022] [Accepted: 06/07/2022] [Indexed: 12/15/2022]
Affiliation(s)
- Nathan J Mackenzie
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
| | - Clarissa Nicholls
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
| | - Abby R Templeton
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
| | - Mahasha PJ Perera
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
- Department of Urology Princess Alexandra Hospital Woolloongabba QLD Australia
| | - Penny L Jeffery
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| | - Kate Zimmermann
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Microbiome Research School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
| | - Arutha Kulasinghe
- University of Queensland Diamantina Institute The University of Queensland Brisbane QLD Australia
| | - Tony J Kenna
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Centre for Immunology and Infection Control School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
- Centre for Microbiome Research School of Biomedical Sciences Queensland University of Technology (QUT) Brisbane QLD Australia
| | - Ian Vela
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
- Department of Urology Princess Alexandra Hospital Woolloongabba QLD Australia
| | - Elizabeth D Williams
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| | - Patrick B Thomas
- School of Biomedical Sciences at Translational Research Institute (TRI) Queensland University of Technology (QUT) Brisbane QLD Australia
- Queensland Bladder Cancer Initiative (QBCI) Brisbane QLD Australia
- Centre for Personalised Analysis of Cancers (CPAC) Brisbane QLD Australia
- Australian Prostate Cancer Research Centre – Queensland (APCRC‐Q) Brisbane QLD Australia
| |
Collapse
|
20
|
Liu Q, Wang X, Yang Y, Wang C, Zou J, Lin J, Qiu L. Immuno-PET imaging of PD-L1 expression in patient-derived lung cancer xenografts with [ 68Ga]Ga-NOTA-Nb109. Quant Imaging Med Surg 2022; 12:3300-3313. [PMID: 35655844 PMCID: PMC9131318 DOI: 10.21037/qims-21-991] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Accepted: 01/17/2022] [Indexed: 09/17/2023]
Abstract
Background Accurate evaluation of programmed death-ligand 1 (PD-L1) expression levels in cancer patients may be useful in the identification of potential candidates for anti-programmed death-1/PD-L1 (anti-PD-1/PD-L1) immune checkpoint therapy to improve the response rate of immune checkpoint blockade therapy. This study evaluated the feasibility of the nanobody-based positron emission tomography (PET) tracer [68Ga]Ga-NOTA-Nb109 for immuno-PET imaging of PD-L1 in lung cancer patient-derived xenograft (PDX). Methods We constructed 2 PDXs of lung adenocarcinoma (ADC) and lung squamous cell carcinoma (SCC) and used them for immuno-PET imaging. A 2-hour dynamic PET scanning was performed on the samples and the in vivo biodistribution and metabolism of [68Ga]Ga-NOTA-Nb109 were investigated using region of interest (ROI) analysis. The ex vivo biodistribution of [68Ga]Ga-NOTA-Nb109 in the 2 PDXs was investigated by static PET scanning. In addition, tumor PD-L1 expression in the 2 PDXs was evaluated by autoradiography, western blot, and immunohistochemical (IHC) analysis. Results Noninvasive PET imaging showed that [68Ga]Ga-NOTA-Nb109 can accurately and sensitively assess the PD-L1 expression in non-small cell lung cancer (NSCLC) PDX models. The maximum [68Ga]Ga-NOTA-Nb109 uptake by the ADC PDX LU6424 and the SCC PDX LU6437 were 3.13%±0.35% and 2.60%±0.32% injected dose per milliliter of tissue volume (ID/mL), respectively, at 20 min post injection. In vivo and ex vivo biodistribution analysis showed that [68Ga]Ga-NOTA-Nb109 was rapidly cleared through renal excretion and an enhanced signal-to-noise ratio (SNR) was achieved. Ex vivo PD-L1 expression analysis showed good agreement with in vivo PET imaging results. Conclusions This study demonstrated that [68Ga]Ga-NOTA-Nb109 could be applied with PET imaging to noninvasively and accurately monitor PD-L1 expression in vivo for screening patients who may be responsive to immunotherapy and to guide the development of appropriate treatment strategies for such patients.
Collapse
Affiliation(s)
- Qingzhu Liu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Xiaodan Wang
- Wuxi Second Hospital Affiliated to Nanjing Medical University, Wuxi, China
| | - Yanling Yang
- Suzhou Smart Nuclide Biopharmaceutical Co. Ltd., Suzhou Industrial Park, Suzhou, China
| | - Chao Wang
- Suzhou Smart Nuclide Biopharmaceutical Co. Ltd., Suzhou Industrial Park, Suzhou, China
| | - Jian Zou
- Center of Clinical Research, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi, China
| | - Jianguo Lin
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, China
| | - Ling Qiu
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
- Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, China
| |
Collapse
|
21
|
Rha SY, Ku GY, Kim HS, Chung HC, Amlashi FG, Maru DM, Fein CA, Tang LH, Zhou W, Wu T, Peter SA, Kelsen DP, Ajani JA. PD-L1 expression and overall survival in Asian and western patients with gastric cancer. Future Oncol 2022; 18:2623-2634. [PMID: 35616013 DOI: 10.2217/fon-2022-0103] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: Data are limited on PD-L1 expression and its association with overall survival (OS) in gastric cancer (GC) patients receiving routine care in different regions. Materials & methods: In a retrospective study, PD-L1 expression was assayed using the 22C3 pharmDx on GC tumor samples collected between 2003 and 2017 at South Korean and US cancer centers. PD-L1 positivity was defined as combined positive score (CPS) ≥1. The relationship between PD-L1 and OS was analyzed. Results: Of 574 GC tumor samples, 67.4% were CPS ≥1 (68.7% in Korean and 65.7% in US patients). PD-L1 expression was not associated with OS (adjusted hazard ratio: 0.94; 95% CI: 0.75-1.17). Conclusion: PD-L1 prevalence and its association with OS was similar between South Korean and US GC patients.
Collapse
Affiliation(s)
- Sun Young Rha
- Yonsei Cancer Center, Yonsei University Health System, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Geoffrey Y Ku
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Hyo Song Kim
- Yonsei Cancer Center, Yonsei University Health System, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Cheol Chung
- Yonsei Cancer Center, Yonsei University Health System, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Songdang Institute for Cancer Research, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | | | | | - Carly A Fein
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Laura H Tang
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Wei Zhou
- Merck & Co., Inc., 126 E. Lincoln Ave. Rahway, 07065, USA
| | - Ting Wu
- Merck & Co., Inc., 126 E. Lincoln Ave. Rahway, 07065, USA
| | - Senaka A Peter
- Merck & Co., Inc., 126 E. Lincoln Ave. Rahway, 07065, USA
| | - David P Kelsen
- Memorial Sloan Kettering Cancer Center, 1275 York Ave, New York, NY 10065, USA
| | - Jaffer A Ajani
- The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
22
|
Wang H, Li ZW, Ou Q, Wu X, Nagasaka M, Shao Y, Ou SHI, Yang Y. NTRK fusion positive colorectal cancer is a unique subset of CRC with high TMB and microsatellite instability. Cancer Med 2022; 11:2541-2549. [PMID: 35506567 PMCID: PMC9249987 DOI: 10.1002/cam4.4561] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 01/20/2023] Open
Abstract
TRK fusions are rare but targetable mutations which occur across a wide variety of cancer types. We report the prevalence of approximately 0.7% for NTRK‐positive colorectal cancer (CRC) by genetically profiling 2519 colonic and rectal tumors. The aberrations of APC and TP53 frequently co‐occurred with NTRK gene fusions, whereas RAS/BRAF oncogenic alterations and NTRK fusions were almost always mutually exclusive. NTRK‐driven colorectal cancer patients demonstrated increased TMB (median = 53 mut/MB, 95% CI: 36.8–68.0 mut/MB), high microsatellite instability, and an enrichment for POLE/POLD1 mutations when compared to molecularly unstratified colorectal cancer population. These data shed light on possible future approach of multimodality treatment regimen including TRK‐targeted therapy and immune checkpoint inhibitor therapy in NTRK‐positive CRCs.
Collapse
Affiliation(s)
- Hui Wang
- Department of Medical Oncology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhi-Wei Li
- Department of Internal Medicine, Harbin Medical University Cancer Hospital, Harbin, China
| | - Qiuxiang Ou
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Xue Wu
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China
| | - Misako Nagasaka
- Karmanos Cancer Institute, Wayne State University, Detroit, Michigan, USA
| | - Yang Shao
- Geneseeq Research Institute, Nanjing Geneseeq Technology Inc., Nanjing, Jiangsu, China.,School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sai-Hong Ignatius Ou
- Chao Family Comprehensive Cancer Center, University of California Irvine School of Medicine, Orange, California, USA
| | - Yu Yang
- Department of Oncology, the Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| |
Collapse
|
23
|
Darmon-Novello M, Adam J, Lamant L, Battistella M, Ortonne N, Balme B, de la Fouchardière A, Chaltiel L, Gerard E, Franchet C, Vergier B. Harmonization of PD-L1 immunohistochemistry and mRNA expression scoring in metastatic melanoma: a multicenter analysis. Histopathology 2022; 80:1091-1101. [PMID: 35322452 DOI: 10.1111/his.14651] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/21/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022]
Abstract
AIMS This multicenter study sought to harmonize programmed death ligand 1 (PD-L1) immunohistochemistry (IHC) data and melanoma scoring. To provide a reference for PD-L1 expression independent of the IHC protocol, PD-L1 mRNA expression was determined then compared to IHC. METHODS Standardized PD-L1 assays (22C3, 28-8, SP142, and SP263) and laboratory-developed tests (QR1 and 22C3) were evaluated on three IHC platforms using a training set of 7 cases. mRNA expression was determined via RNAscope (CD274/PD-L1 probe) and analyzed by image analysis. PD-L1 IHC findings were scored by seven blinded pathologists using the tumor proportion score (TPS), combined positive score (CPS), and MELscore. This method was validated by three blinded pathologists on 40 metastatic melanomas. RESULTS Concordances among various antibody/platforms were high across antibodies (ICC > 0.80 for CPS), except for SP142. Two levels of immunostaining intensities were observed: high (QR1 and SP263) and low (28-8, 22C3, and SP142). Reproducibilities across pathologists were higher for QR1 and SP263 (ICC ≥ 0.87 and ≥ 0.85 for TPS and CPS, respectively). QR1, SP263, and 28-8 showed the highest concordance with mRNA expression (ICC ≥ 0.81 for CPS). We developed a standardized method for PD-L1 immunodetection and scoring, tested on 40 metastatic melanomas. Concordances among antibodies were excellent for all criteria, and concordances among pathologists were better for the MELscore than for other scores. CONCLUSION Harmonization of PD-L1 staining and scoring in melanomas with good concordance is achievable using the PD-L1 IHC protocols applied to other cancers; this reproducible approach can simplify daily practice.
