1
|
Liu M, Ma Y, Hong Z, Yin Q, Gu J, Chen L, Zheng M. Long-term outcomes in patients with persistent EBV-DNA positivity after primary Epstein-Barr virus infection and clinical implications of rituximab therapy. Int Immunopharmacol 2025; 156:114695. [PMID: 40262252 DOI: 10.1016/j.intimp.2025.114695] [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: 07/24/2024] [Revised: 03/22/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND The long-term clinical outcomes of patients with persistent EBV-DNA positivity after primary EBV infection of B cells and the clinical value of rituximab in this group of patients are unknown. METHODS We retrospectively analyzed patients with primary EBV-infected B cells at our center from October 2016 to July 2023. They were divided into two groups based on the presence of infectious mononucleosis. To assess disease transformation under long-term EBV exposure and the impact of rituximab on EBV-DNA clearance, we followed up and monitored the EBV-DNA changes in the conventional observation treatment subgroup and the rituximab subgroup. RESULTS Seventy-one patients were included in this study, and 29 of them exhibited sustained EBV-DNA positivity for over 3 months. Among them, 20 patients were classified as infectious mononucleosis (IM) group, while 9 patients belonged to the non-infectious mononucleosis (NIM) group. In the IM group, 5 out of 7 patients treated with rituximab (71.4 %) became EBV-DNA negative within one month post-treatment, while 6 of the 13 patients (46.2 %) who did not undergo rituximab treatment still remained EBV-DNA positive for over 18 months. These results suggested that rituximab treatment significantly shortened the duration required for EBV-DNA clearance. In the NIM group, 5 of the 6 patients received rituximab had EBV-associated lymphoid tissue proliferation before treatment, which occurred approximately 7 months after EBV infection was detected. Following treatment, EBV-DNA became negative in all five patients within a median of 2 months. Conversely, the three patients who did not receive rituximab treatment in the NIM group, the longest duration of EBV-DNA positivity was 16 months. Disease progression or transformation was not observed in either group of patients. CONCLUSION Patients with primary EBV infection of B cells are unlikely to progress to malignant disease transformation despite persistent EBV-DNA positivity. Nonetheless, there is a potential risk of chronic lymphoproliferation. Rituximab treatment is a safe and efficient method for eliminating EBV-infected B cells and achieving rapid clearance of EBV-DNA.
Collapse
Affiliation(s)
- Min Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China; Department of Hematology, Jingzhou Hospital, Affiliated to Yangtze University, China
| | - Yaxian Ma
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Zetong Hong
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Qing Yin
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Jia Gu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China
| | - Miao Zheng
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, China.
| |
Collapse
|
2
|
Vincken R, Armendáriz-Martínez U, Ruiz-Sáenz A. ADCC: the rock band led by therapeutic antibodies, tumor and immune cells. Front Immunol 2025; 16:1548292. [PMID: 40308580 PMCID: PMC12040827 DOI: 10.3389/fimmu.2025.1548292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/27/2025] [Indexed: 05/02/2025] Open
Abstract
Antibody-dependent cellular cytotoxicity (ADCC) is a critical mechanism by which therapeutic antibodies leverage the immune system to target and eliminate cancer cells. The key agents of ADCC are natural killer (NK) cells, specifically targeting antibody-covered cancer cells through the CD16 receptor. While other immune cells and Fc receptors can contribute and enhance ADCC, NK cells and the CD16 receptor are crucial for the efficacy of cancer therapies such as trastuzumab, cetuximab and rituximab. Co-culture assays are essential for understanding the mechanisms of these therapies, overcoming resistance and optimizing novel therapeutic antibodies. This review highlights the importance of measuring ADCC to assess the efficacy of therapeutic antibodies. Here we also present the various in vitro models and assay methodologies available for studying ADCC, comparing the strengths and limitations of approaches like using PBMCs to better reflect real-life conditions or NK cell lines for standardization. It also covers different readouts for ADCC, either focusing on effector cells activation, including reporter and degranulation assays or in the target cell killing, including different molecule release assays, flow cytometry and immunofluorescence techniques. Selecting the best model for studying ADCC is crucial for the translational significance of therapeutic antibody research.
