1
|
Santos AS, Santos-Bezerra DP, Ferreira LRP, Bando SY, Alves LI, Cunha-Neto E, da Silva MER. Relevance of Circulating microRNA, and their Association with Islet Cell Autoantibodies in Type 1 Diabetes Pathogenesis. Arch Med Res 2024; 56:103114. [PMID: 39489115 DOI: 10.1016/j.arcmed.2024.103114] [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: 05/30/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND AIMS/HYPOTHESIS The role of microRNAs (miRNAs) in the pathogenesis and progression of type 1 diabetes (T1D) has been described, but data remain scarce and conflicting. OBJECTIVES To evaluate the potential biological involvement of miRNA expression in the immune response and beta cell function in T1D. METHODS We screened 10 serum miRNAs from 142 subjects divided into three groups: healthy individuals (control group; n = 52) and patients at different stages of T1D progression, from the initial immunological manifestation, presenting islet cell autoantibodies (AbP group; n = 39), to partial and severe beta cell damage in T1D (recent T1D group; n = 51). RESULTS Three miRNAs (miR-200c-3p, miR-301a-3p, and miR-382-5p) were highly expressed in the AbP and/or recent T1D groups compared to the control group. Furthermore, in the AbP group, miR-301a-3p and miR-382-5p were positively correlated with insulin autoantibody levels and miR-382-5p was negatively correlated with C-peptide levels. In the recent T1D group, miR-200c-3p expression was positively correlated with IA-2A levels. Enrichment analysis of differentially expressed miRNAs showed their involvement in immune response, inflammatory pathways, proliferation/survival/apoptosis mechanisms, bacterial and viral infection, and insulin resistance. CONCLUSION Our data indicated that miR-200c-3p, miR-301a-3p, and miR-382-5p might be involved in T1D pathogenesis. Proliferative, metabolic, and immune responses were main pathways associated with serum miRNA target genes.
Collapse
Affiliation(s)
- Aritania S Santos
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | | | - Ludmila Rodrigues Pinto Ferreira
- Department of Morphology, RNA Systems Biology Laboratory, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Silvia Y Bando
- Department of Pediatrics, Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Laís Isidoro Alves
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - Edecio Cunha-Neto
- Laboratory of Immunology, Heart Institute, School of Medicine, University of São Paulo, Institute for Investigation in Immunology (iii) INCT, São Paulo, Brazil
| | - Maria Elizabeth Rossi da Silva
- Laboratorio de Carboidratos e Radioimunoensaios, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil; Hospital das Clinicas, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil.
| |
Collapse
|
2
|
Kotwal A, Kennedy R, Kikani N, Thosani S, Goldner W, Shariff A. Endocrinopathies Associated With Immune Checkpoint Inhibitor Use. Endocr Pract 2024; 30:584-591. [PMID: 38554775 DOI: 10.1016/j.eprac.2024.03.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 03/12/2024] [Accepted: 03/20/2024] [Indexed: 04/02/2024]
Abstract
OBJECTIVE To provide a clinical approach towards immune checkpoint inhibitor (ICI)-associated endocrinopathies, their link with cancer outcomes, factors which differentiate them from other immune related adverse events, and health systems innovation to improve care for these patients. METHODS A literature search for articles pertaining to ICIs and endocrinopathies was performed and supplemented by expert opinions of the authors. RESULTS While immune related adverse events can affect almost any organ, they frequently target the endocrine glands, most commonly thyroid. Different classes of ICIs have varying frequencies of endocrinopathies related to hypophysitis, thyroiditis, diabetes mellitus, and rarely hypoadrenalism and hypoparathyroidism. ICI-associated endocrinopathies share some features with classic endocrine autoimmunity but appear to be a distinct entity. They can be challenging to diagnose and manage due to nonspecific clinical features, use of exogenous glucocorticoids, and at times rapid and severe hormone deficiency. The role of anti-inflammatory high-dose glucocorticoids is minimal, and the ICI does not usually require permanent discontinuation. ICI-associated endocrinopathies usually cause permanent hormone deficiency necessitating long-term management and patient engagement. ICI-thyroiditis has been associated with improved survival, while other endocrinopathies have not shown a significant association with outcomes in cancer patients receiving ICIs. Oncoendocrinology teams can improve the care of patients with ICI-associated endocrinopathies. CONCLUSION This narrative review provides guidance to clinicians prescribing ICIs and those managing ICI-associated endocrinopathies, and complements the frameworks provided by major scientific societies in this field.
Collapse
Affiliation(s)
- Anupam Kotwal
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska.
| | - Randol Kennedy
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina
| | - Nupur Kikani
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Sonali Thosani
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Whitney Goldner
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, University of Nebraska Medical Center, Omaha, Nebraska; Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska
| | - Afreen Shariff
- Division of Endocrinology, Metabolism, and Nutrition, Department of Medicine, Duke University School of Medicine, Durham, North Carolina; Duke Cancer Institute, Duke Health, Durham, North Carolina
| |
Collapse
|
3
|
Zieliński M, Sakowska J, Iwaszkiewicz-Grześ D, Gliwiński M, Hennig M, Żalińska M, Wołoszyn-Durkiewicz A, Jaźwińska-Curyłło A, Kamińska H, Owczuk R, Młynarski W, Jarosz-Chobot P, Bossowski A, Szadkowska A, Fendler W, Beń-Skowronek I, Chobot A, Myśliwiec M, Siebert J, Marek-Trzonkowska N, Trzonkowski P. PD-1 Receptor (+) T cells are associated with the efficacy of the combined treatment with regulatory t cells and rituximab in type 1 diabetes children via regulatory t cells suppressive activity amelioration. Int Immunopharmacol 2024; 132:111919. [PMID: 38554443 DOI: 10.1016/j.intimp.2024.111919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/01/2024]
Abstract
An imbalance between exaggerated autoaggressive T cell responses, primarily CD8 + T cells, and impaired tolerogenic mechanisms underlie the development of type 1 diabetes mellitus. Disease-modifying strategies, particularly immunotherapy focusing on FoxP3 + T regulatory cells (Treg), and B cells facilitating antigen presentation for T cells, show promise. Selective depletion of B cells may be achieved with an anti-CD20 monoclonal antibody (mAb). In a 2-year-long flow cytometry follow-up, involving 32 peripheral blood T and B cell markers across three trial arms (Treg + rituximab N = 12, Treg + placebo N = 13, control N = 11), we observed significant changes. PD-1 receptor (+) CD4 + Treg, CD4 + effector T cells (Teffs), and CD8 + T cell percentages increased in the combined regimen group by the end of follow-up. Conversely, the control group exhibited a notable reduction in PD-1 receptor (+) CD4 + Teff percentages. Considering clinical endpoints, higher PD-1 receptor (+) expression on T cells correlated with positive responses, including a higher mixed meal tolerance test AUC, and reduced daily insulin dosage. PD-1 receptor (+) T cells emerged as a potential therapy outcome biomarker. In vitro validation confirmed that successful Teff suppression was associated with elevated PD-1 receptor (+) Treg levels. These findings support PD-1 receptor (+) T cells as a reliable indicator of treatment with combined immunotherapy consisting of Tregs and anti-CD20 mAb efficacy in type 1 diabetes mellitus.
Collapse
Affiliation(s)
- Maciej Zieliński
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7 80-210, Poland; Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland
| | - Justyna Sakowska
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7 80-210, Poland; Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland
| | - Dorota Iwaszkiewicz-Grześ
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7 80-210, Poland; Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland
| | - Mateusz Gliwiński
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7 80-210, Poland; Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland
| | - Matylda Hennig
- Department of Pediatric Diabetology and Endocrinology, Medical University of Gdańsk, Debinki 7 80-210, Poland
| | - Magdalena Żalińska
- Department of Pediatric Diabetology and Endocrinology, Medical University of Gdańsk, Debinki 7 80-210, Poland
| | - Anna Wołoszyn-Durkiewicz
- Department of Pediatric Diabetology and Endocrinology, Medical University of Gdańsk, Debinki 7 80-210, Poland
| | - Anna Jaźwińska-Curyłło
- Regional Center of Blood Donation and Treatment, Hoene-Wrońskiego 4, 80-210 Gdańsk, Poland
| | - Halla Kamińska
- Department of Children's Diabetology, Medical University of Silesia, Medykow 16, 40-752 Katowice, Poland
| | - Radosław Owczuk
- Department of Anaesthesiology and Critical Care, Medical University of Gdańsk, Debinki 7 80-210, Poland
| | - Wojciech Młynarski
- Department of Paediatrics, Oncology and Haematology, Medical University of Lodz, Sporna 36/50, 91-738 Lodz, Poland
| | - Przemysława Jarosz-Chobot
- Department of Children's Diabetology, Medical University of Silesia, Medykow 16, 40-752 Katowice, Poland
| | - Artur Bossowski
- Department of Peadiatrics, Endocrinology, Diabetology with Cardiology Division, Medical University of Bialystok, Jana Kilińskiego 1, 15-089 Białystok, Poland
| | - Agnieszka Szadkowska
- Department of Pediatrics, Diabetology, Endocrinology and Nephrology, Medical University of Lodz, Sporna 36/50, 91-738 Lodz, Poland
| | - Wojciech Fendler
- Department of Pediatrics, Diabetology, Endocrinology and Nephrology, Medical University of Lodz, Sporna 36/50, 91-738 Lodz, Poland
| | - Iwona Beń-Skowronek
- Dept. Pediatric Endocrinology and Diabetology, Medical University of Lublin, ul. Prof. A. Gebali 6, 20-093 Lublin, Poland
| | - Agata Chobot
- Department of Paediatrics, Institute of Medical Sciences, University of Opole, Al. Witosa 26, 45-401 Opole, Poland
| | - Małgorzata Myśliwiec
- Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland; Department of Pediatric Diabetology and Endocrinology, Medical University of Gdańsk, Debinki 7 80-210, Poland
| | - Janusz Siebert
- Department of Family Medicine, Laboratory of Immunoregulation and Cellular Therapies, Medical University of Gdańsk, Debinki 2 80-210, Poland
| | - Natalia Marek-Trzonkowska
- Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland; Department of Family Medicine, Laboratory of Immunoregulation and Cellular Therapies, Medical University of Gdańsk, Debinki 2 80-210, Poland; International Centre for Cancer Vaccine Science, University of Gdańsk, Wita Stwosza 63, 80-308 Gdańsk, Poland
| | - Piotr Trzonkowski
- Department of Medical Immunology, Medical University of Gdańsk, Debinki 7 80-210, Poland; Poltreg S.A., Botaniczna 20 Street, 80-298 Gdańsk, Poland.
| |
Collapse
|
4
|
Cao Y, Afzal MZ, Gutmann EJ, Shirai K. Rare immune-related adverse events in a patient with metastatic melanoma: a case report highlighting sarcoidosis-like reactions triggered by immune-checkpoint inhibitors. Melanoma Res 2024; 34:70-75. [PMID: 37830935 DOI: 10.1097/cmr.0000000000000925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Pembrolizumab and ipilimumab/nivolumab (ipi/nivo) combination are FDA-approved immune checkpoint inhibitor (ICI) therapies for metastatic melanoma. ICIs could result in various inflammation responses known as immune-related adverse events (IRAEs). We report a patient with metastatic melanoma who developed multiple IRAEs including sarcoidosis-like reaction (SLR), diabetic ketoacidosis (DKA), and worsening hypothyroidism on ICIs. A 71-year-old man with stage IIIC melanoma and lymph node metastasis began adjuvant therapy with pembrolizumab in May 2021. A surveillance positron emission tomography-computed tomography (PET-CT) scan four months later showed diffuse nodal uptake indicating potential metastases although the patient remained asymptomatic. His treatment was temporarily switched to ipi/nivo before biopsy was obtained for definitive diagnosis, which revealed non-caseating granulomas consistent with SLR. After resuming pembrolizumab, he developed DKA and worsening hypothyroidism in November 2021, both of which were attributed to IRAEs. His surveillance PET scan in March 2022 again revealed new hypermetabolic activity in several bones, subcutaneous tissue, and the left inguinal lymph node. Left inguinal node biopsy showed disease recurrence, while biopsies of hypermetabolic subcutaneous nodules and bone demonstrated non-caseating granulomas. Our case described a patient on ICIs who developed several IRAEs. SLR is often asymptomatic but remains a diagnostic challenge due to its indistinguishable appearance on imaging studies compared to metastasis. Better understanding of IRAEs and improved surveillance strategies are needed for optimal patient outcomes.