Collapse
Affiliation(s)
- M Darmon-Novello
- Department of Pathology, Bordeaux University Hospital and INSERM U1053, Bordeaux, France
| | - J Adam
- Department of Pathology, Gustave Roussy Institute, Paris, France
| | - L Lamant
- Department of Pathology, Oncopole University Hospital Toulouse, France
| | - M Battistella
- Department of Pathology, Hôpital Saint-Louis, AP-HP, Université de Paris, INSERM U976 HIPI, Paris, France
| | - N Ortonne
- Department of Pathology, University Hospital Henri Mondor, Creteil-, Paris, France
| | - B Balme
- Department of Pathology, University Hospital Lyon, France
| | | | - L Chaltiel
- Department of Biostatistics, Institut Claudius Regaud IUCT-O, Toulouse, France
| | - E Gerard
- Department of Dermatology, Bordeaux University Hospital, Bordeaux, France
| | - C Franchet
- Department of Pathology, Oncopole University Hospital Toulouse, France
| | - B Vergier
- Department of Pathology, Bordeaux University Hospital and INSERM U1053, Bordeaux, France
| |
Collapse
|
24
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
25
|
Helweg LP, Windmöller BA, Burghardt L, Storm J, Förster C, Wethkamp N, Wilkens L, Kaltschmidt B, Banz-Jansen C, Kaltschmidt C. The Diminishment of Novel Endometrial Carcinoma-Derived Stem-like Cells by Targeting Mitochondrial Bioenergetics and MYC. Int J Mol Sci 2022; 23:ijms23052426. [PMID: 35269569 PMCID: PMC8910063 DOI: 10.3390/ijms23052426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer stem cells (CSCs) are a small subpopulation of tumor cells harboring properties that include self-renewal, multi-lineage differentiation, tumor reconstitution, drug resistance and invasiveness, making them key players in tumor relapse. In the present paper, we develop new CSC models and analyze the molecular pathways involved in survival to identify targets for the establishment of novel therapies. Endometrial carcinoma-derived stem-like cells (ECSCs) were isolated from carcinogenic gynecological tissue and analyzed regarding their expression of prominent CSC markers. Further, they were treated with the MYC-signaling inhibitor KJ-Pyr-9, chemotherapeutic agent carboplatin and type II diabetes medication metformin. ECSC populations express common CSC markers, such as Prominin-1 and CD44 antigen as well as epithelial-to-mesenchymal transition markers, Twist, Snail and Slug, and exhibit the ability to form free-floating spheres. The inhibition of MYC signaling and treatment with carboplatin as well as metformin significantly reduced the cell survival of ECSC-like cells. Further, treatment with metformin significantly decreased the mitochondrial membrane potential of ECSC-like cells, while the extracellular lactate concentration was increased. The established ECSC-like populations represent promising in vitro models to further study the contribution of ECSCs to endometrial carcinogenesis. Targeting MYC signaling as well as mitochondrial bioenergetics has shown promising results in the diminishment of ECSCs, although molecular signaling pathways need further investigations.
Collapse
Affiliation(s)
- Laureen P. Helweg
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Correspondence: ; Tel.: +49-0521-106-5619
| | - Beatrice A. Windmöller
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Leonie Burghardt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
| | - Jonathan Storm
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| | - Christine Förster
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Nils Wethkamp
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Ludwig Wilkens
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Institute of Pathology, KRH Hospital Nordstadt, Affiliated with the Protestant Hospital of Bethel Foundation, 30167 Hannover, Germany;
| | - Barbara Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Molecular Neurobiology, Faculty of Biology, Bielefeld University, 33615 Bielefeld, Germany
| | - Constanze Banz-Jansen
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
- Department of Gynecology and Obstetrics, and Perinatal Center, Protestant Hospital of Bethel Foundation, University Medical School OWL at Bielefeld, Bielefeld University, Campus Bielefeld-Bethel, 33615 Bielefeld, Germany
| | - Christian Kaltschmidt
- Department of Cell Biology, Faculty of Biology, University of Bielefeld, Universitätsstrasse 25, 33615 Bielefeld, Germany; (B.A.W.); (L.B.); (J.S.); (B.K.); (C.K.)
- Forschungsverbund BioMedizin Bielefeld/OWL FBMB e.V., 33615 Bielefeld, Germany; (C.F.); (L.W.); (C.B.-J.)
| |
Collapse
|
26
|
Lee S, Lin C, Wei C, Chang K, Yuan C, Tsai C, Liu J, Hou H, Tang J, Chou W, Tien H. PD-L1 expression in megakaryocytes and its clinicopathological features in primary myelofibrosis patients. J Pathol Clin Res 2022; 8:78-87. [PMID: 34480529 PMCID: PMC8682945 DOI: 10.1002/cjp2.240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 07/24/2021] [Accepted: 08/05/2021] [Indexed: 11/11/2022]
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by upregulation of proinflammatory cytokines and immune dysregulation, which provide a reasonable basis for immunotherapy in patients. Megakaryocytes are crucial in the pathogenesis of primary myelofibrosis (PMF), the most clinically aggressive subtype of MPN. In this study, we aimed to explore PD-L1 (programmed death-ligand 1) expression in megakaryocytes and its clinical implications in PMF. We analyzed PD-L1 expression on megakaryocytes in PMF patients by immunohistochemistry and correlated the results with clinicopathological features and molecular aberrations. We employed a two-tier grading system considering both the proportion of cells positively stained and the intensity of staining. Among the 85 PMF patients, 41 (48%) showed positive PD-L1 expression on megakaryocytes with the immune-reactive score ranging from 1 to 12. PD-L1 expression correlated closely with higher white blood cell count (p = 0.045), overt myelofibrosis (p = 0.010), JAK2V617F mutation (p = 0.011), and high-molecular risk mutations (p = 0.045), leading to less favorable overall survival in these patients (hazard ratio 0.341, 95% CI 0.135-0.863, p = 0.023). Our study provides unique insights into the interaction between immunologic and molecular phenotypes in PMF patients. Future work to explore the translational potential of PD-L1 in the clinical setting is needed.
Collapse
Affiliation(s)
- Sze‐Hwei Lee
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Tai‐Chen Stem Cell Therapy CenterNational Taiwan UniversityTaipeiTaiwan
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chien‐Chin Lin
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Chao‐Hong Wei
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Ko‐Ping Chang
- Department of PathologyNational Taiwan University HospitalTaipeiTaiwan
| | - Chang‐Tsu Yuan
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of PathologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Cheng‐Hong Tsai
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Jia‐Hao Liu
- Department of Hematology and OncologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Hsin‐An Hou
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Jih‐Lu Tang
- Graduate Institute of Clinical Medicine, College of MedicineNational Taiwan UniversityTaipeiTaiwan
- Department of Hematology and OncologyNational Taiwan University Cancer CenterTaipeiTaiwan
| | - Wen‐Chien Chou
- Department of Laboratory MedicineNational Taiwan University HospitalTaipeiTaiwan
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| | - Hwei‐Fang Tien
- Division of Hematology, Department of Internal MedicineNational Taiwan University HospitalTaipeiTaiwan
| |
Collapse
|
27
|
Sagawa R, Sakata S, Gong B, Seto Y, Takemoto A, Takagi S, Ninomiya H, Yanagitani N, Nakao M, Mun M, Uchibori K, Nishio M, Miyazaki Y, Shiraishi Y, Ogawa S, Kataoka K, Fujita N, Takeuchi K, Katayama R. Soluble PD-L1 through alternative polyadenylation works as a decoy in lung cancer immunotherapy. JCI Insight 2021; 7:153323. [PMID: 34874919 PMCID: PMC8765052 DOI: 10.1172/jci.insight.153323] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 11/23/2021] [Indexed: 11/17/2022] Open
Abstract
Immune checkpoint therapy targeting the PD-1/PD-L1 axis is a novel development in anticancer therapy and has been applied to clinical medicine. However, there are still some problems, including a relatively low response rate, innate mechanisms of resistance against immune checkpoint blockades, and the absence of reliable biomarkers to predict responsiveness. In this study of in vitro and in vivo models, we demonstrate that PD-L1-vInt4, a splicing variant of PD-L1, plays a role as a decoy in anti-PD-L1 antibody treatment. First, we showed that PD-L1-vInt4 was detectable in clinical samples and that it was possible to visualize the secreting variants with IHC. By overexpressing the PD-L1-secreted splicing variant on MC38 cells, we observed that an immune-suppressing effect was not induced by their secretion alone. We then demonstrated that PD-L1-vInt4 secretion resisted anti-PD-L1 antibody treatment, compared with wild type PD-L1, which was explicable by the PD-L1-vInt4's decoying of the anti-PD-L1 antibody. The decoying function of PD-L1 splicing variants may be one of the reasons for cancers being resistant to anti-PD-L1 therapy. Measuring serum PD-L1 levels might be helpful in deciding the therapeutic strategy.