Collapse
Affiliation(s)
- Yuanzhen Cao
- Department of Internal Medicine, Dartmouth-Hitchcock Medical Center
| | - Muhammad Zubair Afzal
- Section of Hematology/Oncology, Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center
| | - Edward J Gutmann
- Department of Pathology, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Keisuke Shirai
- Section of Hematology/Oncology, Dartmouth Cancer Center, Dartmouth-Hitchcock Medical Center
| |
Collapse
|
5
|
Bhanderi H, Khalid F, Bodla ZH, Muhammad T, Du D, Meghal T. Autoimmune diabetes from pembrolizumab: A case report and review of literature. World J Clin Oncol 2023; 14:535-543. [PMID: 38059185 PMCID: PMC10696214 DOI: 10.5306/wjco.v14.i11.535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/13/2023] [Accepted: 10/30/2023] [Indexed: 11/22/2023] Open
Abstract
BACKGROUND Immunotherapy, specifically the use of checkpoint inhibitors such as pembrolizumab, has become an important tool in personalized cancer therapy. These inhibitors target proteins on T-cells that regulate the immune response against tumor cells. Pembrolizumab, which targets the programmed cell death 1 receptor on T-cells, has been approved for the treatment of metastatic melanoma and non-small cell lung cancer. However, it can also lead to immune-related side effects, including pneumonitis, colitis, thyroid abnormalities, and rare cases of type 1 diabetes. CASE SUMMARY The case presented involves an adult patient in 30s with breast cancer who developed hyperglycemia after receiving pembrolizumab treatment. The patient was diagnosed with diabetic ketoacidosis and further investigations were performed to evaluate for new-onset type 1 diabetes. The patient had a history of hypothyroidism and a family history of breast cancer. Treatment for diabetic ketoacidosis was initiated, and the patient was discharged for close follow-up with an endocrinologist. CONCLUSION This literature review highlights the occurrence of diabetic ketoacidosis and new-onset type 1 diabetes in patients receiving pembrolizumab treatment for different types of cancer. Overall, the article emphasizes the therapeutic benefits of immunotherapy in cancer treatment, particularly pembrolizumab, while also highlighting the potential side effect of immune-related diabetes that can occur in a small percentage of patients. Here we present a case where pembrolizumab lead to development of diabetes after a few cycles highlighting one of the rare yet a serious toxicity of the drug.
Collapse
Affiliation(s)
- Hardikkumar Bhanderi
- Department of Internal Medicine, Monmouth Medical Center, Long branch, NJ 07740, United States
| | - Farhan Khalid
- Department of Internal Medicine, Monmouth Medical Center, Long branch, NJ 07740, United States
| | - Zubair Hassan Bodla
- Department of Internal Medicine, University of Central Florida College of Medicine, Gainesville, FL 32303, United States
| | - Tayyeb Muhammad
- Department of Internal Medicine, Monmouth Medical Center, Long branch, NJ 07740, United States
| | - Doantrang Du
- Department of Internal Medicine, Monmouth Medical Center, Long branch, NJ 07740, United States
| | - Trishala Meghal
- Department of Hematology-Oncology, Monmouth Medical Center, Long Branch, NJ 07740, United States
| |
Collapse
|
6
|
Collier JL, Pauken KE, Lee CA, Patterson DG, Markson SC, Conway TS, Fung ME, France JA, Mucciarone KN, Lian CG, Murphy GF, Sharpe AH. Single-cell profiling reveals unique features of diabetogenic T cells in anti-PD-1-induced type 1 diabetes mice. J Exp Med 2023; 220:e20221920. [PMID: 37432393 PMCID: PMC10336233 DOI: 10.1084/jem.20221920] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 04/28/2023] [Accepted: 06/23/2023] [Indexed: 07/12/2023] Open
Abstract
Immune-related adverse events (irAEs) are a notable complication of PD-1 cancer immunotherapy. A better understanding of how these iatrogenic diseases compare with naturally arising autoimmune diseases is needed for treatment and monitoring of irAEs. We identified differences in anti-PD-1-induced type 1 diabetes (T1D) and spontaneous T1D in non-obese diabetic (NOD) mice by performing single-cell RNA-seq and TCR-seq on T cells from the pancreas, pancreas-draining lymph node (pLN), and blood of mice with PD-1-induced T1D or spontaneous T1D. In the pancreas, anti-PD-1 resulted in expansion of terminally exhausted/effector-like CD8+ T cells, an increase in T-bethi CD4+FoxP3- T cells, and a decrease in memory CD4+FoxP3- and CD8+ T cells in contrast to spontaneous T1D. Notably, anti-PD-1 caused increased TCR sharing between the pancreas and the periphery. Moreover, T cells in the blood of anti-PD-1-treated mice expressed markers that differed from spontaneous T1D, suggesting that the blood may provide a window to monitor irAEs rather than relying exclusively on the autoimmune target organ.
Collapse
Affiliation(s)
- Jenna L. Collier
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Kristen E. Pauken
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Dillon G. Patterson
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Samuel C. Markson
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Thomas S. Conway
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Megan E. Fung
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | - Joshua A. France
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
| | | | - Christine G. Lian
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - George F. Murphy
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
| | - Arlene H. Sharpe
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA, USA
- Evergrande Center for Immunological Diseases, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA, USA
- Department of Pathology, Brigham and Women’s Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
7
|
Rezayi M, Hosseini A. Structure of PD1 and its mechanism in the treatment of autoimmune diseases. Cell Biochem Funct 2023; 41:726-737. [PMID: 37475518 DOI: 10.1002/cbf.3827] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/27/2023] [Accepted: 07/08/2023] [Indexed: 07/22/2023]
Abstract
PD-1 and CTLA-4 can play an important role in addressing the issue of autoimmune diseases. PD-1 is a transmembrane glycoprotein expressed on T, B, and Dentric cells. This molecule functions as a checkpoint in T cell proliferation. Ligation of PD-1 with its ligands inhibits the production of IL-2, IL-7, IL-10, and IL-12 as well as other cytokines by macrophages, natural killer (NK) cells, and T cells, which can suppress cell proliferation and inflammation. Today, scientists attempt to protect against autoimmune diseases by PD-1 inhibitory signals. In this review, we discuss the structure, expression, and signaling pathway of PD-1. In addition, we discuss the importance of PD-1 in regulating several autoimmune diseases, reflecting how manipulating this molecule can be an effective method in the immunotherapy of some autoimmune diseases.
Collapse
Affiliation(s)
- Mahdi Rezayi
- Department of Medical Sciences, Marand Baranch, Islamic Azad University, Marand, Iran
| | - Arezoo Hosseini
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
8
|
Liao D, Liu C, Chen S, Liu F, Li W, Shangguan D, Shi Y. Recent advances in immune checkpoint inhibitor-induced type 1 diabetes mellitus. Int Immunopharmacol 2023; 122:110414. [PMID: 37390646 DOI: 10.1016/j.intimp.2023.110414] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/23/2023] [Accepted: 05/29/2023] [Indexed: 07/02/2023]
Abstract
As a new group of anticancer drugs, immune checkpoint inhibitors (ICIs) have exhibited favorable antitumor efficacy in numerous malignant tumors. Anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4), anti-programmed cell death-1 (PD-1) and anti-programmed cell death ligand-1 (PD-L1) are three kinds of ICIs widely used in clinical practice. However, ICI therapy (monotherapy or combination therapy) is always accompanied by a unique toxicity profile known as immune-related adverse events (irAEs) affecting multiple organs. The endocrine glands are common targets of irAEs induced by ICIs, which cause type 1 diabetes mellitus (T1DM) when the pancreas is affected. Although the incidence rate of ICI-induced T1DM is rare, it will always lead to an irreversible impairment of β-cells and be potentially life-threatening. Hence, it is vital for endocrinologists and oncologists to obtain a comprehensive understanding of ICI-induced T1DM and its management. In our present manuscript, we have reviewed the epidemiology, pathology and mechanism, diagnosis, management, and treatments of ICI-induced T1DM.
Collapse
Affiliation(s)
- Dehua Liao
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Chaoyi Liu
- Department of Information, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Shanshan Chen
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Fen Liu
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Wei Li
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China
| | - Dangang Shangguan
- Department of Pharmacy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China.
| | - Yingrui Shi
- Department of Radiation Oncology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410011, China.
| |
Collapse
|
9
|
Bode K, MacDonald T, Stewart T, Mendez B, Cai EP, Morrow N, Lee YC, Yi P, Kissler S. Protective Renalase Deficiency in β-Cells Shapes Immune Metabolism and Function in Autoimmune Diabetes. Diabetes 2023; 72:1127-1143. [PMID: 37216639 PMCID: PMC10382656 DOI: 10.2337/db23-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 04/30/2023] [Indexed: 05/24/2023]
Abstract
Type 1 diabetes (T1D) is caused by the immune-mediated loss of pancreatic β-cells that produce insulin. The latest advances in stem cell (SC) β-cell differentiation methods have made a cell replacement therapy for T1D feasible. However, recurring autoimmunity would rapidly destroy transplanted SC β-cells. A promising strategy to overcome immune rejection is to genetically engineer SC β-cells. We previously identified Renalase (Rnls) as a novel target for β-cell protection. Here we show that Rnls deletion endows β-cells with the capacity to modulate the metabolism and function of immune cells within the local graft microenvironment. We used flow cytometry and single-cell RNA sequencing to characterize β-cell graft-infiltrating immune cells in a mouse model for T1D. Loss of Rnls within transplanted β-cells affected both the composition and the transcriptional profile of infiltrating immune cells in favor of an anti-inflammatory profile with decreased antigen-presenting capacity. We propose that changes in β-cell metabolism mediate local immune regulation and that this feature could be exploited for therapeutic goals. ARTICLE HIGHLIGHTS Protective Renalase (Rnls) deficiency impacts β-cell metabolism. Rnls-deficient β-cell grafts do not exclude immune infiltration. Rnls deficiency in transplanted β-cells broadly modifies local immune function. Immune cell in Rnls mutant β-cell grafts adopt a noninflammatory phenotype.
Collapse
Affiliation(s)
- Kevin Bode
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Tara MacDonald
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Taylor Stewart
- Department of Medicine, Harvard Medical School, Boston, MA
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Bryhan Mendez
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA
| | - Erica P. Cai
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Noelle Morrow
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Yu-Chi Lee
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
| | - Peng Yi
- Department of Medicine, Harvard Medical School, Boston, MA
- Section for Islet Cell and Regenerative Biology, Joslin Diabetes Center, Boston, MA
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA
| | - Stephan Kissler
- Section for Immunobiology, Joslin Diabetes Center, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Diabetes Program, Harvard Stem Cell Institute, Cambridge, MA
| |
Collapse
|
10
|
Kotwal A, Perlman JE, Goldner WS, Marr A, Mammen JS. Endocrine Dysfunction From Immune Checkpoint Inhibitors: Pearls and Pitfalls in Evaluation and Management. JCO Oncol Pract 2023:OP2300023. [PMID: 37023383 DOI: 10.1200/op.23.00023] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023] Open
Abstract
Immune checkpoint inhibitors (ICPis) have proven extremely efficacious in cancer therapy but also lead to a plethora of immune-related adverse events (irAEs). The endocrine irAEs are not only quite common but also may pose a challenge to the clinician while managing a patient with cancer treated with ICPis. The clinical features of endocrine dysfunction are usually nonspecific and may overlap with concurrent illnesses, underlying the importance of accurate hormone testing and efforts toward case-finding. The management of endocrine irAEs is unique in the focus being on hormone replacement rather than curtailing the autoimmune process. Although the management of thyroid irAEs appears straightforward, adrenal insufficiency and insulin-dependent diabetes can be life-threatening if not promptly recognized and treated. This clinical review synthesizes the studies to provide pearls and pitfalls in the evaluation and management of endocrine irAEs with specific reference to guidelines from oncologic societies.