Collapse
Affiliation(s)
- Ray Sagawa
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Seiji Sakata
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Bo Gong
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yosuke Seto
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ai Takemoto
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Satoshi Takagi
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hironori Ninomiya
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Noriko Yanagitani
- Department of Thoracic Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Masayuki Nakao
- Division of Thoracic Surgery, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Mingyon Mun
- Division of Thoracic Surgery, Cancer Institute Hospital of Japanese Foundation of Cancer Research, Tokyo, Japan
| | - Ken Uchibori
- Department of Thoracic Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Makoto Nishio
- Department of Thoracic Medical Oncology, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yasunari Miyazaki
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yuichi Shiraishi
- Division of Genome Analysis Platform Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keisuke Kataoka
- Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Kengo Takeuchi
- Pathology Project for Molecular Targets, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
28
|
Rugo HS, Loi S, Adams S, Schmid P, Schneeweiss A, Barrios CH, Iwata H, Diéras V, Winer EP, Kockx MM, Peeters D, Chui SY, Lin JC, Nguyen-Duc A, Viale G, Molinero L, Emens LA. PD-L1 Immunohistochemistry Assay Comparison in Atezolizumab Plus nab-Paclitaxel-Treated Advanced Triple-Negative Breast Cancer. J Natl Cancer Inst 2021; 113:1733-1743. [PMID: 34097070 PMCID: PMC8634452 DOI: 10.1093/jnci/djab108] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 01/07/2021] [Accepted: 05/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND In the phase III IMpassion130 study, atezolizumab plus nab-paclitaxel (A+nP) showed clinical benefit in advanced or metastatic triple-negative breast cancer patients who were programmed death-ligand 1 (PD-L1)+ (tumor-infiltrating immune cells [IC] ≥1%) using the SP142 immunohistochemistry assay. Here we evaluate 2 other PD-L1 assays for analytical concordance with SP142 and patient-associated clinical outcomes. METHODS Samples from 614 patients (68.1% of intention-to-treat population) were centrally evaluated by immunohistochemistry for PD-L1 status on IC (VENTANA SP142, SP263, Dako 22C3) or as a combined positive score (CPS; 22C3). RESULTS Using SP142, SP263, and 22C3 assays, PD-L1 IC ≥1% prevalence was 46.4% (95% confidence interval [CI] = 42.5% to 50.4%), 74.9% (95% CI = 71.5% to 78.3%), and 73.1% (95% CI = 69.6% to 76.6%), respectively; 80.9% were 22C3 CPS ≥1. At IC ≥1% (+), the analytical concordance between SP142 and SP263 and 22C3 was 69.2% and 68.7%, respectively. Almost all SP142+ cases were captured by other assays (double positive), but several SP263+ (29.6%) or 22C3+ (29.0%) cases were SP142- (single positive). A+nP clinical activity vs placebo+nP in SP263+ and 22C3+ patients (progression-free survival [PFS] hazard ratios [HRs] = 0.64 to 0.68; overall survival [OS] HRs = 0.75 to 0.79) was driven by double-positive cases (PFS HRs = 0.60 to 0.61; OS HRs = 0.71 to 0.75) rather than single-positive cases (PFS HRs = 0.68 to 0.81; OS HRs = 0.87 to 0.95). Concordance for harmonized cutoffs for SP263 (IC ≥4%) and 22C3 (CPS ≥10) to SP142 (IC ≥1%) was subpar (approximately 75%). CONCLUSIONS 22C3 and SP263 assays identified more patients as PD-L1+ (IC ≥1%) than SP142. No inter-assay analytical equivalency was observed. Consistent improved A+nP efficacy was captured by the SP142 PD-L1 IC ≥1% subgroup nested within 22C3 and SP263 PD-L1+ (IC ≥1%) populations.
Collapse
Affiliation(s)
- Hope S Rugo
- Department of Medicine (Hematology/Oncology), University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, USA
| | - Sherene Loi
- Peter MacCallum Cancer Centre and University of Melbourne, Melbourne, Victoria, Australia
| | - Sylvia Adams
- New York University Langone Health, Perlmutter Cancer Center, New York, NY, USA
| | - Peter Schmid
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Andreas Schneeweiss
- National Center for Tumor Diseases (NCT), Heidelberg University Hospital and German Cancer Research Center, Heidelberg, Germany
| | - Carlos H Barrios
- Centro de Pesquisa em Oncologia, Hospital São Lucas, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | | | - Véronique Diéras
- Department of Medical Oncology, Institut Curie, Paris, and Centre Eugène Marquis, Rennes, France
| | | | | | | | | | | | | | - Giuseppe Viale
- Post-graduate Medical School in Pathology, University of Milan, Milan, Italy
- Division of Pathology and Laboratory Medicine, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Leisha A Emens
- University of Pittsburgh Medical Center Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Akhtar M, Rashid S, Al-Bozom IA. PD-L1 immunostaining: what pathologists need to know. Diagn Pathol 2021; 16:94. [PMID: 34689789 PMCID: PMC8543866 DOI: 10.1186/s13000-021-01151-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 09/22/2021] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Immune checkpoint proteins, especially PD-L1 and PD-1, play a crucial role in controlling the intensity and duration of the immune response, thus preventing the development of autoimmunity. These proteins play a vital role in enabling cancer cells to escape immunity, proliferate and progress. METHODS This brief review highlights essential points related to testing for immune checkpoint therapy that histopathologists need to know. RESULTS In recent years, several inhibitors of these proteins have been used to reactivate the immune system to fight cancer. Selection of patients for such therapy requires demonstration of PD-L1 activation on the tumor cells, best done by immunohistochemical staining of the tumor and immune cells using various antibodies with predetermined thresholds. CONCLUSIONS Immune checkpoint therapy appears to be promising and is rapidly expanding to include a large variety of cancers.
Collapse
Affiliation(s)
- Mohammed Akhtar
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| | - Sameera Rashid
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Issam A Al-Bozom
- Department of Laboratory Medicine and Pathology, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar
| |
Collapse
|
30
|
Placke JM, Soun C, Bottek J, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Pfeiffer C, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Zimmer L, Livingstone E, Becker JC, Lodde G, Sucker A, Griewank K, Horn S, Hadaschik E, Roesch A, Schadendorf D, Engel DR, Ugurel S. Digital Quantification of Tumor PD-L1 Predicts Outcome of PD-1-Based Immune Checkpoint Therapy in Metastatic Melanoma. Front Oncol 2021; 11:741993. [PMID: 34621681 PMCID: PMC8491983 DOI: 10.3389/fonc.2021.741993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Background PD-1-based immune checkpoint blockade (ICB) is a highly effective therapy in metastatic melanoma. However, 40-60% of patients are primarily resistant, with valid predictive biomarkers currently missing. This study investigated the digitally quantified tumor PD-L1 expression for ICB therapy outcome prediction. Patients and Methods Tumor tissues taken prior to PD-1-based ICB for unresectable metastatic disease were collected within the prospective multicenter Tissue Registry in Melanoma (TRIM). PD-L1 expression (clone 28-8; cut-off=5%) was determined by digital and physician quantification, and correlated with therapy outcome (best overall response, BOR; progression-free survival, PFS; overall survival, OS). Results Tissue samples from 156 patients were analyzed (anti-PD-1, n=115; anti-CTLA-4+anti-PD-1, n=41). Patients with PD-L1-positive tumors showed an improved response compared to patients with PD-L1-negative tumors, by digital (BOR 50.5% versus 32.2%; p=0.026) and physician (BOR 54.2% versus 36.6%; p=0.032) quantification. Tumor PD-L1 positivity was associated with a prolonged PFS and OS by either digital (PFS, 9.9 versus 4.6 months, p=0.021; OS, not reached versus 13.0 months, p=0.001) or physician (PFS, 10.6 versus 5.6 months, p=0.051; OS, not reached versus 15.6 months, p=0.011) quantification. Multivariable Cox regression revealed digital (PFS, HR=0.57, p=0.007; OS, HR=0.44, p=0.001) and physician (OS, HR=0.54, p=0.016) PD-L1 quantification as independent predictors of survival upon PD-1-based ICB. The combination of both methods identified a patient subgroup with particularly favorable therapy outcome (PFS, HR=0.53, p=0.011; OS, HR=0.47, p=0.008). Conclusion Pre-treatment tumor PD-L1 positivity predicted a favorable outcome of PD-1-based ICB in melanoma. Herein, digital quantification was not inferior to physician quantification, and should be further validated for clinical use.
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Camille Soun
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Jenny Bottek
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Rudolf Herbst
- Department of Dermatology, Medical Hospital, Erfurt, Germany
| | | | - Jochen Utikal
- Department of Dermatology, Venerology, and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg, Germany
| | - Jens Ulrich
- Department of Dermatology, Medical Hospital of Quedlinburg, Quedlinburg, Germany
| | - Alexander Kreuter
- Department of Dermatology, Venereology, and Allergology, Helios St. Elisabeth Hospital Oberhausen, University of Witten-Herdecke, Oberhausen, Germany
| | - Christiane Pfeiffer
- Department of Dermatology, Venereology, and Allergology University Ulm, Ulm, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, Buxtehude, Germany
| | - Ralf Gutzmer
- Skin Cancer Center, Department of Dermatology, Hannover Medical School, Hannover, Germany
| | - Friedegund Meier
- German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, University Hospital Dresden, Dresden, Germany
| | - Edgar Dippel
- Hautklinik, Klinikum der Stadt Ludwigshafen am Rhein gGmbH, Ludwigshafen, Germany
| | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translationale Hautkrebsforschung, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Robert Engel
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
31
|
Turchini J, Sioson L, Clarkson A, Sheen A, Gill AJ. PD-L1 Is Preferentially Expressed in PIT-1 Positive Pituitary Neuroendocrine Tumours. Endocr Pathol 2021; 32:408-414. [PMID: 33694064 DOI: 10.1007/s12022-021-09673-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 12/19/2022]
Abstract
Pituitary neuroendocrine tumours (PitNETs) cause lifelong morbidity, some requiring extensive surgical intervention, radiotherapy, or chemotherapy. A small percentage still cause debilitating disease, resistant to standard treatments, and may benefit from novel therapies. We assessed PD-L1 expression in a large cohort of PitNETs to investigate whether immunotherapy could represent a rational therapeutic choice. Unselected PitNETs undergoing surgical resection were reclassified according to the WHO 2017 system and underwent PD-L1 immunohistochemistry (clone SP263) in tissue microarray format. Membranous expression was scored as 0 (no expression), 1+ (< 50% expression) and 2+ (> 50% expression). A total of 265 PitNETs underwent PD-L1 immunohistochemistry. Prominent non-specific cytoplasmic staining was noted making assessment of true membrane expression difficult. Allowing for this, 40 of 264 (15%) PitNETs demonstrated strong staining (> 50% of neoplastic cells positive). These included 5/10 (50%) somatotrophs, 7/17 (41%) lactotrophs, 2/5 (40%) mammosomatotrophs, 4/8 (50%) mixed somatotroph-lactotrophs, 3/5 (60%) PIT-1 positive plurihormonal tumours with TSH expression, 10/28 (36%) of PIT-1 positive plurihormonal tumours, and 4/10 (40%) of PIT-1 positive tumours with no hormonal expression. Only 2/32 (6%) transcription factor triple negative, hormone negative tumours, 5/113 (4%) of gonadotrophs, and 0/6 thyrotrophs or 0/30 corticotrophs showed significant staining. We conclude that PD-L1 expression is common in somatotrophs, lactotrophs, and PIT-1 positive plurihormonal PitNETs but rare in transcription factor negative, hormone negative PitNETs, gonadotrophs, and corticotrophs. If the therapeutic role of immunotherapy is to be explored in PitNETs, it may be that it is of most benefit in the PD-L1 high subgroup.
Collapse
Affiliation(s)
- John Turchini
- Anatomical Pathology, Douglass Hanly Moir Pathology, 14 Giffnock Avenue, Macquarie Park, NSW, 2113, Australia.
- Discipline of Pathology, MQ Health, Macquarie University, Macquarie Park, NSW, 2113, Australia.
- Sydney Medical School, The University of Sydney, Sydney, 2006, Australia.
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia.
| | - Loretta Sioson
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
| | - Adele Clarkson
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, NSW Health Pathology, St Leonards, NSW, 2065, Australia
| | - Amy Sheen
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
| | - Anthony J Gill
- Sydney Medical School, The University of Sydney, Sydney, 2006, Australia
- Cancer Diagnosis and Pathology Group, Kolling Institute of Medical Research, St Leonards, NSW, 2065, Australia
- Department of Anatomical Pathology, Royal North Shore Hospital, NSW Health Pathology, St Leonards, NSW, 2065, Australia
| |
Collapse
|
32
|
Emens LA, Adams S, Cimino-Mathews A, Disis ML, Gatti-Mays ME, Ho AY, Kalinsky K, McArthur HL, Mittendorf EA, Nanda R, Page DB, Rugo HS, Rubin KM, Soliman H, Spears PA, Tolaney SM, Litton JK. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immunotherapy for the treatment of breast cancer. J Immunother Cancer 2021; 9:e002597. [PMID: 34389617 PMCID: PMC8365813 DOI: 10.1136/jitc-2021-002597] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer has historically been a disease for which immunotherapy was largely unavailable. Recently, the use of immune checkpoint inhibitors (ICIs) in combination with chemotherapy for the treatment of advanced/metastatic triple-negative breast cancer (TNBC) has demonstrated efficacy, including longer progression-free survival and increased overall survival in subsets of patients. Based on clinical benefit in randomized trials, ICIs in combination with chemotherapy for the treatment of some patients with advanced/metastatic TNBC have been approved by the United States (US) Food and Drug Administration (FDA), expanding options for patients. Ongoing questions remain, however, about the optimal chemotherapy backbone for immunotherapy, appropriate biomarker-based selection of patients for treatment, the optimal strategy for immunotherapy treatment in earlier stage disease, and potential use in histological subtypes other than TNBC. To provide guidance to the oncology community on these and other important concerns, the Society for Immunotherapy of Cancer (SITC) convened a multidisciplinary panel of experts to develop a clinical practice guideline (CPG). The expert panel drew upon the published literature as well as their clinical experience to develop recommendations for healthcare professionals on these important aspects of immunotherapeutic treatment for breast cancer, including diagnostic testing, treatment planning, immune-related adverse events (irAEs), and patient quality of life (QOL) considerations. The evidence-based and consensus-based recommendations in this CPG are intended to give guidance to cancer care providers treating patients with breast cancer.
Collapse
Affiliation(s)
- Leisha A Emens
- Department of Medicine, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Sylvia Adams
- Perlmutter Cancer Center, New York University Langone, New York, New York, USA
| | - Ashley Cimino-Mathews
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Mary L Disis
- Cancer Vaccine Institute, University of Washington, Seattle, Washington, USA
| | - Margaret E Gatti-Mays
- Pelotonia Institute for Immuno-Oncology, Division of Medical Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio, USA
| | - Alice Y Ho
- Department of Radiation Oncology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kevin Kalinsky
- Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | | | - Elizabeth A Mittendorf
- Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Breast Oncology Program, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts, USA
| | - Rita Nanda
- Department of Medicine, Section of Hematology/Oncology, The University of Chicago Medicine Comprehensive Cancer Center, Chicago, Illinois, USA
| | - David B Page
- Earle A Chiles Research Institute, Portland, Oregon, USA
| | - Hope S Rugo
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, California, USA
| | - Krista M Rubin
- Center for Melanoma, Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - Hatem Soliman
- Department of Breast Oncology, H Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Patricia A Spears
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill, North Carolina, USA
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Jennifer K Litton
- Department of Breast Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
33
|
Miracco C, Toti P, Gelmi MC, Aversa S, Baldino G, Galluzzi P, De Francesco S, Petrelli F, Sorrentino E, Belmonte G, Galimberti D, Bracco S, Hadjistilianou T. Retinoblastoma Is Characterized by a Cold, CD8+ Cell Poor, PD-L1- Microenvironment, Which Turns Into Hot, CD8+ Cell Rich, PD-L1+ After Chemotherapy. Invest Ophthalmol Vis Sci 2021; 62:6. [PMID: 33538768 PMCID: PMC7862737 DOI: 10.1167/iovs.62.2.6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Purpose To investigate the impact of chemotherapy (CHT) on human retinoblastoma (RB) tumor microenvironment (TME). Cases and Methods Ninety-four RBs were studied, including 44 primary RBs treated by upfront surgery (Group 1) and 50 primary RBs enucleated after CHT (CHT), either intra-arterial (IAC; Group 2, 33 cases) or systemic (S-CHT; Group 3, 17 cases). Conventional and multiplexed immunohistochemistry were performed to make quantitative comparisons among the three groups, for the following parameters: tumor-infiltrating inflammatory cells (TI-ICs); programmed cell death protein 1 (PD-1) positive TI-ICs; Ki67 proliferation index; gliosis; PD-1 ligand (PD-L1) protein expression; vessel number. We also correlated these TME factors with the presence of histological high-risk factors (HHRF+) and RB anaplasia grade (AG). Results After CHT, a decrease in both RB burden and Ki67 positivity was observed. In parallel, most subsets of TI-ICs, PD-1+ TI-ICs, gliosis, and PD-L1 protein expression significantly increased (P < 0.001, P = 0.02, P < 0.001, respectively). Vessel number did not significantly vary. Age, HHRFs+ and AG were significantly different between primary and chemoreduced RBs (P < 0.001, P = 0.006, P = 0.001, respectively) and were correlated with most TME factors. Conclusions CHT modulates host antitumor immunity by reorienting the RB TME from anergic into an active, CD8+, PD-L1+ hot state. Furthermore, some clinicopathological characteristics of RB correlate with several factors of TME. Our study adds data in favor of the possibility of a new therapeutic scenario in human RB.
Collapse
Affiliation(s)
- Clelia Miracco
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Paolo Toti
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Maria Chiara Gelmi
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| | - Sara Aversa
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Gennaro Baldino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Paolo Galluzzi
- Department of Medicine, Surgery and Neuroscience, Unit of Neuroimaging and Neurointervention, University Hospital of Siena, Siena, Italy
| | - Sonia De Francesco
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| | - Federica Petrelli
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Ester Sorrentino
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Giuseppe Belmonte
- Department of Medicine, Surgery and Neuroscience, Pathological Anatomy Section, University Hospital of Siena, Siena, Italy
| | - Daniela Galimberti
- Department of Maternal, Newborn and Child Health, Unit of Pediatrics, University Hospital of Siena, Siena, Italy
| | - Sandra Bracco
- Department of Medicine, Surgery and Neuroscience, Unit of Neuroimaging and Neurointervention, University Hospital of Siena, Siena, Italy
| | - Theodora Hadjistilianou
- Department of Medicine, Surgery and Neuroscience, Ophthalmology Unit, University Hospital of Siena, Siena, Italy
| |
Collapse
|
34
|
Koumarianou A, Kaltsas GA, Chatzellis E, Kyriakopoulos G, Kolomodi D, Alexandraki KI. Immunotherapeutics at the spearhead: current status in targeting neuroendocrine neoplasms. Endocrine 2021; 73:232-239. [PMID: 33544352 DOI: 10.1007/s12020-021-02639-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/18/2021] [Indexed: 01/18/2023]
Abstract
PURPOSE Recent advances in the field of immunotherapy have significantly prolonged the survival of patients with aggressive carcinomas, but the role of immunotherapy in neuroendocrine neoplasms (NENs) remains to be elucidated. METHODS AND RESULTS We report a patient diagnosed with a well-differentiated grade 3 pancreatic NEN (pNEN) and type 3 liver metastases who received compassionate nivolumab as a fifth line treatment and achieved a durable partial response of more than 34 months. We have performed a systematic review to the literature on tumor microenvironment and potential biomarkers in the field of NEN including the tumor mutational burden, the tumor infiltrating lymphocytes, the programmed cell death ligand 1, and the mismatch repair system. The potential role of the immune system modulation together with a critical assessment of the recent phase II clinical studies in NEN including monotherapy with anti-PD-1/PD-L1 monoclonal antibodies, and combination therapies including anti-PD-1 along with anti-CTLA-4 monoclonal antibodies are also provided. CONCLUSION Immunotherapeutics are gaining a post in the field of NENs in cases progressing during the course of the disease, dictating urgently the identification of biomarkers that will enable selection of NEN patients who may benefit from this treatment.