Collapse
Affiliation(s)
- Anupam Kotwal
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Jordan E Perlman
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University Medical Center, Baltimore, MD
| | - Whitney S Goldner
- Division of Diabetes, Endocrinology, and Metabolism, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Alissa Marr
- Division of Oncology and Hematology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE
| | - Jennifer S Mammen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Johns Hopkins University Medical Center, Baltimore, MD
| |
Collapse
|
11
|
Pancreatic Islet Cells Response to IFNγ Relies on Their Spatial Location within an Islet. Cells 2022; 12:cells12010113. [PMID: 36611907 PMCID: PMC9818682 DOI: 10.3390/cells12010113] [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: 11/27/2022] [Revised: 12/19/2022] [Accepted: 12/23/2022] [Indexed: 12/30/2022] Open
Abstract
Type 1 diabetes (T1D) is an auto-immune disease characterized by the progressive destruction of insulin-producing pancreatic beta cells. While beta cells are the target of the immune attack, the other islet endocrine cells, namely the alpha and delta cells, can also be affected by the inflammatory milieu. Here, using a flow cytometry-based strategy, we compared the impact of IFNγ, one of the main cytokines involved in T1D, on the three endocrine cell subsets isolated from C57BL/6 mouse islets. RNA-seq analyses revealed that alpha and delta cells exposed in vitro to IFNγ display a transcriptomic profile very similar to that of beta cells, with an increased expression of inflammation key genes such as MHC class I molecules, the CXCL10 chemokine and the programmed death-ligand 1 (PD-L1), three hallmarks of IFNγ signaling. Interestingly, at low IFNγ concentration, we observed two beta cell populations (responders and non-responders) based on PD-L1 protein expression. Our data indicate that this differential sensitivity relies on the location of the cells within the islet rather than on the existence of two different beta cells subsets. The same findings were corroborated by the in vivo analysis of pancreatic islets from the non-obese diabetic mouse model of T1D, showing more intense PD-L1 staining on endocrine cells close to immune infiltrate. Collectively, our work demonstrates that alpha and delta cells are as sensitive as beta cells to IFNγ, and suggests a gradual diffusion of the cytokine into an islet. These observations provide novel insights into the in situ inflammatory processes occurring in T1D progression.
Collapse
|
12
|
Ikeda M, Tamada T, Takebayashi R, Okuno G, Yagura I, Nakamori S, Matsumura T, Yoshioka T, Kaneko S, Kanda N. Development of Fulminant Type 1 Diabetes Mellitus in the Course of Treatment with Atezolizumab for Hepatocellular Carcinoma: A Case Report. Intern Med 2022. [PMID: 36288990 DOI: 10.2169/internalmedicine.0860-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/07/2023] Open
Abstract
A 71-year-old woman with recurring stage IV hepatocellular carcinoma (HCC) was admitted to our hospital while being treated with atezolizumab and bevacizumab and complained of fatigue, vomiting, and appetite loss. The following were noted on admission: serum glucose level, 633 mg/dL; metabolic acidemia (HCO3- of 19.5 mmol/L); remarkably low serum and urinary C-peptide levels (0.16 ng/mL and ≤1.5 μg/day, respectively); and urinary ketone body level, 4,197 μmol/L. She was diagnosed with atezolizumab-induced fulminant type 1 diabetes mellitus (T1DM), and insulin therapy improved the symptoms. To our knowledge, this a novel report of atezolizumab-induced fulminant T1DM in an HCC patient.
Collapse
Affiliation(s)
- Munehiro Ikeda
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Takashi Tamada
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Risa Takebayashi
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Gaku Okuno
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Iori Yagura
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Shohei Nakamori
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Taishiro Matsumura
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Takuto Yoshioka
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| | - Shizuka Kaneko
- Department of Diabetes/ Endocrinology/ Metabolism, Takatsuki Red Cross Hospital, Japan
| | - Naoki Kanda
- Department of Gastroenterology and Hepatology, Takatsuki Red Cross Hospital, Japan
| |
Collapse
|
13
|
Califaretti E, Dall'armellina S, Rovera G, Finessi M, Deandreis D. The role of PET/CT in thyroid autoimmune diseases. THE QUARTERLY JOURNAL OF NUCLEAR MEDICINE AND MOLECULAR IMAGING : OFFICIAL PUBLICATION OF THE ITALIAN ASSOCIATION OF NUCLEAR MEDICINE (AIMN) [AND] THE INTERNATIONAL ASSOCIATION OF RADIOPHARMACOLOGY (IAR), [AND] SECTION OF THE SOCIETY OF... 2022; 66:218-228. [PMID: 35612371 DOI: 10.23736/s1824-4785.22.03464-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Autoimmune thyroid diseases (AITD) are a heterogeneous group of disorders. They include, in particular, Graves' disease and Hashimoto's thyroiditis with a wide range of different functional status ranging from subclinical biochemical abnormalities to severe hyperthyroidism or severe hypothyroidism respectively. Furthermore, other conditions more frequently infectious or drug related can cause an immune reaction in the thyroid tissue. In AITDs, positron emission tomography/computed tomography (PET/CT) does not play a primary role for disease diagnosis or management, but accidental findings can occur in both symptomatic and asymptomatic patients, and they should be recognized and well interpreted. A comprehensive literature search of the PubMed databases was conducted to identify papers (systematic review, prospective and retrospective study, case report) evaluating the role of PET/CT in thyroid autoimmune diseases. Thyroid diffuse uptake of 18F-fluoro-2-deoxy-2-d-glucose ([18F]FDG) has been shown to be frequently associated with AITDs, but also with immune-induced thyroid disorders related to SARS-CoV-2 or immunotherapy, while malignant lesions more often have a focal aspect. Other radiopharmaceuticals as [68Ga]-DOTA-peptides, [68Ga]-fibroblast activation protein inhibitors (FAPIs) and [68Ga]-prostate specific membrane antigen ([68Ga]-PSMA) showed similar findings. In conclusion, PET/CT scan in AITDs does not play a primary role in the diagnosis, but the occasional finding of a thyroid uptake must always be described in the report and possibly investigated for a better patient's management.
Collapse
Affiliation(s)
- Elena Califaretti
- Unit of Nuclear Medicine, Department of Medical Sciences, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Sara Dall'armellina
- Unit of Nuclear Medicine, Department of Medical Sciences, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Guido Rovera
- Unit of Nuclear Medicine, Department of Medical Sciences, Città della Salute e della Scienza, University of Turin, Turin, Italy
| | - Monica Finessi
- Unit of Nuclear Medicine, Department of Diagnostic Imaging and Interventional Radiology, Città della Salute e della Scienza, Turin, Italy -
| | - Désirée Deandreis
- Unit of Nuclear Medicine, Department of Medical Sciences, Città della Salute e della Scienza, University of Turin, Turin, Italy
- Unit of Nuclear Medicine, Department of Diagnostic Imaging and Interventional Radiology, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
14
|
Beenen AC, Sauerer T, Schaft N, Dörrie J. Beyond Cancer: Regulation and Function of PD-L1 in Health and Immune-Related Diseases. Int J Mol Sci 2022; 23:ijms23158599. [PMID: 35955729 PMCID: PMC9369208 DOI: 10.3390/ijms23158599] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 07/28/2022] [Accepted: 07/29/2022] [Indexed: 12/20/2022] Open
Abstract
Programmed Cell Death 1 Ligand 1 (PD-L1, CD274, B7-H1) is a transmembrane protein which is strongly involved in immune modulation, serving as checkpoint regulator. Interaction with its receptor, Programmed Cell Death Protein 1 (PD-1), induces an immune-suppressive signal, which modulates the activity of T cells and other effector cells. This mediates peripheral tolerance and contributes to tumor immune escape. PD-L1 became famous due to its deployment in cancer therapy, where blockage of PD-L1 with the help of therapeutic antagonistic antibodies achieved impressive clinical responses by reactivating effector cell functions against tumor cells. Therefore, in the past, the focus has been placed on PD-L1 expression and its function in various malignant cells, whereas its role in healthy tissue and diseases apart from cancer remained largely neglected. In this review, we summarize the function of PD-L1 in non-cancerous cells, outlining its discovery and origin, as well as its involvement in different cellular and immune-related processes. We provide an overview of transcriptional and translational regulation, and expression patterns of PD-L1 in different cells and organs, and illuminate the involvement of PD-L1 in different autoimmune diseases as well as in the context of transplantation and pregnancy.
Collapse
Affiliation(s)
- Amke C. Beenen
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (A.C.B.); (T.S.); (N.S.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Tatjana Sauerer
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (A.C.B.); (T.S.); (N.S.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (A.C.B.); (T.S.); (N.S.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
| | - Jan Dörrie
- Department of Dermatology, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Hartmannstraße 14, 91052 Erlangen, Germany; (A.C.B.); (T.S.); (N.S.)
- Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Östliche Stadtmauerstraße 30, 91054 Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Ulmenweg 18, 91054 Erlangen, Germany
- Correspondence: ; Tel.: +49-9131-85-31127
| |
Collapse
|
15
|
Zhu B, Qu S. The Relationship Between Diabetes Mellitus and Cancers and Its Underlying Mechanisms. Front Endocrinol (Lausanne) 2022; 13:800995. [PMID: 35222270 PMCID: PMC8873103 DOI: 10.3389/fendo.2022.800995] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 01/12/2022] [Indexed: 12/27/2022] Open
Abstract
Epidemiological studies suggest associations between diabetes mellitus and some cancers. The risk of a number of cancers appears to be increased in diabetes mellitus. On the other hand, some cancer and cancer therapies could lead to diabetes mellitus. Genetic factors, obesity, inflammation, oxidative stress, hyperglycemia, hyperinsulinemia, cancer therapies, insulin and some oral hypoglycemic drugs appear to play a role in the crosstalk between diabetes mellitus and cancers. This review summarized the associations between various types of diabetes and cancers and updated available evidence of underlying mechanisms between diabetes and cancers.
Collapse
Affiliation(s)
| | - Shen Qu
- Department of Endocrinology and Metabolism, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
16
|
[Immune Checkpoint Inhibitors Related Diabetes Mellitus:
A Report of 2 Cases and Literature Review]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2022; 25:61-65. [PMID: 35078286 PMCID: PMC8796132 DOI: 10.3779/j.issn.1009-3419.2021.102.54] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in clinic, and the incidence of rare adverse events are increasing. The aim of this paper is to better define the rare adverse effect of diabetes mellitus associated with ICIs. We report 2 cases of diabetes mellitus associated with ICIs. Literature review was conducted and we discussed the clinical presentation, potential mechanisms and suggestions for optimal management. Two patients were both elderly women, case 1 had increased blood glucose after 7 months of using Durvalumab, and cases 2 had diabetic ketoacidosis after 6 weeks of using Pembrolizumab. Both patients were administered exogenous insulin to control blood glucose. Case 1 has been treated with Durvalumab until now and case 2 discontinued using of Pembrolizumab. HLA genotypes and other factors may explain the risk factors of diabetes associated with ICIs in some individuals. Diabetes mellitus associated with ICIs is an uncommon but potentially life-threatening endocrine system adverse event, which requires doctors to be vigilant. The patients who use ICIs need to monitor blood glucose. If they have hyperglycemia, endocrinologists should be asked to assist in diagnosis and treatment.
.
Collapse
|
17
|
Zand Irani A, Almuwais A, Gibbons H. Immune checkpoint inhibitor-induced diabetes mellitus with pembrolizumab. BMJ Case Rep 2022; 15:15/1/e245846. [PMID: 35039353 PMCID: PMC8768469 DOI: 10.1136/bcr-2021-245846] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An 81-year-old woman with a background of metastatic melanoma on pembrolizumab with no history of diabetes was brought into the emergency department with polyuria, polydipsia and weight loss. The initial assessment was consistent with severe diabetic ketoacidosis (DKA) and prerenal acute kidney injury with no clinical evidence of infection. The patient was treated with fluid resuscitation and an insulin infusion and eventually transitioned to a basal-bolus insulin regime, which was continued after discharge. Diabetes autoantibody screen returned negative, and she was diagnosed with immune checkpoint inhibitor–induced diabetes mellitus (ICI-induced DM) due to pembrolizumab. The patient has clinically improved and pembrolizumab was continued. The aim of this report is to highlight the importance of recognising ICI-induced DM as a rare immune-related adverse event in patients receiving programmed cell death protein 1/programmed cell death protein-ligand 1 inhibitor therapy and provide clinicians with insight into immune checkpoint endocrinopathies with an emphasis on diabetes and DKA.
Collapse
Affiliation(s)
- Anis Zand Irani
- Endocrinology, Gympie Hospital, Gympie, Queensland, Australia
| | - Ahmed Almuwais
- Medicine, Princess Alexandra Hospital, Woolloongabba, Queensland, Australia
| | - Holly Gibbons
- Medicine, Sunshine Coast University Hospital, Sunshine Coast, Queensland, Australia
| |
Collapse
|
18
|
Brodnicki TC. A Role for lncRNAs in Regulating Inflammatory and Autoimmune Responses Underlying Type 1 Diabetes. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1363:97-118. [DOI: 10.1007/978-3-030-92034-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
19
|
Loretelli C, Abdelsalam A, D'Addio F, Ben Nasr M, Assi E, Usuelli V, Maestroni A, Seelam AJ, Ippolito E, Di Maggio S, Loreggian L, Radovanovic D, Vanetti C, Yang J, El Essawy B, Rossi A, Pastore I, Montefusco L, Lunati ME, Bolla AM, Biasin M, Antinori S, Santus P, Riva A, Zuccotti G, Galli M, Rusconi S, Fiorina P. PD-1 blockade counteracts post-COVID-19 immune abnormalities and stimulates the anti-SARS-CoV-2 immune response. JCI Insight 2021; 6:146701. [PMID: 34784300 PMCID: PMC8783674 DOI: 10.1172/jci.insight.146701] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
A substantial proportion of patients who have recovered from coronavirus disease-2019 (COVID-19) experience COVID-19–related symptoms even months after hospital discharge. We extensively immunologically characterized patients who recovered from COVID-19. In these patients, T cells were exhausted, with increased PD-1+ T cells, as compared with healthy controls. Plasma levels of IL-1β, IL-1RA, and IL-8, among others, were also increased in patients who recovered from COVID-19. This altered immunophenotype was mirrored by a reduced ex vivo T cell response to both nonspecific and specific stimulation, revealing a dysfunctional status of T cells, including a poor response to SARS-CoV-2 antigens. Altered levels of plasma soluble PD-L1, as well as of PD1 promoter methylation and PD1-targeting miR–15-5p, in CD8+ T cells were also observed, suggesting abnormal function of the PD-1/PD-L1 immune checkpoint axis. Notably, ex vivo blockade of PD-1 nearly normalized the aforementioned immunophenotype and restored T cell function, reverting the observed post–COVID-19 immune abnormalities; indeed, we also noted an increased T cell–mediated response to SARS-CoV-2 peptides. Finally, in a neutralization assay, PD-1 blockade did not alter the ability of T cells to neutralize SARS-CoV-2 spike pseudotyped lentivirus infection. Immune checkpoint blockade ameliorates post–COVID-19 immune abnormalities and stimulates an anti–SARS-CoV-2 immune response.
Collapse
Affiliation(s)
- Cristian Loretelli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Ahmed Abdelsalam
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Francesca D'Addio
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, United States of America
| | - Emma Assi
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Vera Usuelli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Anna Maestroni
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Andy Joe Seelam
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Elio Ippolito
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Stefania Di Maggio
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Lara Loreggian
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Dejan Radovanovic
- Division of Respiratory Diseases, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Claudia Vanetti
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Jun Yang
- Institute of Organ Transplantation, Tongji Hospital and Medical College,, Wuhan, China
| | | | - Antonio Rossi
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Ida Pastore
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Laura Montefusco
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | | | - Andrea M Bolla
- Endocrinology Division, ASST Fatebenefratelli-Sacco, Milan, Italy
| | - Mara Biasin
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Spinello Antinori
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Pierachille Santus
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Gianvincenzo Zuccotti
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Massimo Galli
- Department of Biomedical and Clinical Science L. Sacco, Università di Milano, Milan, Italy
| | - Stefano Rusconi
- Infectious Diseases Unit, Legnano General Hospital, ASST Ovest Milanese, Legnano, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|
20
|
An in vivo method for diversifying the functions of therapeutic antibodies. Proc Natl Acad Sci U S A 2021; 118:2025596118. [PMID: 33658386 DOI: 10.1073/pnas.2025596118] [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] [Indexed: 12/12/2022] Open
Abstract
V(D)J recombination generates mature B cells that express huge repertoires of primary antibodies as diverse immunoglobulin (Ig) heavy chain (IgH) and light chain (IgL) of their B cell antigen receptors (BCRs). Cognate antigen binding to BCR variable region domains activates B cells into the germinal center (GC) reaction in which somatic hypermutation (SHM) modifies primary variable region-encoding sequences, with subsequent selection for mutations that improve antigen-binding affinity, ultimately leading to antibody affinity maturation. Based on these principles, we developed a humanized mouse model approach to diversify an anti-PD1 therapeutic antibody and allow isolation of variants with novel properties. In this approach, component Ig gene segments of the anti-PD1 antibody underwent de novo V(D)J recombination to diversify the anti-PD1 antibody in the primary antibody repertoire in the mouse models. Immunization of these mouse models further modified the anti-PD1 antibodies through SHM. Known anti-PD1 antibodies block interaction of PD1 with its ligands to alleviate PD1-mediated T cell suppression, thereby boosting antitumor T cell responses. By diversifying one such anti-PD1 antibody, we derived many anti-PD1 antibodies, including anti-PD1 antibodies with the opposite activity of enhancing PD1/ligand interaction. Such antibodies theoretically might suppress deleterious T cell activities in autoimmune diseases. The approach we describe should be generally applicable for diversifying other therapeutic antibodies.
Collapse
|
21
|
Abstract
PURPOSE OF REVIEW Programmed death-1 (PD-1) is an inhibitory receptor that controls T and B cell proliferation and function through interacting with its ligand PD-L1 or PD-L2. PD-1/PD-L1 blockade reboots anti-tumor immunity and is currently used to treat > 15 different types of cancer. However, the response rate is not at 100% and some patients relapse. Importantly, up to 37% of patients treated with PD-1/PD-L1 blocking antibodies develop immune-related adverse events, including overt autoimmunity, such as type 1 diabetes (T1D). Herein, we discuss the role of PD-1, PD-L1, and PD-L2 signaling in pre-clinical models of T1D, including recent work from our laboratory. RECENT FINDINGS We highlight ongoing efforts to harness PD-1/PD-L1 signaling and treat autoimmunity. We also evaluate studies aimed at defining biomarkers that could reliably predict the development of immune-related adverse events after clinical PD-1/PD-L1 blockade. With increasing use of PD-1 blockade in the clinic, onset of autoimmunity is a growing health concern. In this review, we discuss what is known about the role of PD-1 pathway signaling in T1D and comment on ongoing efforts to identify patients at risk of T1D development after PD-1 pathway blockade.
Collapse
Affiliation(s)
- Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA
| | - Alexander J Dwyer
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA.
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, 2101 6th St SE, Wallin Medical Biosciences Building, 3-146, Minneapolis, MN, 55455, USA.
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Thomas Building, D3-100, Seattle, WA, 98109, USA.
| |
Collapse
|
22
|
Liu J, Zhao Z, Qiu N, Zhou Q, Wang G, Jiang H, Piao Y, Zhou Z, Tang J, Shen Y. Co-delivery of IOX1 and doxorubicin for antibody-independent cancer chemo-immunotherapy. Nat Commun 2021; 12:2425. [PMID: 33893275 PMCID: PMC8065121 DOI: 10.1038/s41467-021-22407-6] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 03/10/2021] [Indexed: 12/12/2022] Open
Abstract
Anti-programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) antibodies are currently used in the clinic to interupt the PD-1/PD-L1 immune checkpoint, which reverses T cell dysfunction/exhaustion and shows success in treating cancer. Here, we report a histone demethylase inhibitor, 5-carboxy-8-hydroxyquinoline (IOX1), which inhibits tumour histone demethylase Jumonji domain-containing 1A (JMJD1A) and thus downregulates its downstream β-catenin and subsequent PD-L1, providing an antibody-independent paradigm interrupting the PD-1/PD-L1 checkpoint. Synergistically, IOX1 inhibits cancer cells’ P-glycoproteins (P-gp) through the JMJD1A/β-catenin/P-gp pathway and greatly enhances doxorubicin (DOX)-induced immune-stimulatory immunogenic cell death. As a result, the IOX1 and DOX combination greatly promotes T cell infiltration and activity and significantly reduces tumour immunosuppressive factors. Their liposomal combination reduces the growth of various murine tumours, including subcutaneous, orthotopic, and lung metastasis tumours, and offers a long-term immunological memory function against tumour rechallenging. This work provides a small molecule-based potent cancer chemo-immunotherapy. Some chemotherapeutic drugs, such as doxorubicin, induce immunogenic cell death (ICD) and promote anti-tumor immune responses. Here the authors report that the histone demethylase inhibitor 5-carboxy-8-hydroxyquinoline (IOX1) reduces the expression of PD-L1 in cancer cells and enhances doxorubicin-induced ICD, promoting T cell infiltration and reducing tumor growth in preclinical models.
Collapse
Affiliation(s)
- Jing Liu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhihao Zhao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Nasha Qiu
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Quan Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Guowei Wang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Haiping Jiang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Ying Piao
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Zhuxian Zhou
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China.,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China
| | - Jianbin Tang
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Youqing Shen
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China. .,Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, China.
| |
Collapse
|
23
|
Han P, Yu T, Hou Y, Zhao Y, Liu Y, Sun Y, Wang H, Xu P, Li G, Sun T, Hu X, Liu X, Li L, Peng J, Zhou H, Hou M. Low-Dose Decitabine Inhibits Cytotoxic T Lymphocytes-Mediated Platelet Destruction via Modulating PD-1 Methylation in Immune Thrombocytopenia. Front Immunol 2021; 12:630693. [PMID: 33679776 PMCID: PMC7925841 DOI: 10.3389/fimmu.2021.630693] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/27/2021] [Indexed: 12/31/2022] Open
Abstract
Cytotoxic T lymphocytes (CTLs)-mediated platelet destruction plays an important role in the pathogenesis of primary immune thrombocytopenia (ITP). The programmed cell death protein 1 (PD-1) signaling can turn off autoreactive T cells and induce peripheral tolerance. Herein, we found that the expression of PD-1 and its ligand PD-L1 on CD8+ T cells from ITP patients was decreased. Activating PD-1 pathway by PD-L1-Fc fusion protein inhibited CTLs-mediated platelet destruction in ITP in vitro. PD-1 promoter hypermethylation in CD8+ T cells was found in ITP patients, resulting in decreased PD-1 expression. The demethylating agent decitabine at a low dose was proved to restore the methylation level and expression of PD-1 on CD8+ T cells and reduce the cytotoxicity of CTLs of ITP patients. The phosphorylation levels of phosphatidylinositol 3-kinase (PI3K) and AKT in CD8+ T cells were significantly downregulated by low-dose decitabine. Furthermore, blocking PD-1 could counteract the effect of low-dose decitabine on CTLs from ITP patients. Therefore, our data suggest that the aberrant PD-1/PD-L1 pathway is involved in the pathophysiology of ITP and enhancing PD-1/PD-L1 signaling is a promising therapeutic approach for ITP management. Our results reveal the immunomodulatory mechanism of low-dose decitabine in ITP by inhibiting CTLs cytotoxicity to autologous platelets through PD-1 pathway.
Collapse
Affiliation(s)
- Panpan Han
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianshu Yu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yu Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yajing Zhao
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yang Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yunqi Sun
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haoyi Wang
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Pengcheng Xu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Guosheng Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tao Sun
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiang Hu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xinguang Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lizhen Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hai Zhou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Key Laboratory of Immunohematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Clinical Medicine Research Center for Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
24
|
Zheng Z, Liu Y, Yang J, Tan C, Zhou L, Wang X, Xiao L, Zhang S, Chen Y, Liu X. Diabetes mellitus induced by immune checkpoint inhibitors. Diabetes Metab Res Rev 2021; 37:e3366. [PMID: 32543027 DOI: 10.1002/dmrr.3366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 05/22/2020] [Accepted: 06/12/2020] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are widely used in oncology for their favourable antitumor efficacy. ICI therapy is associated with a unique toxicity profile known as immune-related adverse events (irAEs). One such irAE is ICI-related diabetes mellitus (DM), which is relatively uncommon but can become extremely severe, leading to irreversible impairment of β-cells, and even lead to death if not promptly recognised and properly managed. The precise mechanisms of ICI-related DM are not well understood. In this review, we summarise the clinical characteristics, pathophysiology, and management of this adverse effect caused by ICI therapy. Deeper investigation of ICI-related DM may contribute to elucidate the molecular mechanisms of classical type 1 DM.