Collapse
Affiliation(s)
- Anna Koumarianou
- Hematology Oncology Unit, Fourth Department of Internal Medicine, Attikon University Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece.
| | - Gregory A Kaltsas
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
- 1st Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Eleftherios Chatzellis
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | - Georgios Kyriakopoulos
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | - Denise Kolomodi
- European Neuroendocrine Tumor Society, ENETS Center of Excellence, EKPA-LAIKO CENTER, Athens, Greece
| | | |
Collapse
|
35
|
Clinicopathologic and genomic characterization of PD-L1-positive uterine cervical carcinoma. Mod Pathol 2021; 34:1425-1433. [PMID: 33637877 DOI: 10.1038/s41379-021-00780-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 12/28/2022]
Abstract
Positive program death-ligand 1 (PD-L1) immunohistochemistry (IHC) is an approved companion diagnostic guiding the use of immune checkpoint inhibitors in uterine cervical carcinoma (CXC). The clinical and genomic features of PD-L1-positive (PD-L1positive) CXC have not been previously described. We reviewed the clinicopathologic and molecular features of 647 CXC cases that were tested using DAKO 22C3 PD-L1 IHC and comprehensive genomic profiling during the course of clinical care. PD-L1positive cases were defined via a combined positive score of ≥ 1. No differences were found in age, genetic ancestry, and HPV status of the PD-L1positive (n = 548) and PD-L1negative disease subset. The PD-L1 positivity rate varied by histologic subtype of CXC with squamous cell carcinoma (SCC) having a PD-L1 positivity rate of 91% (397/437) and usual-type adenocarcinoma's PD-L1 positivity rate being 60% (35/58). In addition, the PD-L1 positivity rate varied depending on site of the specimen with 89.1% (261/293) positivity rate observed in cervix specimens compared to 25% (2/8) in brain metastases specimens. No significant difference in tumor mutational burden (TMB), microsatellite instability, and CD274 (encoding PD-L1) amplification was observed between PD-L1positive and PD-L1negative CXC subsets. By combining TMB with PD-L1, an additional 17 patients are eligible for pembrolizumab when compared to PD-L1 testing alone. TERT promoter alterations and APOBEC mutational signature were enriched in the PD-L1positive CXC SCC (p = 0.011, and p = 0.004, respectively). Our study reveals important prevalence data on PD-L1 positivity in CXC non-SCC and suggests that further studies in these histologic subtypes are warranted. In addition, we also provide a key framework to guide both specimen selection and future investigations of predictors of immunotherapy response in cervical cancer patients. Lastly, TERT promoter alterations and APOBEC mutational signature may be a biologically unique subset of PD-L1positive CXC SCC.
Collapse
|
36
|
Huang RSP, Decker B, Murugesan K, Hiemenz M, Mata DA, Li G, Creeden J, Ramkissoon SH, Ross JS. Pan-cancer analysis of CD274 (PD-L1) mutations in 314,631 patient samples and subset correlation with PD-L1 protein expression. J Immunother Cancer 2021; 9:e002558. [PMID: 34130989 PMCID: PMC8207995 DOI: 10.1136/jitc-2021-002558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2021] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND The effects of non-amplification short variant (SV) mutations in CD274 (programmed death-ligand 1 (PD-L1)) on PD-L1 protein expression and immune checkpoint inhibitors (ICPIs) therapy are unknown. Here, we present a retrospective analysis of CD274 mutations detected by comprehensive genomic profiling (CGP) and correlate these results with tumor-cell PD-L1 immunohistochemistry (IHC)-based expression assessment to better understand the relationship between mutations and protein expression of PD-L1. METHODS CGP was performed on hybridization-captured, adaptor ligation-based libraries using DNA and/or RNA extracted from 314,631 tumor samples that were sequenced for up to 406 cancer-related genes and select gene rearrangements. PD-L1 IHC was performed on a subset of cases (n=58,341) using the DAKO 22C3 PD-L1 IHC assay and scored with the tumor proportion score (TPS). RESULTS Overall, the prevalence of CD274 SV mutations was low (0.3%, 1081/314,631) with 577 unique variants. The most common CD274 SV mutations were R260H (n=51), R260C (n=18), R125Q (n=12), C272fs*13 (n=11), R86W (n=10), and R113H (n=10). The prevalence of CD274 mutations varied depending on tumor type with diffuse large B-cell lymphoma (1.9%, 19/997), cutaneous squamous cell carcinoma (1.6%, 14/868), endometrial adenocarcinoma (1.0%, 36/3740), unknown primary melanoma (0.9%, 33/3679), and cutaneous melanoma (0.8%, 32/3874) having the highest frequency of mutations. Of the R260H cases concurrently tested with PD-L1 IHC, most (81.8%, 9/11) had no PD-L1 expression, which contrasts to the five E237K cases where most (80%, 4/5) had PD-L1 expression. In addition, we saw a significantly lower level of PD-L1 expression in samples with a clonal truncating variant (nonsense or frameshift indel) when compared with samples with a subclonal truncating variants (mean: TPS=1 vs TPS=38; p<0.001), and also in clonal versus subclonal missense mutations (mean: TPS=11 vs TPS=22, respectively; p=0.049) CONCLUSIONS: We defined the landscape of CD274 mutations in a large cohort of tumor types that can be used as a reference for examining CD274 mutations as potential resistance biomarkers for ICPI. Furthermore, we presented novel data on the correlation of CD274 mutations and PD-L1 protein expression, providing important new information on the potential functionality of these mutations and can serve as a basis for future research.
Collapse
Affiliation(s)
| | | | | | | | | | - Gerald Li
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
| | - James Creeden
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
| | - Shakti H Ramkissoon
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
- Wake Forest Comprehensive Cancer Center, Winston-Salem, North Carolina, USA
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jeffrey S Ross
- Foundation Medicine Inc, Cambridge, Massachusetts, USA
- Department of Pathology, State University of New York (SUNY) Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
37
|
Huang X, Zhang ZH, Chen J, Mao Z, Zhu H, Liu Y, Zhu Z, Chen H. One dimensional magneto-optical nanocomplex from silver nanoclusters and magnetite nanorods containing ordered mesocages for sensitive detection of PD-L1. Biosens Bioelectron 2021; 189:113385. [PMID: 34091282 DOI: 10.1016/j.bios.2021.113385] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/26/2021] [Accepted: 05/28/2021] [Indexed: 10/21/2022]
Abstract
Programmed death ligand 1 (PD-L1) is a typical immune checkpoint protein, whose up-regulation on the membrane of different tumor cells inhibits the immune response of T cells and leads to the escape of tumor cells. In this work, we designed a facile and highly specific surface plasmon resonance (SPR) biosensor to detect PD-L1 in human plasma based on magnetite nanorods containing ordered mesocages (MNOM) and silver nanoclusters (AgNCs). Magneto-optical nanocomplex MNOM@AgNCs with superior magneto-optical properties and high signal-to-noise ratio were fabricated to improve the detection sensitivity owing to the high specific surface area of MNOM and excellent localized SPR of AgNCs. The PD-L1 Antibody on the surface of gold chip and the PD-L1 aptamer on MNOM@AgNCs could realize dual selective recognition of PD-L1, providing the specificity of the sensor and reducing non-specific binding. The SPR sensor showed a good linear range of PD-L1 from 10 ng/mL to 300 ng/mL with the detection limit of 3.29 ng/mL. The practical performance of this immunosensing platform had been successfully verified by clinical samples which included healthy donors and cancer patients. Based on the analysis, the developed immunosensor provided a new strategy for point-of-care detection of PD-L1 and could be used as clinical companion diagnosis of PD-1/PD-L1 inhibitor therapy.
Collapse
Affiliation(s)
- Xing Huang
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Zhao-Huan Zhang
- Department of Laboratory Medicine, Changzheng Hospital, Naval Medical University, Shanghai, 200003, PR China
| | - Jie Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China; School of Medicine, Shanghai University, Shanghai, 200444, PR China
| | - Zhihui Mao
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China; School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Han Zhu
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China
| | - Yawen Liu
- School of Medicine, Shanghai University, Shanghai, 200444, PR China; School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, PR China
| | - Zhongzheng Zhu
- Department of Oncology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 301 Middle Yanchang Road, Shanghai, 200072, PR China.
| | - Hongxia Chen
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai, 200444, PR China.
| |
Collapse
|
38
|
Pembrolizumab in a Patient with Treatment-Naïve Unresectable BRAF-Mutation Negative Anaplastic Thyroid Cancer. Case Rep Endocrinol 2021; 2021:5521649. [PMID: 34123437 PMCID: PMC8166484 DOI: 10.1155/2021/5521649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 05/12/2021] [Indexed: 11/17/2022] Open
Abstract
Immune check point inhibitor (ICI) therapy can be a potentially effective salvage treatment for anaplastic thyroid cancer (ATC) with progression despite standard of care therapies. We report a case of unresectable treatment-naïve ATC showing a dramatic and durable response to first-line pembrolizumab therapy. A 69-year-old male presented with a large, right-sided neck mass associated with compressive symptoms. A neck ultrasound showed a large, right-sided, and highly suspicious thyroid nodule. A fine needle aspiration (FNA) biopsy revealed tumor cells consistent with ATC that were positive for PD-L1, with an expression score of >95% and negative for the BRAF V600E mutation. Imaging studies were negative for distant metastases. The disease was declared surgically inoperable, and the patient declined chemotherapy/radiation therapy (XRT), but agreed to ICI therapy with intravenous pembrolizumab 200 mg every three weeks. The patient has received 25 doses of pembrolizumab to date, with rapid resolution of symptoms and a significant reduction in tumor size. He remains alive without disease progression 18 months since initial diagnosis.
Collapse
|
39
|
Garutti M, Bonin S, Buriolla S, Bertoli E, Pizzichetta MA, Zalaudek I, Puglisi F. Find the Flame: Predictive Biomarkers for Immunotherapy in Melanoma. Cancers (Basel) 2021; 13:cancers13081819. [PMID: 33920288 PMCID: PMC8070445 DOI: 10.3390/cancers13081819] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 03/24/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy has revolutionized the therapeutic landscape of melanoma. In particular, checkpoint inhibition has shown to increase long-term outcome, and, in some cases, it can be virtually curative. However, the absence of clinically validated predictive biomarkers is one of the major causes of unpredictable efficacy of immunotherapy. Indeed, the availability of predictive biomarkers could allow a better stratification of patients, suggesting which type of drugs should be used in a certain clinical context and guiding clinicians in escalating or de-escalating therapy. However, the difficulty in obtaining clinically useful predictive biomarkers reflects the deep complexity of tumor biology. Biomarkers can be classified as tumor-intrinsic biomarkers, microenvironment biomarkers, and systemic biomarkers. Herein we review the available literature to classify and describe predictive biomarkers for checkpoint inhibition in melanoma with the aim of helping clinicians in the decision-making process. We also performed a meta-analysis on the predictive value of PDL-1.