Collapse
Affiliation(s)
- Zhenjiang Zheng
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ya Liu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Yang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Chunlu Tan
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Li Zhou
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, China
| | - Xing Wang
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Li Xiao
- Department of Traditional Chinese Medicine, Chengdu Third People's Hospital, Chengdu, China
| | - Shu Zhang
- Department of General Surgery, Chengdu Third People's Hospital, Chengdu, China
| | - Yonghua Chen
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xubao Liu
- Department of Pancreatic Surgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
25
|
Wu L, Li B. A Case of Severe Diabetic Ketoacidosis Associated with Pembrolizumab Therapy in a Patient with Metastatic Melanoma. Diabetes Metab Syndr Obes 2021; 14:753-757. [PMID: 33628041 PMCID: PMC7899305 DOI: 10.2147/dmso.s297709] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 01/29/2021] [Indexed: 11/27/2022] Open
Abstract
Pembrolizumab, a monoclonal antibody against programmed cell death-1 receptor, was licensed for advanced cancers. Although the use of pembrolizumab can enhance the effect of cancer treatment, it can increase immune-related adverse events. We describe an elderly woman who developed ketoacidosis after receiving pembrolizumab to treat metastatic melanoma. In the presentation, laboratory analysis showed that hyperglycemia and anion gap metabolic acidosis was consistent with diabetic ketoacidosis. Except for pembrolizumab, no other predisposing factors were found. The blood glucose levels before using pembrolizumab were normal. The patient responded well to intravenous fluids, insulin therapy, and treatment to correct electrolyte disturbances. She was diagnosed with severe diabetic ketoacidosis (DKA) because of new-onset diabetes mellitus which associated with pembrolizumab therapy. Two months after she was discharged from the hospital, she continued to take insulin as well as metformin to treat her diabetes. Clinicians need to be alert about diabetes mellitus and ketoacidosis for patients undergoing pembrolizumab treatment.
Collapse
Affiliation(s)
- Lili Wu
- Department of Integrated Medicine, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
| | - Bixun Li
- Department of Integrated Medicine, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China
- Correspondence: Bixun Li Department of Integrated Medicine, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, 530021, People’s Republic of China Email
| |
Collapse
|
26
|
Affiliation(s)
| | - Ruth Poole
- Poole Hospital NHS Foundation Trust Poole UK
| |
Collapse
|
27
|
Yamamoto Y, Kakizaki M, Shimizu T, Carreras J, Chiba T, Chamoto K, Kagawa T, Aoki T, Nakamura N, Ando K, Kotani A. PD-L1 is induced on the hepatocyte surface via CKLF-like MARVEL transmembrane domain-containing protein 6 up-regulation by the anti-HBV drug Entecavir. Int Immunol 2020; 32:519-531. [PMID: 32219331 DOI: 10.1093/intimm/dxaa018] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 03/26/2020] [Indexed: 12/15/2022] Open
Abstract
Chronic hepatitis B is now controllable when treated with nucleoside reverse transcriptase inhibitors (NRTIs), which inhibit hepatitis B virus (HBV) replication. However, once the NRTIs are discontinued, most patients relapse, necessitating lifelong NRTIs treatment. HBV infection relapse is assumed to be caused by the persistent existence of covalently closed circular DNA (cccDNA) in the nuclei of infected hepatocytes. The mechanism by which cccDNA-positive hepatocytes escape immune surveillance during NRTIs treatment remains elusive. Entecavir (ETV), a commonly used NRTI, post-transcriptionally up-regulates programmed cell death-ligand 1 (PD-L1), an immune checkpoint molecule, on the cell surface of hepatocytes regardless of HBV infection. Up-regulation by ETV depends on up-regulation of CKLF-like MARVEL transmembrane domain-containing 6, a newly identified potent regulator of PD-L1 expression on the cell surface. ETV-treated hepatic cells suppressed the activity of primary CD3 T cells and programmed cell death protein-1 (PD-1)-over-expressed Jurkat cells. Finally, ETV induces PD-L1 in primary hepatocytes infected by HBV. These results provide evidence that ETV considerably up-regulates PD-L1 on the cell surface of infected hepatocytes, which may be one of the mechanisms by which infected hepatocytes subvert immune surveillance.
Collapse
Affiliation(s)
- Yuichiro Yamamoto
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan
| | - Masatoshi Kakizaki
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan.,Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Takayuki Shimizu
- Department of Gastroenterological Surgery, Dokkyo Medical University, Kitakobayashi, Mibu, Japan
| | - Joaquim Carreras
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Tetsuhiro Chiba
- Department of Gastroenterology, Chiba University, Graduate School of Medicine, Inohana, Chuo-ku, Chiba, Japan
| | - Kenji Chamoto
- Department of Immunology and Genomic Medicine, Kyoto University, Graduate School of Medicine, Yoshida Konoe-cho, Sakyo-ku, Kyoto, Japan
| | - Tatehiro Kagawa
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Tokai University School of Medicine, Isehara, Japan
| | - Taku Aoki
- Department of Gastroenterological Surgery, Dokkyo Medical University, Kitakobayashi, Mibu, Japan
| | - Naoya Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Japan
| | - Kiyoshi Ando
- Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Ai Kotani
- Division of Hematological Malignancy, Institute of Medical Sciences, Tokai University, Isehara, Japan.,Department of Hematology and Oncology, Tokai University School of Medicine, Isehara, Japan
| |
Collapse
|
28
|
Dendritic cell upregulation of programmed death ligand-1 via DNA demethylation inhibits experimental autoimmune encephalomyelitis. J Autoimmun 2020; 107:102362. [DOI: 10.1016/j.jaut.2019.102362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 11/04/2019] [Accepted: 11/06/2019] [Indexed: 01/08/2023]
|
29
|
Martinov T, Fife BT. Type 1 diabetes pathogenesis and the role of inhibitory receptors in islet tolerance. Ann N Y Acad Sci 2020; 1461:73-103. [PMID: 31025378 PMCID: PMC6994200 DOI: 10.1111/nyas.14106] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/25/2019] [Accepted: 04/03/2019] [Indexed: 12/15/2022]
Abstract
Type 1 diabetes (T1D) affects over a million Americans, and disease incidence is on the rise. Despite decades of research, there is still no cure for this disease. Exciting beta cell replacement strategies are being developed, but in order for such approaches to work, targeted immunotherapies must be designed. To selectively halt the autoimmune response, researchers must first understand how this response is regulated and which tolerance checkpoints fail during T1D development. Herein, we discuss the current understanding of T1D pathogenesis in humans, genetic and environmental risk factors, presumed roles of CD4+ and CD8+ T cells as well as B cells, and implicated autoantigens. We also highlight studies in non-obese diabetic mice that have demonstrated the requirement for CD4+ and CD8+ T cells and B cells in driving T1D pathology. We present an overview of central and peripheral tolerance mechanisms and comment on existing controversies in the field regarding central tolerance. Finally, we discuss T cell- and B cell-intrinsic tolerance mechanisms, with an emphasis on the roles of inhibitory receptors in maintaining islet tolerance in humans and in diabetes-prone mice, and strategies employed to date to harness inhibitory receptor signaling to prevent or reverse T1D.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
30
|
Parackova Z, Vrabcova P, Zentsova I, Kayserova J, Richtrova I, Sojka L, Stechova K, Sumnik Z, Sediva A. Enhanced STAT3 phosphorylation and PD-L1 expression in myeloid dendritic cells indicate impaired IL-27Ralpha signaling in type 1 diabetes. Sci Rep 2020; 10:493. [PMID: 31949260 PMCID: PMC6965661 DOI: 10.1038/s41598-020-57507-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 01/02/2020] [Indexed: 02/06/2023] Open
Abstract
Interleukin 27 (IL-27), a member of the IL-12 family, is important for T cell differentiation; however, little is known about its effect on dendritic cells (DCs). IL-27 can activate multiple signaling cascades, including the JAK/STAT pathway, and depending on the setting it can both promote and antagonize inflammatory responses. An anti-inflammatory function of IL-27 has been reported in several autoimmune diseases; however, in type 1 diabetes (T1D), an autoimmune disease where autoreactive cytotoxic T cells attack insulin-producing beta cells, IL-27 has been shown to have a dual role and contradictory effects. Here, we show impaired IL-27 signaling in a large cohort of T1D patients (n = 51) compared to age- and gender-matched healthy donors. Increased expression of the IL-27 receptor subunit IL-27Ralpha mRNA in purified myeloid DCs (mDCs), detected by gene expression microarrays was mirrored by enhanced signal transduction in T1D mDCs in response to IL-27 stimulation. Higher STAT phosphorylation in T1D patients was also accompanied by elevated expression of the inhibitory molecules PD-L1, PD-L2 and PD-1, which may suggest not only immunomodulatory mechanisms of IL-27 in T1D but also a compensatory effort of T1D dendritic cells against the ongoing inflammation.
Collapse
Affiliation(s)
- Z Parackova
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.
| | - P Vrabcova
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - I Zentsova
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - J Kayserova
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | | - L Sojka
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic.,Sotio, A. S., Prague, Czech Republic
| | - K Stechova
- Department of Internal Medicine, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Z Sumnik
- Department of Pediatrics, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - A Sediva
- Department of Immunology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| |
Collapse
|
31
|
Falcone M, Fousteri G. Role of the PD-1/PD-L1 Dyad in the Maintenance of Pancreatic Immune Tolerance for Prevention of Type 1 Diabetes. Front Endocrinol (Lausanne) 2020; 11:569. [PMID: 32973682 PMCID: PMC7466754 DOI: 10.3389/fendo.2020.00569] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022] Open
Abstract
The human pancreas, like almost all organs in the human body, is immunologically tolerated despite the presence of innate and adaptive immune cells that promptly mediate protective immune responses against pathogens in situ. The PD-1/PD-L1 inhibitory pathway seems to play a key role in the maintenance of immune tolerance systemically and within the pancreatic tissue. Tissue resident memory T cells (TRM), T regulatory cells (Treg), macrophages and even β cells exhibit PD-1 or PD-L1 expression that contributes in controlling pancreatic immune homeostasis and tolerance. Dysregulation of the PD-1/PD-L1 axis as shown by animal studies and our recent experience with checkpoint inhibitory blockade in humans can lead to immune dysfunctions leading to chronic inflammatory disease and to type 1 diabetes (T1D) in genetically susceptible individuals. In this review, we discuss the role of the PD-1/PD-L1 axis in pancreatic tissue homeostasis and tolerance, speculate how genetic and environmental factors can regulate the PD-1/PD-L1 pathway, and discuss PD-1/PD-L1-based therapeutic approaches for pancreatic islet transplantation and T1D treatment.
Collapse
|
32
|
Marchand L, Disse E, Dalle S, Reffet S, Vouillarmet J, Fabien N, Thivolet C, Cugnet-Anceau C. The multifaceted nature of diabetes mellitus induced by checkpoint inhibitors. Acta Diabetol 2019; 56:1239-1245. [PMID: 31423559 DOI: 10.1007/s00592-019-01402-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2019] [Accepted: 08/05/2019] [Indexed: 12/30/2022]
Abstract
Immune checkpoint inhibitors (CPI) are increasingly being used in oncology, and many autoimmune side effects have been described. Diabetes mellitus (DM) has been reported in approximately 1% of subjects treated with programmed cell death-1 and programmed death ligand 1 (PD-1/PD-L1) inhibitors, alone or in association with CTLA-4 inhibitors. In the present mini-review, we aimed to describe different clinical pictures and pathophysiology associated with these forms of diabetes. Data on CPI-related DM was gathered from the largest case series in the literature and from our centre dedicated to immunotherapy complications (ImmuCare-Hospices Civils de Lyon). Most cases are acute autoimmune insulin-dependent diabetes which are similar to fulminant diabetes (extremely acute onset with concomitant near-normal HbA1c levels). Other cases, however, have a phenotype close to type 2 diabetes or appear as a decompensation of previously known type 2 diabetes. The occurrence of diabetes can also be a complication of autoimmune pancreatitis induced by CPI use. Finally, two cases of diabetes in a context of autoimmune lipoatrophy have recently been described. Regarding the wide variety of CPI-induced diabetes, the discovery of a glucose disorder under CPI should motivate specialised care for aetiological diagnosis and appropriate treatment.
Collapse
MESH Headings
- Autoimmune Diseases/chemically induced
- Autoimmune Diseases/epidemiology
- Autoimmune Diseases/etiology
- B7-H1 Antigen/antagonists & inhibitors
- B7-H1 Antigen/immunology
- CTLA-4 Antigen/antagonists & inhibitors
- CTLA-4 Antigen/immunology
- Cell Cycle Checkpoints/drug effects
- Cell Cycle Checkpoints/immunology
- Diabetes Mellitus, Lipoatrophic/chemically induced
- Diabetes Mellitus, Lipoatrophic/epidemiology
- Diabetes Mellitus, Lipoatrophic/immunology
- Diabetes Mellitus, Type 1/chemically induced
- Diabetes Mellitus, Type 1/complications
- Diabetes Mellitus, Type 1/diagnosis
- Diabetes Mellitus, Type 1/epidemiology
- Diabetes Mellitus, Type 2/chemically induced
- Diabetes Mellitus, Type 2/diagnosis
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/immunology
- Humans
- Immunotherapy/adverse effects
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
Collapse
Affiliation(s)
- Lucien Marchand
- Department of Endocrinology and Diabetes, St. Joseph - St. Luc Hospital, Quai Claude Bernard, 69007, Lyon, France.
| | - Emmanuel Disse
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
- CarMeN Laboratory (INSERM U1060, INRA U1235, Université Claude Bernard Lyon1, INSA-Lyon), Lyon 1 University, Oullins, France
| | - Stéphane Dalle
- Department of Dermatology, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
- ImmuCare (Immunology Cancer Research), Hospices Civils de Lyon, Lyon, France
| | - Sophie Reffet
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Julien Vouillarmet
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Nicole Fabien
- Department of Immunology, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
| | - Charles Thivolet
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
- CarMeN Laboratory (INSERM U1060, INRA U1235, Université Claude Bernard Lyon1, INSA-Lyon), Lyon 1 University, Oullins, France
| | - Christine Cugnet-Anceau
- Department of Endocrinology and Diabetes, Hospices Civils de Lyon, Lyon-Sud Hospital, Pierre-Bénite, France
- ImmuCare (Immunology Cancer Research), Hospices Civils de Lyon, Lyon, France
| |
Collapse
|
33
|
Dendritic cells license regulatory B cells to produce IL-10 and mediate suppression of antigen-specific CD8 T cells. Cell Mol Immunol 2019; 17:843-855. [PMID: 31728048 DOI: 10.1038/s41423-019-0324-z] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 10/23/2019] [Indexed: 01/08/2023] Open
Abstract
Regulatory B cells (Bregs) suppress and reduce autoimmune pathology. However, given the variety of Breg subsets, the role of Bregs in the pathogenesis of type 1 diabetes is still unclear. Here, we dissect this fundamental mechanism. We show that natural protection from type 1 diabetes in nonobese diabetic (NOD) mice is associated with increased numbers of IL-10-producing B cells, while development of type 1 diabetes in NOD mice occurs in animals with compromised IL-10 production by B cells. However, B cells from diabetic mice regain IL-10 function if activated by the innate immune receptor TLR4 and can suppress insulin-specific CD8 T cells in a dendritic cell (DC)-dependent, IL-10-mediated fashion. Suppression of CD8 T cells is reliant on B-cell contact with DCs. This cell contact results in deactivation of DCs, inducing a tolerogenic state, which in turn can regulate pathogenic CD8 T cells. Our findings emphasize the importance of DC-Breg interactions during the development of type 1 diabetes.
Collapse
|
34
|
Martinov T, Swanson LA, Breed ER, Tucker CG, Dwyer AJ, Johnson JK, Mitchell JS, Sahli NL, Wilson JC, Singh LM, Hogquist KA, Spanier JA, Fife BT. Programmed Death-1 Restrains the Germinal Center in Type 1 Diabetes. THE JOURNAL OF IMMUNOLOGY 2019; 203:844-852. [PMID: 31324724 DOI: 10.4049/jimmunol.1801535] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 06/18/2019] [Indexed: 01/22/2023]
Abstract
Programmed death-1 (PD-1) inhibits T and B cell function upon ligand binding. PD-1 blockade revolutionized cancer treatment, and although numerous patients respond, some develop autoimmune-like symptoms or overt autoimmunity characterized by autoantibody production. PD-1 inhibition accelerates autoimmunity in mice, but its role in regulating germinal centers (GC) is controversial. To address the role of PD-1 in the GC reaction in type 1 diabetes, we used tetramers to phenotype insulin-specific CD4+ T and B cells in NOD mice. PD-1 or PD-L1 deficiency, and PD-1 but not PD-L2 blockade, unleashed insulin-specific T follicular helper CD4+ T cells and enhanced their survival. This was concomitant with an increase in GC B cells and augmented insulin autoantibody production. The effect of PD-1 blockade on the GC was reduced when mice were treated with a mAb targeting the insulin peptide:MHC class II complex. This work provides an explanation for autoimmune side effects following PD-1 pathway inhibition and suggests that targeting the self-peptide:MHC class II complex might limit autoimmunity arising from checkpoint blockade.
Collapse
Affiliation(s)
- Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Linnea A Swanson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Elise R Breed
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Alexander J Dwyer
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Jenna K Johnson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Jason S Mitchell
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Nathanael L Sahli
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Joseph C Wilson
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Lovejot M Singh
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Kristin A Hogquist
- Department of Laboratory Medicine and Pathology, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455
| | - Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN 55455; and
| |
Collapse
|
35
|
Bajwa R, Cheema A, Khan T, Amirpour A, Paul A, Chaughtai S, Patel S, Patel T, Bramson J, Gupta V, Levitt M, Asif A, Hossain MA. Adverse Effects of Immune Checkpoint Inhibitors (Programmed Death-1 Inhibitors and Cytotoxic T-Lymphocyte-Associated Protein-4 Inhibitors): Results of a Retrospective Study. J Clin Med Res 2019; 11:225-236. [PMID: 30937112 PMCID: PMC6436564 DOI: 10.14740/jocmr3750] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 02/13/2019] [Indexed: 12/11/2022] Open
Abstract
In recent years the use of immunomodulating therapy to treat various cancers has been on the rise. Three checkpoint inhibitors have been approved by the Food and Drug Administration (ipilimumab, pembrolizumab and nivolumab). The use of these drugs comes with serious adverse events related to excessive immune activation, collectively known as immune-related adverse events (irAEs). We conducted a system-based review of 139 case reports/case series that have described these adverse events between January 2016 and April 2018, found in the PubMed database. There was a broad spectrum of presentations, doses and checkpoint inhibitors used. The most common check point inhibitor observed in our literature review was nivolumab. The most common adverse effects encountered were colitis (14/139), hepatitis (11/139), adrenocorticotropic hormone insufficiency (12/139), hypothyroidism (7/139), type 1 diabetes (22/139), acute kidney injury (16/139) and myocarditis (10/139). The treatment most commonly consisted of cessation of the immune checkpoint inhibitor, initiation of steroids and supportive therapy. This approach provided a complete resolution in a majority of cases; however, there were many that developed long-term adverse events with deaths reported in a few cases. The endocrine system was the mostly commonly affected with the development of type 1 diabetes mellitus or diabetic ketoacidosis being the most frequently reported adverse events. While immunomodulating therapy is a significant advance in the management of various malignancies, it is capable of serious adverse effects. Because the majority of the cases developed pancreatic dysfunction within five cycles of therapy, in addition to the evaluation of other systems, pancreatic function should be closely monitored to minimize adverse impact on patients.
Collapse
Affiliation(s)
- Ravneet Bajwa
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Anmol Cheema
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Taimoor Khan
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Alireza Amirpour
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Anju Paul
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Saira Chaughtai
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Shrinil Patel
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Tejas Patel
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Joshua Bramson
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Varsha Gupta
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Michael Levitt
- Department of Hematology/Oncology, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Arif Asif
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Mohammad A Hossain
- Department of Medicine, Jersey Shore University Medical Center, Neptune, NJ, USA
| |
Collapse
|
36
|
Abstract
The clinical onset of type 1 diabetes is characterized by the destruction of the insulin-producing β cells of the pancreas and is caused by autoantigen-induced inflammation (insulitis) of the islets of Langerhans. The current standard of care for type 1 diabetes mellitus patients allows for management of the disease with exogenous insulin, but patients eventually succumb to many chronic complications such as limb amputation, blindness, and kidney failure. New therapeutic approaches now on the horizon are looking beyond glycemic management and are evaluating new strategies from protecting and regenerating endogenous islets to treating the underlying autoimmunity through selective modulation of key immune cell populations. Currently, there are no effective treatments for the autoimmunity that causes the disease, and strategies that aim to delay or prevent the onset of the disease will play an important role in the future of diabetes research. In this review, we summarize many of the key efforts underway that utilize molecular approaches to selectively modulate this disease and look at new therapeutic paradigms that can transform clinical treatment.
Collapse
Affiliation(s)
- Daniel Sheehy
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Sean Quinnell
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| | - Arturo J. Vegas
- Department of Chemistry, Boston University, Boston, Massachusetts 02215, United States
| |
Collapse
|
37
|
Sothornwit J, Phunmanee A, Pongchaiyakul C. Atezolizumab-Induced Autoimmune Diabetes in a Patient With Metastatic Lung Cancer. Front Endocrinol (Lausanne) 2019; 10:352. [PMID: 31244772 PMCID: PMC6581023 DOI: 10.3389/fendo.2019.00352] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 05/17/2019] [Indexed: 12/20/2022] Open
Abstract
Context: Immune checkpoint inhibitors (ICIs), now FDA-approved, are increasingly used as an effective treatment of various cancers. Autoimmune diabetes is a rare but life-threatening endocrine adverse event, which has been reported in patients treated with anti-programmed-cell death-1 (anti-PD-1) and anti-programmed-cell death-1 ligand (anti-PD-L1) therapies. Case description: We report a 52-year-old woman with advanced-stage non-small cell lung cancer who presented with diabetic ketoacidosis (DKA) at 24 weeks after atezolizumab initiation. She initially received oral antidiabetic medication from primary care hospital and experienced recurrent DKA 3 days later. Her plasma glucose on the day that she had recurrent DKA was 332 mg/dL (18.4 mmol/L), A1c was 7.9% (63 mmol/mol), fasting C-peptide was <0.03 nmol/L (0.1 ng/ml), fasting insulin level was <1 μIU/ml, anti-glutamic acid decarboxylase 65 (GADA) was 7.2 U/ml (normal, >5 U/ml), and human leukocyte antigen (HLA) class II typing was DR3-DQ2/DR14-DQ5. A diagnosis of autoimmune diabetes was made. After treatment for DKA, she recovered and received basal-bolus insulin treatment. Atezolizumab had been discontinued after the fifth cycle, prior to the development of DKA, due to progression of lung cancer. Conclusion: To date, there has been neither an effective way to detect if a patient is at high risk for autoimmune diabetes nor to prevent the complications associated with it. Regular glucose monitoring is the best method of early diabetes detection. In patients with new onset diabetes following treatment with ICIs, C-peptide levels and GADA should be screened, and insulin therapy should be prescribed to prevent hyperglycemic emergency while waiting for definite diagnosis.