Collapse
Affiliation(s)
- Mattia Garutti
- CRO Aviano National Cancer Institute IRCCS, 33081 Aviano, Italy; (E.B.); (M.A.P.); (F.P.)
- Correspondence:
| | - Serena Bonin
- DSM—Department of Medical Sciences, University of Trieste, 34123 Trieste, Italy;
| | - Silvia Buriolla
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
- Dipartimento di Oncologia, Azienda Sanitaria Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Elisa Bertoli
- CRO Aviano National Cancer Institute IRCCS, 33081 Aviano, Italy; (E.B.); (M.A.P.); (F.P.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
| | - Maria Antonietta Pizzichetta
- CRO Aviano National Cancer Institute IRCCS, 33081 Aviano, Italy; (E.B.); (M.A.P.); (F.P.)
- Department of Dermatology, University of Trieste, 34123 Trieste, Italy;
| | - Iris Zalaudek
- Department of Dermatology, University of Trieste, 34123 Trieste, Italy;
| | - Fabio Puglisi
- CRO Aviano National Cancer Institute IRCCS, 33081 Aviano, Italy; (E.B.); (M.A.P.); (F.P.)
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy;
| |
Collapse
|
40
|
Wang JY, Xiu J, Baca Y, Arai H, Battaglin F, Kawanishi N, Soni S, Zhang W, Millstein J, Shields AF, Grothey A, Weinberg BA, Marshall JL, Lou E, Khushman M, Sohal DPS, Hall MJ, Oberley M, Spetzler D, Shen L, Korn WM, Lenz HJ. Distinct genomic landscapes of gastroesophageal adenocarcinoma depending on PD-L1 expression identify mutations in RAS-MAPK pathway and TP53 as potential predictors of immunotherapy efficacy. Ann Oncol 2021; 32:906-916. [PMID: 33798656 DOI: 10.1016/j.annonc.2021.03.203] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 03/21/2021] [Accepted: 03/28/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The impact of molecular alterations on programmed death-ligand 1 (PD-L1) combined positive score (CPS) is not well studied in gastroesophageal adenocarcinomas (GEAs). We aimed to characterize genomic features of tumors with different CPSs in GEAs. PATIENTS AND METHODS Genomic alterations of 2518 GEAs were compared in three groups (PD-L1 CPS ≥ 10, high; CPS = 1-9, intermediate; CPS < 1, low) using next-generation sequencing. We assessed the impact of gene mutations on the efficacy of immune checkpoint inhibitors (ICIs) and tumor immune environment based on the Memorial Sloan Kettering Cancer Center and The Cancer Genome Atlas databases. RESULTS High, intermediate, and low CPSs were seen in 18%, 54% and 28% of GEAs, respectively. PD-L1 positivity was less prevalent in women and in tissues derived from metastatic sites. PD-L1 CPS was positively associated with mismatch repair deficiency/microsatellite instability-high, but independent of tumor mutation burden distribution. Tumors with mutations in KRAS, TP53, and RAS-mitogen-activated protein kinase (MAPK) pathway were associated with higher PD-L1 CPSs in the mismatch repair proficiency and microsatellite stability (pMMR&MSS) subgroup. Patients with RAS-MAPK pathway alterations had longer overall survival (OS) from ICIs compared to wildtype (WT) patients [27 versus 13 months, hazard ratio (HR) = 0.36, 95% confidence interval (CI): 0.19-0.7, P = 0.016] and a similar trend was observed in the MSS subgroup (P = 0.11). In contrast, patients with TP53 mutations had worse OS from ICIs compared to TP53-WT patients in the MSS subgroup (5 versus 21 months, HR = 2.39, 95% CI: 1.24-4.61, P = 0.016). CONCLUSIONS This is the largest study to investigate the distinct genomic landscapes of GEAs with different PD-L1 CPSs. Our data may provide novel insights for patient selection using mutations in TP53 and RAS-MAPK pathway and for the development of rational combination immunotherapies in GEAs.
Collapse
Affiliation(s)
- J Y Wang
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China; Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - J Xiu
- Caris Life Sciences, Phoenix, USA
| | - Y Baca
- Caris Life Sciences, Phoenix, USA
| | - H Arai
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - F Battaglin
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - N Kawanishi
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - S Soni
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - W Zhang
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - J Millstein
- Department of Preventive Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - A F Shields
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, USA
| | - A Grothey
- GI Cancer Research, West Cancer Center and Research Institute, Germantown, USA
| | - B A Weinberg
- Division of Hematology and Oncology, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, USA
| | - J L Marshall
- Division of Hematology and Oncology, Ruesch Center for the Cure of Gastrointestinal Cancers, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, USA
| | - E Lou
- Division of Hematology, Oncology and Transplantation, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| | - M Khushman
- Department of Interdisciplinary Clinical Oncology, Mitchell Cancer Institute, University of South Alabama, Mobile, USA
| | - D P S Sohal
- Division of Hematology/Oncology, University of Cincinnati, Cincinnati, USA
| | - M J Hall
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, USA
| | | | | | - L Shen
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - W M Korn
- Caris Life Sciences, Phoenix, USA
| | - H J Lenz
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, USA.
| |
Collapse
|
41
|
Development of a Novel Weighted Ranking Method for Immunohistochemical Quantification of a Heterogeneously Expressed Protein in Gastro-Esophageal Cancers. Cancers (Basel) 2021; 13:cancers13061286. [PMID: 33805812 PMCID: PMC7998246 DOI: 10.3390/cancers13061286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 03/06/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022] Open
Abstract
Simple Summary High levels of the protein Junctional Adhesion Molecule-A (JAM-A) have been linked with aggressive disease in patients with several different cancers. However, its distribution is often non-uniform (heterogeneous) across tumors, and can be difficult to quantify. JAM-A has also been linked with high levels of HER2 (an important oncogene) in breast tumors, and the development of resistance to HER2-targeted drugs in those patients. Since gastro-esophageal (GE) cancers are often high in HER2 and have also been approved for HER2-targeted drugs, the aim of this study was to investigate if levels of JAM-A and HER2 are linked in GE cancer. JAM-A was expressed very heterogeneously across miniaturized tissue sections called tissue microarrays (TMAs) of GE cancer patients. In this model, therefore, there was no correlation between JAM-A and HER2 expression. However, when we used larger tissue sections and developed a scoring system to account for heterogeneity, a significant correlation between JAM-A and HER2 levels emerged. This work illustrates the importance of taking intra-tumor heterogeneity into account, particularly in an era when analysis of protein levels by this method is increasingly used to select patients for targeted cancer drugs. Abstract High expression of Junctional Adhesion Molecule-A (JAM-A) has been linked with poor prognosis in several cancers, including breast cancers overexpressing the human epidermal growth factor receptor-2 (HER2). Furthermore, JAM-A expression has been linked with regulating that of HER2, and associated with the development of resistance to HER2-targeted therapies in breast cancer patients. The purpose of this study was to establish a potential relationship between JAM-A and HER2 in HER2-overexpressing gastro-esophageal (GE) cancers. Interrogation of gene expression datasets revealed that high JAM-A mRNA expression was associated with poorer survival in HER2-positive gastric cancer patients. However, high intra-tumoral heterogeneity of JAM-A protein expression was noted upon immunohistochemical scoring of a GE cancer tissue microarray (TMA), precluding a simple confirmation of any relationship between JAM-A and HER2 at protein level. However, in a test-set of 25 full-face GE cancer tissue sections, a novel weighted ranking system proved effective in capturing JAM-A intra-tumoral heterogeneity and confirming statistically significant correlations between JAM-A/HER2 expression. Given the growing importance of immunohistochemistry in stratifying cancer patients for the receipt of new targeted therapies, this may sound a cautionary note against over-relying on cancer TMAs in biomarker discovery studies of heterogeneously expressed proteins. It also highlights a timely need to develop validated mechanisms of capturing intra-tumoral heterogeneity to aid in future biomarker/therapeutic target development for the benefit of cancer patients.
Collapse
|
42
|
PD-L1 immunohistochemistry for canine cancers and clinical benefit of anti-PD-L1 antibody in dogs with pulmonary metastatic oral malignant melanoma. NPJ Precis Oncol 2021; 5:10. [PMID: 33580183 PMCID: PMC7881100 DOI: 10.1038/s41698-021-00147-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy targeting programmed cell death 1 (PD-1) and PD-ligand 1 (PD-L1) represents promising treatments for human cancers. Our previous studies demonstrated PD-L1 overexpression in some canine cancers, and suggested the therapeutic potential of a canine chimeric anti-PD-L1 monoclonal antibody (c4G12). However, such evidence is scarce, limiting the clinical application in dogs. In the present report, canine PD-L1 expression was assessed in various cancer types, using a new anti-PD-L1 mAb, 6C11-3A11, and the safety and efficacy of c4G12 were explored in 29 dogs with pulmonary metastatic oral malignant melanoma (OMM). PD-L1 expression was detected in most canine malignant cancers including OMM, and survival was significantly longer in the c4G12 treatment group (median 143 days) when compared to a historical control group (n = 15, median 54 days). In dogs with measurable disease (n = 13), one dog (7.7%) experienced a complete response. Treatment-related adverse events of any grade were observed in 15 dogs (51.7%). Here we show that PD-L1 is a promising target for cancer immunotherapy in dogs, and dogs could be a useful large animal model for human cancer research.