Collapse
Affiliation(s)
- Jin Sothornwit
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
- *Correspondence: Jin Sothornwit
| | - Anakapong Phunmanee
- Division of Critical Care, Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Chatlert Pongchaiyakul
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
38
|
Tezza S, Ben Nasr M, D'Addio F, Vergani A, Usuelli V, Falzoni S, Bassi R, Dellepiane S, Fotino C, Rossi C, Maestroni A, Solini A, Corradi D, Giani E, Mameli C, Bertuzzi F, Pezzolesi MG, Wasserfall CH, Atkinson MA, Füchtbauer EM, Ricordi C, Folli F, Di Virgilio F, Pileggi A, Dhe-Paganon S, Zuccotti GV, Fiorina P. Islet-Derived eATP Fuels Autoreactive CD8 + T Cells and Facilitates the Onset of Type 1 Diabetes. Diabetes 2018; 67:2038-2053. [PMID: 30065030 PMCID: PMC6905486 DOI: 10.2337/db17-1227] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 07/06/2018] [Indexed: 01/13/2023]
Abstract
Extracellular ATP (eATP) activates T cells by engaging the P2X7R receptor. We identified two loss-of-function P2X7R mutations that are protective against type 1 diabetes (T1D) and thus hypothesized that eATP/P2X7R signaling may represent an early step in T1D onset. Specifically, we observed that in patients with newly diagnosed T1D, P2X7R is upregulated on CD8+ effector T cells in comparison with healthy control subjects. eATP is released at high levels by human/murine islets in vitro in high-glucose/inflammatory conditions, thus upregulating P2X7R on CD8+ T cells in vitro. P2X7R blockade with oxidized ATP reduces the CD8+ T cell-mediated autoimmune response in vitro and delays diabetes onset in NOD mice. Autoreactive CD8+ T-cell activation is highly dependent upon eATP/P2X7R-mediated priming, while a novel sP2X7R recombinant protein abrogates changes in metabolism and the autoimmune response associated with CD8+ T cells. eATP/P2X7R signaling facilitates the onset of autoimmune T1D by fueling autoreactive CD8+ cells and therefore represents a novel targeted therapeutic for the disorder.
Collapse
Affiliation(s)
- Sara Tezza
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Moufida Ben Nasr
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Francesca D'Addio
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Andrea Vergani
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Vera Usuelli
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Simonetta Falzoni
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | - Roberto Bassi
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Sergio Dellepiane
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
| | - Carmen Fotino
- Diabetes Research Institute, University of Miami, FL
| | - Chiara Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Anna Maestroni
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
| | - Anna Solini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Domenico Corradi
- Pathology and Laboratory Medicine, University of Parma, Parma, Italy
| | - Elisa Giani
- Pediatric Clinical Romeo and Enrica Invernizzi Research Center, L. Sacco Department of Biomedical and Clinical Science, University of Milan, and Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Chiara Mameli
- Pediatric Clinical Romeo and Enrica Invernizzi Research Center, L. Sacco Department of Biomedical and Clinical Science, University of Milan, and Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Federico Bertuzzi
- Diabetology Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Marcus G Pezzolesi
- Division of Nephrology & Hypertension and Diabetes & Metabolism Research Center, University of Utah, Salt Lake City, UT
| | - Clive H Wasserfall
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | - Mark A Atkinson
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL
| | | | | | - Franco Folli
- Endocrinology and Metabolism, Department of Health Science, University of Milan, Milan, Italy
- ASST Santi Paolo e Carlo, Ospedali San Paolo e San Carlo Borromeo, Milan, Italy
| | - Francesco Di Virgilio
- Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Ferrara, Italy
| | | | | | - Gian Vincenzo Zuccotti
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
- Pediatric Clinical Romeo and Enrica Invernizzi Research Center, L. Sacco Department of Biomedical and Clinical Science, University of Milan, and Department of Pediatrics, Children's Hospital Buzzi, Milan, Italy
| | - Paolo Fiorina
- Division of Nephrology, Boston Children's Hospital, Harvard Medical School, Boston, MA
- International Center for Type 1 Diabetes, Pediatric Clinical Romeo and Enrica Invernizzi Research Center, and L. Sacco Department of Biomedical and Clinical Science, University of Milan, Milan, Italy
- Division of Endocrinology, ASST Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
39
|
Shahbazi M, Soltanzadeh-Yamchi M, Mohammadnia-Afrouzi M. T cell exhaustion implications during transplantation. Immunol Lett 2018; 202:52-58. [PMID: 30130559 DOI: 10.1016/j.imlet.2018.08.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 08/05/2018] [Accepted: 08/16/2018] [Indexed: 12/20/2022]
Abstract
Exhaustion of lymphocyte function, particularly T cell exhaustion, due to prolonged exposure to a high load of foreign antigen is commonly seen during chronic viral infection as well as antitumor immune responses. This phenomenon has been associated with a determined molecular mechanism and phenotypic manifestations on the cell surface. In spite of investigation of exhaustion, mostly about CD8 responses toward viral infections, recent studies have reported that chronic exposure to antigen may develop exhaustion in CD4 + T cells, B cells, and NK cells. Little is known with respect to lymphocyte exhaustion during transplantation and its effect on aberrant anti-graft responses. Through a same mechanobiology observed during chronic exposure of foreign viral antigens, alloantigen persistence mediated by allograft could develop a favorable circumstance for exhaustion of T cells responding to allograft. However, to achieve better manipulation approaches of this event to reduce the complications during transplantation, we need to be armed with a bulk of knowledge with regard to quality and quantity of T cell exhaustion occurring in various allografts, the kinetics of exhaustion development, the impression of immunosuppressive agents on the exhaustion, and the influence of exhaustion on graft survival and immune tolerance.
Collapse
Affiliation(s)
- Mehdi Shahbazi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mehdi Soltanzadeh-Yamchi
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Mousa Mohammadnia-Afrouzi
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Immunoregulation Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran; Department of Immunology, School of Medicine, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
40
|
Ben Nasr M, Tezza S, D'Addio F, Mameli C, Usuelli V, Maestroni A, Corradi D, Belletti S, Albarello L, Becchi G, Fadini GP, Schuetz C, Markmann J, Wasserfall C, Zon L, Zuccotti GV, Fiorina P. PD-L1 genetic overexpression or pharmacological restoration in hematopoietic stem and progenitor cells reverses autoimmune diabetes. Sci Transl Med 2018; 9:9/416/eaam7543. [PMID: 29141886 DOI: 10.1126/scitranslmed.aam7543] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/01/2017] [Accepted: 08/14/2017] [Indexed: 12/13/2022]
Abstract
Immunologically based clinical trials performed thus far have failed to cure type 1 diabetes (T1D), in part because these approaches were nonspecific. Because the disease is driven by autoreactive CD4 T cells, which destroy β cells, transplantation of hematopoietic stem and progenitor cells (HSPCs) has been recently offered as a therapy for T1D. Our transcriptomic profiling of HSPCs revealed that these cells are deficient in programmed death ligand 1 (PD-L1), an important immune checkpoint, in the T1D nonobese diabetic (NOD) mouse model. Notably, the immunoregulatory molecule PD-L1 plays a determinant role in controlling/inhibiting activated T cells and thus maintains immune tolerance. Furthermore, our genome-wide and bioinformatic analysis revealed the existence of a network of microRNAs (miRNAs) controlling PD-L1 expression, and silencing one of key altered miRNAs restored PD-L1 expression in HSPCs. We therefore sought to determine whether restoration of this defect would cure T1D as an alternative to immunosuppression. Genetically engineered or pharmacologically modulated HSPCs overexpressing PD-L1 inhibited the autoimmune response in vitro, reverted diabetes in newly hyperglycemic NOD mice in vivo, and homed to the pancreas of hyperglycemic NOD mice. The PD-L1 expression defect was confirmed in human HSPCs in T1D patients as well, and pharmacologically modulated human HSPCs also inhibited the autoimmune response in vitro. Targeting a specific immune checkpoint defect in HSPCs thus may contribute to establishing a cure for T1D.
Collapse
Affiliation(s)
- Moufida Ben Nasr
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy
| | - Sara Tezza
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Francesca D'Addio
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy
| | - Chiara Mameli
- Department of Pediatrics, Buzzi Children's Hospital, Milan 20154, Italy
| | - Vera Usuelli
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA.,International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy
| | - Anna Maestroni
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy
| | | | | | - Luca Albarello
- Pathology Unit, Ospedale San Raffaele, Milan 20132, Italy
| | | | | | - Christian Schuetz
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - James Markmann
- Department of Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Clive Wasserfall
- Department of Pathology, University of Florida, Gainesville, FL 32611, USA
| | - Leonard Zon
- Division of Hematology/Oncology, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Gian Vincenzo Zuccotti
- International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy.,Department of Pediatrics, Buzzi Children's Hospital, Milan 20154, Italy
| | - Paolo Fiorina
- Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA. .,International Center for T1D, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, "L. Sacco" Department of Biomedical and Clinical Sciences, University of Milan, Milan 20157, Italy.,Department of Endocrinology, ASST Fatebenefratelli-Sacco, Milan 20121, Italy
| |
Collapse
|
41
|
Osum KC, Burrack AL, Martinov T, Sahli NL, Mitchell JS, Tucker CG, Pauken KE, Papas K, Appakalai B, Spanier JA, Fife BT. Interferon-gamma drives programmed death-ligand 1 expression on islet β cells to limit T cell function during autoimmune diabetes. Sci Rep 2018; 8:8295. [PMID: 29844327 PMCID: PMC5974126 DOI: 10.1038/s41598-018-26471-9] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 05/09/2018] [Indexed: 02/06/2023] Open
Abstract
Type 1 diabetes is caused by autoreactive T cell-mediated β cell destruction. Even though co-inhibitory receptor programmed death-1 (PD-1) restrains autoimmunity, the expression and regulation of its cognate ligands on β cell remains unknown. Here, we interrogated β cell-intrinsic programmed death ligand-1 (PD-L1) expression in mouse and human islets. We measured a significant increase in the level of PD-L1 surface expression and the frequency of PD-L1+ β cells as non-obese diabetic (NOD) mice aged and developed diabetes. Increased β cell PD-L1 expression was dependent on T cell infiltration, as β cells from Rag1-deficient mice lacked PD-L1. Using Rag1-deficient NOD mouse islets, we determined that IFN-γ promotes β cell PD-L1 expression. We performed analogous experiments using human samples, and found a significant increase in β cell PD-L1 expression in type 1 diabetic samples compared to type 2 diabetic, autoantibody positive, and non-diabetic samples. Among type 1 diabetic samples, β cell PD-L1 expression correlated with insulitis. In vitro experiments with human islets from non-diabetic individuals showed that IFN-γ promoted β cell PD-L1 expression. These results suggest that insulin-producing β cells respond to pancreatic inflammation and IFN-γ production by upregulating PD-L1 expression to limit self-reactive T cells.
Collapse
Affiliation(s)
- Kevin C Osum
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Adam L Burrack
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Tijana Martinov
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Nathanael L Sahli
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Jason S Mitchell
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Christopher G Tucker
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Kristen E Pauken
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Klearchos Papas
- Department of Surgery, University of Arizona, Tucson, AZ, USA
| | | | - Justin A Spanier
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA
| | - Brian T Fife
- Department of Medicine, Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, 55455, USA.
| |
Collapse
|
42
|
Helminth infection in mice improves insulin sensitivity via modulation of gut microbiota and fatty acid metabolism. Pharmacol Res 2018; 132:33-46. [PMID: 29653264 DOI: 10.1016/j.phrs.2018.04.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/14/2018] [Accepted: 04/09/2018] [Indexed: 12/19/2022]
Abstract
Intestinal helminths are prevalent in individuals who live in rural areas of developing countries, where obesity, type 2 diabetes, and metabolic syndrome are rare. In the present study, we analyzed the modulation of the gut microbiota in mice infected with the helminth Strongyloides venezuelensis, and fed either a standard rodent chow diet or high-fat diet (HFD). To investigate the effects of the microbiota modulation on the metabolism, we analyzed the expression of tight-junction proteins present in the gut epithelium, inflammatory markers in the serum and tissue and quantified glucose tolerance and insulin sensitivity and resistance. Additionally, the levels of lipids related to inflammation were evaluated in the feces and serum. Our results show that infection with Strongyloides venezuelensis results in a modification of the gut microbiota, most notably by increasing Lactobacillus spp. These modifications in the microbiota alter the host metabolism by increasing the levels of anti-inflammatory cytokines, switching macrophages from a M1 to M2 pattern in the adipose tissue, increasing the expression of tight junction proteins in the intestinal cells (thereby reducing the permeability) and decreasing LPS in the serum. Taken together, these changes correlate with improved insulin signaling and sensitivity, which could also be achieved with HFD mice treated with probiotics. Additionally, helminth infected mice produce higher levels of oleic acid, which participates in anti-inflammatory pathways. These results suggest that modulation of the microbiota by helminth infection or probiotic treatment causes a reduction in subclinical inflammation, which has a positive effect on the glucose metabolism of the host.