Collapse
|
43
|
Huang RSP, Haberberger J, Severson E, Duncan DL, Hemmerich A, Edgerly C, Ferguson NL, Williams E, Elvin J, Vergilio JA, Killian JK, Lin DI, Tse J, Hiemenz M, Owens C, Danziger N, Hegde PS, Venstrom J, Alexander B, Ross JS, Ramkissoon SH. A pan-cancer analysis of PD-L1 immunohistochemistry and gene amplification, tumor mutation burden and microsatellite instability in 48,782 cases. Mod Pathol 2021; 34:252-263. [PMID: 32884129 DOI: 10.1038/s41379-020-00664-y] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 01/20/2023]
Abstract
PD-L1 immunohistochemistry (IHC) currently has the most Food and Drug Administration (FDA) approvals as a companion diagnostic (CDx) for immunotherapies in specific tumor types; however, multiple other immunotherapy biomarkers exist. We performed this study to examine and report the prevalence of PD-L1 expression in a wide variety of tumor types and examine its relationship to microsatellite instability (MSI), tumor mutational burden (TMB), and CD274 (PD-L1) gene amplification. We performed a retrospective analysis of all cases in which both PD-L1 IHC (using the DAKO 22C3 IHC assay with either tumor proportion score (TPS) or combined positive score (CPS); or the VENTANA SP142 assay with infiltrating immune cell score (IC)) and comprehensive genomic profiling (CGP) were tested at Foundation Medicine between January 2016 and November 2019. Of note, PD-L1 positivity is defined per the CDx indication and tumor proportion score (TPS ≥ 1) for indications without a CDx claim; and TMB positivity is defined as ≥10 mutations/Mb. A total of 48,782 cases were tested for PD-L1 IHC and CGP. Immune cell expression of PD-L1 was more frequently identified than tumor cell expression of PD-L1. We saw a high correlation between PD-L1 expression and CD274 gene amplification (p < 0.0001), MSI and TMB (p < 0.0001), and PD-L1 and TMB (p < 0.0001). In addition, the combination of PD-L1 and TMB identified four unique disease subsets PD-L1-/TMB-, PD-L1+/TMB-, PD-L1-/TMB+, and PD-L1+/TMB+ with varying prevalence dependent on tumor type. Lastly, 50.3% (24527/48782) of the overall cohort was positive for at least one of the CDx or exploratory biomarkers described above. This is the largest pan-cancer analysis of relevant biomarkers associated with response to checkpoint inhibitors to date, including more than 48,000 cases. Additional clinical trials with treatment outcome data in individual tumor types are needed to determine whether the double positive PD-L1+/TMB+ disease subset would respond best to immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Julia Elvin
- Foundation Medicine, Inc., Cambridge, MA, USA
| | | | | | | | - Julie Tse
- Foundation Medicine, Inc., Cambridge, MA, USA
| | | | | | | | | | | | | | - Jeffrey S Ross
- Foundation Medicine, Inc., Cambridge, MA, USA.,Department of Pathology, State University of New York (SUNY) Upstate Medical University, Syracuse, NY, USA
| | - Shakti H Ramkissoon
- Foundation Medicine, Inc., Morrisville, NC, USA.,Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| |
Collapse
|
44
|
Weng CY, Kao CX, Chang TS, Huang YH. Immuno-Metabolism: The Role of Cancer Niche in Immune Checkpoint Inhibitor Resistance. Int J Mol Sci 2021; 22:1258. [PMID: 33514004 PMCID: PMC7865434 DOI: 10.3390/ijms22031258] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICI) in treating cancer has revolutionized the approach to eradicate cancer cells by reactivating immune responses. However, only a subset of patients benefits from this treatment; the majority remains unresponsive or develops resistance to ICI therapy. Increasing evidence suggests that metabolic machinery in the tumor microenvironment (TME) plays a role in the development of ICI resistance. Within the TME, nutrients and oxygen are scarce, forcing immune cells to undergo metabolic reprogramming to adapt to harsh conditions. Cancer-induced metabolic deregulation in immune cells can attenuate their anti-cancer properties, but can also increase their immunosuppressive properties. Therefore, targeting metabolic pathways of immune cells in the TME may strengthen the efficacy of ICIs and prevent ICI resistance. In this review, we discuss the interactions of immune cells and metabolic alterations in the TME. We also discuss current therapies targeting cellular metabolism in combination with ICIs for the treatment of cancer, and provide possible mechanisms behind the cellular metabolic rewiring that may improve clinical outcomes.
Collapse
Affiliation(s)
- Chao-Yuan Weng
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Cheng-Xiang Kao
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
45
|
Cimino-Mathews A. Novel uses of immunohistochemistry in breast pathology: interpretation and pitfalls. Mod Pathol 2021; 34:62-77. [PMID: 33110239 DOI: 10.1038/s41379-020-00697-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 12/22/2022]
Abstract
Immunohistochemistry is an essential component of diagnostic breast pathology. The emergence of novel assays and applications is accompanied by new interpretation criteria and potential pitfalls. Immunohistochemistry assists in supporting breast origin for primary or metastatic carcinomas and identifying non-mammary metastases to the breast; however, no single immunostain is perfectly sensitive nor specific. GATA3 and Sox10 are particularly useful immunostains to identify triple negative breast carcinoma, which are often negative for other markers of mammary differentiation. Sox10 labeling is a major potential diagnostic pitfall, as Sox10 and S-100 label both triple negative breast carcinoma and metastatic melanoma; a pan-cytokeratin immunostain should always be included for this differential diagnosis. Novel immunohistochemistry serves as surrogates for the molecular alterations unique to several of special-type breast carcinomas, including the use of MYB in adenoid cystic carcinoma, pan-TRK in secretory carcinoma, and mutant IDH2 in tall cell carcinoma with reversed polarity (TCCRP). In addition, PD-L1 immunohistochemistry is an emerging, albeit imperfect, biomarker for breast cancer immunotherapy, with different assay parameters and scoring criteria in breast carcinoma compared to other tumor types. The expanding repertoire of novel immunohistochemistry provides additional diagnostic tools and biomarkers that improve diagnostic breast pathology and patient care.
Collapse
Affiliation(s)
- Ashley Cimino-Mathews
- Department of Pathology and Oncology, The Johns Hopkins University School of Medicine, 401N Broadway St Weinberg Bldg 2242, Baltimore, MD, 21231, USA.
| |
Collapse
|
46
|
Paver EC, Cooper WA, Colebatch AJ, Ferguson PM, Hill SK, Lum T, Shin JS, O'Toole S, Anderson L, Scolyer RA, Gupta R. Programmed death ligand-1 (PD-L1) as a predictive marker for immunotherapy in solid tumours: a guide to immunohistochemistry implementation and interpretation. Pathology 2020; 53:141-156. [PMID: 33388161 DOI: 10.1016/j.pathol.2020.10.007] [Citation(s) in RCA: 153] [Impact Index Per Article: 30.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 10/05/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022]
Abstract
Immunotherapy with checkpoint inhibitors is well established as an effective treatment for non-small cell lung cancer and melanoma. The list of approved indications for treatment with PD-1/PD-L1 checkpoint inhibitors is growing rapidly as clinical trials continue to show their efficacy in patients with a wide range of solid tumours. Clinical trials have used a variety of PD-L1 immunohistochemical assays to evaluate PD-L1 expression on tumour cells, immune cells or both as a potential biomarker to predict response to immunotherapy. Requests to pathologists for PD-L1 testing to guide choice of therapy are rapidly becoming commonplace. Thus, pathologists need to be aware of the different PD-L1 assays, methods of evaluation in different tumour types and the impact of the results on therapeutic decisions. This review discusses the key practical issues relating to the implementation of PD-L1 testing for solid tumours in a pathology laboratory, including evidence for PD-L1 testing, different assay types, the potential interchangeability of PD-L1 antibody clones and staining platforms, scoring criteria for PD-L1, validation, quality assurance, and pitfalls in PD-L1 assessment. This review also explores PD-L1 IHC in solid tumours including non-small cell lung carcinoma, head and neck carcinoma, triple negative breast carcinoma, melanoma, renal cell carcinoma, urothelial carcinoma, gastric and gastroesophageal carcinoma, colorectal carcinoma, hepatocellular carcinoma, and endometrial carcinoma. The review aims to provide pathologists with a practical guide to the implementation and interpretation of PD-L1 testing by immunohistochemistry.
Collapse
Affiliation(s)
- Elizabeth C Paver
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Andrew J Colebatch
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Peter M Ferguson
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Sean K Hill
- Gold Coast University Hospital, Southport, Qld, Australia
| | - Trina Lum
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
| | - Joo-Shik Shin
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Sandra O'Toole
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia
| | - Lyndal Anderson
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Richard A Scolyer
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - Ruta Gupta
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
47
|
Rotman J, den Otter LAS, Bleeker MCG, Samuels SS, Heeren AM, Roemer MGM, Kenter GG, Zijlmans HJMAA, van Trommel NE, de Gruijl TD, Jordanova ES. PD-L1 and PD-L2 Expression in Cervical Cancer: Regulation and Biomarker Potential. Front Immunol 2020; 11:596825. [PMID: 33424844 PMCID: PMC7793653 DOI: 10.3389/fimmu.2020.596825] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Accepted: 11/16/2020] [Indexed: 12/27/2022] Open
Abstract
PD-1/PD-L1 immune checkpoint inhibitors show potential for cervical cancer treatment. However, low response rates suggest that patient selection based on PD-L1 protein expression is not optimal. Here, we evaluated different PD-L1 detection methods and studied transcriptional regulation of PD-L1/PD-L2 expression by The Cancer Genome Atlas (TCGA) mRNAseq analysis. First, we determined the copy number of the PD-L1/PD-L2 locus by fluorescence in situ hybridization (FISH), PD-L1 mRNA expression by RNA in situ hybridization (RNAish), and PD-L1/PD-L2 protein expression by immunohistochemistry (IHC) on tissue microarrays containing a cohort of 60 patients. Additionally, distribution of PD-L1/PD-L2 was visualized based on flow cytometry analysis of single-cell suspensions (n = 10). PD-L1/PD-L2 locus amplification was rare (2%). PD-L1 mRNA expression in tumor cells was detected in 56% of cases, while 41% expressed PD-L1 protein. Discordant scores for PD-L1 protein expression on tumor cells between cores from one patient were observed in 27% of cases. Interestingly, with RNAish, PD-L1 heterogeneity was observed in only 11% of the cases. PD-L2 protein expression was found in 53%. PD-L1 mRNA and protein expression on tumor cells were strongly correlated (p < 0.001). PD-L1 and PD-L2 protein expression showed no correlation on tumor cells (p = 0.837), but a strong correlation on cells in stromal fields (p < 0.001). Co-expression of PD-L1 and PD-L2 on macrophage-like populations was also observed with flow cytometry analysis. Both PD-L1 and PD-L2 TCGA transcript levels strongly correlated in the TCGA data, and both PD-L1 and PD-L2 strongly correlated with interferon gamma (IFNG) expression/transcript levels (p < 0.0001). Importantly, patients with high PD-L1/PD-L2/IFNG transcript levels had a survival advantage over patients with high PD-L1/PD-L2 and low IFNG expression. Based on these findings, we conclude that PD-L1/PD-L2 expression in cervical cancer is mainly associated with interferon induction and not gene amplification, which makes FISH unsuitable as biomarker. The heterogeneous PD-L1 and PD-L2 expression patterns suggest IHC unreliable for patient selection. RNAish, in conjunction with interferon signaling evaluation, seems a promising technique for immune checkpoint detection. These results warrant further investigation into their prognostic and predictive potential.
Collapse
Affiliation(s)
- Jossie Rotman
- Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center (UMC), Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Medical Oncology Amsterdam UMC, Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leontine A S den Otter
- Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center (UMC), Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Maaike C G Bleeker
- Department of Pathology, Cancer Center Amsterdam (CCA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Sanne S Samuels
- Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center (UMC), Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - A Marijne Heeren
- Department of Medical Oncology Amsterdam UMC, Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Margaretha G M Roemer
- Department of Pathology, Cancer Center Amsterdam (CCA), Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Gemma G Kenter
- Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center (UMC), Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Center for Gynecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute-Antoni van Leeuwenhoek (NKI-AVL), Amsterdam, Netherlands
| | - Henry J M A A Zijlmans
- Center for Gynecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute-Antoni van Leeuwenhoek (NKI-AVL), Amsterdam, Netherlands
| | - Nienke E van Trommel
- Center for Gynecologic Oncology Amsterdam (CGOA), Netherlands Cancer Institute-Antoni van Leeuwenhoek (NKI-AVL), Amsterdam, Netherlands
| | - Tanja D de Gruijl
- Department of Medical Oncology Amsterdam UMC, Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Ekaterina S Jordanova
- Center for Gynecologic Oncology Amsterdam (CGOA), Amsterdam University Medical Center (UMC), Cancer Center Amsterdam (CCA), Vrije Universiteit Amsterdam, Amsterdam, Netherlands.,Department of Pathology, Leiden University Medical Center, Leiden, Netherlands
| |
Collapse
|
48
|
Ganbaatar O, Konnai S, Okagawa T, Nojima Y, Maekawa N, Minato E, Kobayashi A, Ando R, Sasaki N, Miyakoshi D, Ichii O, Kato Y, Suzuki Y, Murata S, Ohashi K. PD-L1 expression in equine malignant melanoma and functional effects of PD-L1 blockade. PLoS One 2020; 15:e0234218. [PMID: 33216754 PMCID: PMC7678989 DOI: 10.1371/journal.pone.0234218] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 11/03/2020] [Indexed: 12/31/2022] Open
Abstract
Programmed death-1 (PD-1) is an immunoinhibitory receptor expressed on lymphocytes. Interaction of PD-1 with its ligand PD-ligand 1 (PD-L1) delivers inhibitory signals and impairs proliferation, cytokine production, and cytotoxicity of T cells. In our previous studies, we have developed anti-bovine PD-L1 monoclonal antibodies (mAbs) and reported that the PD-1/PD-L1 pathway was closely associated with T-cell exhaustion and disease progression in bovine chronic infections and canine tumors. Furthermore, we found that blocking antibodies that target PD-1 and PD-L1 restore T-cell functions and could be used in immunotherapy in cattle and dogs. However, the immunological role of the PD-1/PD-L1 pathway for chronic equine diseases, including tumors, remains unclear. In this study, we identified cDNA sequences of equine PD-1 (EqPD-1) and PD-L1 (EqPD-L1) and investigated the role of anti-bovine PD-L1 mAbs against EqPD-L1 using in vitro assays. In addition, we evaluated the expression of PD-L1 in tumor tissues of equine malignant melanoma (EMM). The amino acid sequences of EqPD-1 and EqPD-L1 share a considerable identity and similarity with homologs from non-primate species. Two clones of the anti-bovine PD-L1 mAbs recognized EqPD-L1 in flow cytometry, and one of these cross-reactive mAbs blocked the binding of equine PD-1/PD-L1. Of note, immunohistochemistry confirmed the PD-L1 expression in EMM tumor tissues. A cultivation assay revealed that PD-L1 blockade enhanced the production of Th1 cytokines in equine immune cells. These findings showed that our anti-PD-L1 mAbs would be useful for analyzing the equine PD-1/PD-L1 pathway. Further research is warranted to discover the immunological role of PD-1/PD-L1 in chronic equine diseases and elucidate a future application in immunotherapy for horses.
Collapse
Affiliation(s)
- Otgontuya Ganbaatar
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- * E-mail:
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yutaro Nojima
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Erina Minato
- Department of Veterinary Clinical Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Atsushi Kobayashi
- Department of Veterinary Clinical Medicine, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Ryo Ando
- Laboratory of Veterinary Pathology, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, School of Veterinary Medicine, Kitasato University, Towada, Japan
| | | | - Osamu Ichii
- Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
- New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Yasuhiko Suzuki
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Division of Bioresources, Research Center for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
49
|
Ariyarathna H, Thomson NA, Aberdein D, Perrott MR, Munday JS. Increased programmed death ligand (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) expression is associated with metastasis and poor prognosis in malignant canine mammary gland tumours. Vet Immunol Immunopathol 2020; 230:110142. [PMID: 33129194 DOI: 10.1016/j.vetimm.2020.110142] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 08/04/2020] [Accepted: 10/15/2020] [Indexed: 12/31/2022]
Abstract
Aberrant expression of immune check point molecules, programmed death ligand (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) has been reported in many human cancers with increased protein and gene expression correlated with an aggressive behaviour in some neoplasms. Additionally, PD-L1 blockade has been shown to be an effective therapy for some human cancers. Canine mammary gland tumours have previously been shown to produce PD-L1 protein, but there are no previous studies investigating CTLA-4 in these common canine neoplasms. The present study investigated protein and gene expression of PD-L1 and CTLA-4 using immunohistochemistry and RT-PCR in 41 histologically-malignant, outcome-known CMGTs. The PD-L1 and CTLA-4 immunostaining scores of the mammary gland tumours that subsequently metastasised were significantly higher than those of tumours which did not metastasise (PD-L1: p = 0.005, CTLA-4: p = 0.003). Gene expression of PD-L1 and CTLA-4 was also significantly higher in tumours which subsequently metastasised (PD-L1: p = 0.023, CTLA-4: p = 0.022). Further, higher PD-L1 or CTLA-4 immunostaining scores correlated with shorter survival times of dogs (PD-L1: rs = - 0.42, p = 0.008, CTLA-4: rs = - 0.4, p = 0.01) while PD-L1 immunostaining was independently prognostic of survival time (Δ F = 4.9, p = 0.035). These findings suggest that higher protein and gene expression of PD-L1 and CTLA-4 by tumour cells increases the chances of metastasis and measuring these proteins may predict likely neoplasm behaviour. Additionally, if increased expression of these proteins promotes metastasis, blocking PD-L1 or CTLA-4 may be beneficial to treat canine mammary gland tumours.
Collapse
Affiliation(s)
- Harsha Ariyarathna
- Department of Pathobiology, School of Veterinary Science, Massey University, Private Bag 11 222, Palmerston North, New Zealand.
| | - Neroli A Thomson
- Department of Pathobiology, School of Veterinary Science, Massey University, Private Bag 11 222, Palmerston North, New Zealand.
| | - Danielle Aberdein
- Department of Pathobiology, School of Veterinary Science, Massey University, Private Bag 11 222, Palmerston North, New Zealand.
| | - Matthew R Perrott
- Department of Pathobiology, School of Veterinary Science, Massey University, Private Bag 11 222, Palmerston North, New Zealand.
| | - John S Munday
- Department of Pathobiology, School of Veterinary Science, Massey University, Private Bag 11 222, Palmerston North, New Zealand.
| |
Collapse
|
50
|
Girolami I, Pantanowitz L, Munari E, Martini M, Nocini R, Bisi N, Molteni G, Marchioni D, Ghimenton C, Brunelli M, Eccher A. Prevalence of PD-L1 expression in head and neck squamous precancerous lesions: a systematic review and meta-analysis. Head Neck 2020; 42:3018-3030. [PMID: 32567746 DOI: 10.1002/hed.26339] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/23/2020] [Accepted: 05/30/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Studies concerning programmed death-ligand 1 (PD-L1) expression in precancerous lesions of head and neck (HN) region have shown variable results. METHODS We systematically reviewed the published evidence on PD-L1 expression in HN precancerous lesions. RESULTS Of 1058 original articles, 14 were included in systematic review and 9 in meta-analysis. The pooled estimate of PD-L1 expression was 48.25% (confidence interval [CI] 21.07-75.98, I2 98%, tau2 0.18). PD-L1 expression appeared to be more frequent in precancerous lesions than in normal mucosa (risk ratio [RR] 1.65, CI 0.65-4.03, I2 91%, tau2 0.82) and less frequent than in invasive squamous cell carcinoma (RR 0.68, CI 0.43-1.08, I2 91%, tau2 0.22). CONCLUSIONS PD-L1 expression could reflect a point of balance between host immune response and cancer escape ability. High heterogeneity and moderate quality suggest that further studies with larger sample size and more rigorous case selection will allow more precise assessment of PD-L1 expression in HN precancerous lesions.
Collapse
Affiliation(s)
- Ilaria Girolami
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Enrico Munari
- Department of Pathology, Sacro Cuore Don Calabria Hospital, Italy
| | - Maurizio Martini
- Division of Anatomic Pathology and Histology, Catholic University-Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Riccardo Nocini
- Department of Otorhinolaringology and Head & Neck Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Nicola Bisi
- Department of Otorhinolaringology and Head & Neck Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Gabriele Molteni
- Department of Otorhinolaringology and Head & Neck Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Daniele Marchioni
- Department of Otorhinolaringology and Head & Neck Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Ghimenton
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|