Collapse
|
43
|
Zaied AA, Akturk HK, Joseph RW, Lee AS. New-onset insulin-dependent diabetes due to nivolumab. Endocrinol Diabetes Metab Case Rep 2018; 2018:EDM170174. [PMID: 29623210 PMCID: PMC5881429 DOI: 10.1530/edm-17-0174] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 03/02/2018] [Indexed: 01/05/2023] Open
Abstract
Nivolumab, a monoclonal antibody against programmed cell death-1 receptor, is increasingly used in advanced cancers. While nivolumab use enhances cancer therapy, it is associated with increased immune-related adverse events. We describe an elderly man who presented in ketoacidosis after receiving nivolumab for metastatic renal cell carcinoma. On presentation, he was hyperpneic and laboratory analyses showed hyperglycemia and anion-gapped metabolic acidosis consistent with diabetic ketoacidosis. No other precipitating factors, besides nivolumab, were identified. Pre-nivolumab blood glucose levels were normal. The patient responded to treatment with intravenous fluids, insulin and electrolyte replacement. He was diagnosed with insulin-dependent autoimmune diabetes mellitus secondary to nivolumab. Although nivolumab was stopped, he continued to require multiple insulin injection therapy till his last follow-up 7 months after presentation. Clinicians need to be alerted to the development of diabetes mellitus and diabetic ketoacidosis in patients receiving nivolumab.
Collapse
Affiliation(s)
- Ali A Zaied
- Divisions of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, Florida, USA
| | - Halis K Akturk
- Divisions of Endocrinology, Mayo Clinic, Jacksonville, Florida, USA
| | - Richard W Joseph
- Divisions of Hematology and Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Augustine S Lee
- Divisions of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, Florida, USA
| |
Collapse
|
44
|
Araújo M, Ligeiro D, Costa L, Marques F, Trindade H, Correia JM, Fonseca C. A case of fulminant Type 1 diabetes following anti-PD1 immunotherapy in a genetically susceptible patient. Immunotherapy 2018; 9:531-535. [PMID: 28595520 DOI: 10.2217/imt-2017-0020] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Programmed cell death-1 protein (PD-1) is an immune checkpoint that has gained popularity in the treatment of several advanced cancers. Inhibiting this checkpoint is known to enhance immune response, but is also known to diminish immune tolerance and to increase autoimmune toxicity. We discuss a case of rapid onset fulminant Type 1 diabetes induced by treatment with anti-programmed cell death-1 monoclonal antibody, nivolumab, in a patient with late-stage non-small-cell lung adenocarcinoma. The patient had no history of previous diabetes but did reveal a high-risk genotype for Type 1 diabetes development (DR3-DQ2; DR4-DQ8). This finding supports that acute Type 1 diabetes can be an important adverse effect of immunotherapies targeting T-cell activation regulation. Because of the severity of this adverse effect, physicians should be aware of it, and studies directed to the detection of new biomarkers for early risk stratification (e.g., HLA) should be sought.
Collapse
Affiliation(s)
- Manuel Araújo
- Internal Medicine Department & Hospital Dia, Hospital São Francisco Xavier - Centro Hospitalar de Lisboa Ocidental, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Dário Ligeiro
- Immunogenetics Laboratory - Centro de Sangue e Transplantação de Lisboa, Instituto Português de Sangue e Transplantação, IP, Lisbon, Portugal
| | - Luís Costa
- Internal Medicine Department & Hospital Dia, Hospital São Francisco Xavier - Centro Hospitalar de Lisboa Ocidental, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Filipa Marques
- Internal Medicine Department & Hospital Dia, Hospital São Francisco Xavier - Centro Hospitalar de Lisboa Ocidental, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Helder Trindade
- Immunogenetics Laboratory - Centro de Sangue e Transplantação de Lisboa, Instituto Português de Sangue e Transplantação, IP, Lisbon, Portugal
| | - José Manuel Correia
- Pneumology Department, Hospital Egas Moniz - Centro Hospitalar de Lisboa Ocidental, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Candida Fonseca
- Internal Medicine Department & Hospital Dia, Hospital São Francisco Xavier - Centro Hospitalar de Lisboa Ocidental, NOVA Medical School, Faculdade de Ciências Médicas, Universidade Nova de Lisboa, Lisbon, Portugal
| |
Collapse
|
45
|
Diabetic Ketoacidosis as an Immune-related Adverse Event from Pembrolizumab in Non-Small Cell Lung Cancer. J Immunother 2018; 40:249-251. [PMID: 28557813 DOI: 10.1097/cji.0000000000000173] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Programmed cell death protein 1 pathway inhibitors are now routinely administered to patients with non-small cell lung cancer, and prompt recognition of immune-related adverse events is critical to managing serious drug toxicities. Here, we describe a 66-year-old man with no known history of diabetes who presented with diabetic ketoacidosis after receiving 3 doses of pembrolizumab for lung adenocarcinoma. Autoimmune diabetes is a rare but potentially life-threatening complication of programmed cell death protein 1 inhibitors.
Collapse
|
46
|
Cheema A, Makadia B, Karwadia T, Bajwa R, Hossain M. Autoimmune Diabetes Associated With Pembrolizumab: A Review of Published Case Reports. World J Oncol 2018; 9:1-4. [PMID: 29581809 PMCID: PMC5862076 DOI: 10.14740/wjon1085w] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 02/26/2018] [Indexed: 12/13/2022] Open
Abstract
The utility of immunotherapy, such as pembrolizumab, is becoming essential in the treatment of certain cancers. Pembrolizumab works through binding of programmed cell death 1 receptor that blocks the binding of the programmed cell death ligand 1 and is commonly used in non-small cell lung cancer and melanoma. Pembrolizumab has been reported to be associated with multiple adverse reactions such as pneumonitis, colitis, hepatitis, hypophysitis, hyperthyroidism, hypothyroidism, nephritis, and type 1 diabetes; however, pembrolizumab causing type 1 diabetes was only reported in 0.1% of the patients in clinical trials. A review of the literature generated 1,001 unique citations of which six reported cases of autoimmune diabetes associated with pembrolizumab were selected and compared. Review of the cases showed no sexual predilection and the average age of onset was 58 years old. The majority of the patients were treated for melanoma (5/6 cases), initially presented with diabetic ketoacidosis (4/6 cases), and had at one point taken ipilimumab (4/6 cases). There was no association found between the number of treatments received and the development of diabetes. With the increasing use of pembrolizumab in cancer treatment regular blood glucose monitoring during treatment, especially in patients who had also taken ipilimumab, may prevent the onset of this life-threatening complication.
Collapse
Affiliation(s)
- Anmol Cheema
- Jersey Shore University Medical Center, Neptune, NJ, USA
| | | | - Tejas Karwadia
- Jersey Shore University Medical Center, Neptune, NJ, USA
| | - Ravneet Bajwa
- Jersey Shore University Medical Center, Neptune, NJ, USA
| | | |
Collapse
|
47
|
Barroso-Sousa R, Ott PA, Hodi FS, Kaiser UB, Tolaney SM, Min L. Endocrine dysfunction induced by immune checkpoint inhibitors: Practical recommendations for diagnosis and clinical management. Cancer 2018; 124:1111-1121. [PMID: 29313945 DOI: 10.1002/cncr.31200] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 11/21/2017] [Accepted: 11/28/2017] [Indexed: 11/11/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy. However, because ICIs block coinhibitory molecules on T cells and other immune cells, unleashing them to mediate tumor cell killing, they also can disrupt the maintenance of immunological tolerance to self-antigens. Compared with chemotherapy, ICIs have a different toxicity profile, especially the occurrence of autoimmune-like manifestations against multiple organ systems, including endocrine glands, commonly referred to as immune-related adverse events. The aim of this review was to provide practical recommendations regarding the proper assessment and clinical management related to the new onset of endocrinopathies after the use of ICIs in patients with cancer. Cancer 2018;124:1111-21. © 2018 American Cancer Society.
Collapse
Affiliation(s)
| | - Patrick A Ott
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts
| | - Sara M Tolaney
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Le Min
- Division of Endocrinology, Diabetes and Hypertension, Brigham and Women's Hospital, Boston, Massachusetts
| |
Collapse
|
48
|
A case of pembrolizumab-induced type 1 diabetes mellitus presenting with severe diabetic ketoacidosis. JOURNAL OF CLINICAL AND TRANSLATIONAL ENDOCRINOLOGY CASE REPORTS 2017. [DOI: 10.1016/j.jecr.2017.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
49
|
|
50
|
Gauci ML, Laly P, Vidal-Trecan T, Baroudjian B, Gottlieb J, Madjlessi-Ezra N, Da Meda L, Madelaine-Chambrin I, Bagot M, Basset-Seguin N, Pages C, Mourah S, Boudou P, Lebbé C, Gautier JF. Autoimmune diabetes induced by PD-1 inhibitor-retrospective analysis and pathogenesis: a case report and literature review. Cancer Immunol Immunother 2017; 66:1399-1410. [PMID: 28634815 PMCID: PMC11028556 DOI: 10.1007/s00262-017-2033-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/12/2017] [Indexed: 12/16/2022]
Abstract
Anti-PD-1 antibody treatment is approved in advanced melanoma and provides median overall survival over 24 months. The main treatment-related side effects are immune-related adverse events, which include rash, pruritus, vitiligo, thyroiditis, diarrhoea, hepatitis and pneumonitis. We report a case of autoimmune diabetes related to nivolumab treatment. A 73-year-old man was treated in second line with nivolumab at 3 mg/kg every two weeks for metastatic melanoma. At 6 weeks of treatment, he displayed diabetic ketoacidosis. Nivolumab was withheld 3.5 weeks and insulin therapy was initiated, enabling a normalization of glycaemia and the disappearance of symptoms. Laboratory investigations demonstrated the presence of islet cell autoantibodies, while C-peptide was undetectable. Retrospective explorations on serum banked at week 0 and 3 months before the start of nivolumab, already showed the presence of autoantibodies, but normal insulin, C-peptide secretion and glycaemia. Partial response was obtained at month 3, and nivolumab was then resumed at the same dose. The clinical context and biological investigations before, at and after nivolumab initiation suggest the autoimmune origin of this diabetes, most likely induced by anti-PD-1 antibody in a predisposed patient. The role of PD-1/PD-L1 binding is well known in the pathogenesis of type 1 diabetes. Therefore, this rare side effect can be expected in a context of anti-PD-1 treatment. Glycaemia should be monitored during PD-1/PD-L1 blockade. The presence of autoantibodies before treatment could identify individuals at risk of developing diabetes, but systematic titration may not be relevant considering the rarity of this side effect.
Collapse
Affiliation(s)
- Marie-Léa Gauci
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France.
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France.
| | - Pauline Laly
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Tiphaine Vidal-Trecan
- AP-HP Diabetology Department, Lariboisière Hospital, Paris, France
- INSERM U1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Barouyr Baroudjian
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Jérémy Gottlieb
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Nika Madjlessi-Ezra
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Laetitia Da Meda
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Isabelle Madelaine-Chambrin
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
- AP-HP Pharmacology Department, Saint-Louis Hospital, Paris, France
| | - Martine Bagot
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Nicole Basset-Seguin
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Cécile Pages
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Samia Mourah
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
- AP-HP Pharmacogenomic Laboratory, Saint-Louis Hospital, Paris, France
| | - Philippe Boudou
- AP-HP Hormonology Department, Saint-Louis Hospital, Paris, France
- Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Céleste Lebbé
- AP-HP Dermatology Department, Saint-Louis Hospital, 1 avenue Claude Vellefaux, 75475, Paris Cedex 10, France
- INSERM U976, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| | - Jean-François Gautier
- AP-HP Diabetology Department, Lariboisière Hospital, Paris, France
- INSERM U1138, Université Paris Diderot-Paris VII, Sorbonne Paris Cité, Paris, France
| |
Collapse
|