Collapse
Affiliation(s)
- Roos Vincken
- Department of Cell Biology, Erasmus University Medical Center Rotterdam, CN, Rotterdam, Netherlands
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Uxue Armendáriz-Martínez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
| | - Ana Ruiz-Sáenz
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
3
|
Jafari P, Hakimian D, Westerhoff M, Cheng J, Cao W, Kohnehshahri MN, Choi WT, Evaristo G, Graham RP, Liao X, Liu X, Pai RK, Salomao MA, Zhao L, Hart J, Micic D, Semrad CE, Alpert L. The Histologic Spectrum of Rituximab-Associated Common Variable Immunodeficiency-Like Enteropathy. Mod Pathol 2025; 38:100770. [PMID: 40222650 DOI: 10.1016/j.modpat.2025.100770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 03/09/2025] [Accepted: 03/30/2025] [Indexed: 04/15/2025]
Abstract
Rituximab (RTX) is a monoclonal anti-CD20 antibody widely used to treat B-cell neoplasms and autoimmune conditions. RTX has recently been linked to an enteropathy characterized by diarrhea, malabsorption, and hypogammaglobulinemia, closely resembling common variable immunodeficiency (CVID) enteropathy. We present the first dedicated histopathologic assessment of RTX-associated CVID-like enteropathy. Study inclusion criteria were the presence of diarrhea, weight loss, or other gastrointestinal symptoms in the setting of current/prior RTX use and associated hypogammaglobulinemia. Twenty-two patients (15 male:7 female; mean age at biopsy/resection, 63.4 years) across 9 tertiary medical centers met inclusion criteria and had small bowel (N = 20) and/or colon (N = 17) specimens (biopsies/resections) available for review; 71.4% of specimens dated from ≤5 years of last RTX dose. Cases were systematically evaluated by gastrointestinal pathologists at each institution. Key histologic features in the small bowel included sparse/absent lamina propria plasma cells (N = 10; 50%), intraepithelial lymphocytosis (N = 12; 60%), villous atrophy (N = 11; 55%), increased crypt apoptotic bodies (N = 6; 30%), and active inflammation (N = 5; 25%). Common features in the colon included sparse/absent plasma cells (N = 7; 41.2%), increased crypt apoptotic bodies (N = 7; 41.2%), active inflammation (N = 5; 29.4%), and intraepithelial lymphocytosis (N = 4; 23.5%). Goblet cell loss was appreciated in small bowel and/or colon specimens from 2 patients. Follow-up biopsies (interval, 2 months to 4 years) were available for 7 patients and largely recapitulated the histology of the index specimens, though 1 patient demonstrated improvement in villous blunting and intraepithelial lymphocytosis. In summary, the histologic spectrum of post-RTX CVID-like enteropathy encompasses lamina propria plasma cell depletion, increased crypt apoptotic bodies, small bowel villous atrophy, and goblet cell loss. While the underlying pathophysiology remains uncertain, the clinicopathologic picture may reflect post-RTX B-cell/plasma cell impairment. Although histologic findings may be subtle and variable, pathologists should be aware of this entity and should seek a history of RTX use in patients whose biopsies exhibit these CVID enteropathy-like features.
Collapse
Affiliation(s)
- Pari Jafari
- Department of Pathology, University of Chicago Medicine, Chicago, Illinois.
| | - David Hakimian
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, University of Chicago Medicine, Chicago, Illinois
| | - Maria Westerhoff
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Jerome Cheng
- Department of Pathology, Michigan Medicine, University of Michigan, Ann Arbor, Michigan
| | - Wenqing Cao
- Department of Pathology, New York University Langone Health, New York, New York
| | | | - Won-Tak Choi
- Department of Pathology, University of California San Francisco, San Francisco, California
| | - Gertruda Evaristo
- Department of Pathology, McGill University Health Centre, Montreal, Quebec
| | - Rondell P Graham
- Department of Laboratory Medicine and Pathology, Mayo Clinic Minnesota, Rochester, Minnesota
| | - Xiaoyan Liao
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York
| | - Xiuli Liu
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, Missouri
| | - Rish K Pai
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Marcela A Salomao
- Department of Laboratory Medicine and Pathology, Mayo Clinic Arizona, Phoenix, Arizona
| | - Lei Zhao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - John Hart
- Department of Pathology, University of Chicago Medicine, Chicago, Illinois
| | - Dejan Micic
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, University of Chicago Medicine, Chicago, Illinois
| | - Carol E Semrad
- Section of Gastroenterology, Hepatology, and Nutrition, Department of Internal Medicine, University of Chicago Medicine, Chicago, Illinois
| | - Lindsay Alpert
- Department of Pathology, University of Chicago Medicine, Chicago, Illinois
| |
Collapse
|
4
|
Gao P, Zhang Y, Ma J, Zhang Y. Immunotherapy in chronic lymphocytic leukemia: advances and challenges. Exp Hematol Oncol 2025; 14:53. [PMID: 40211406 PMCID: PMC11984025 DOI: 10.1186/s40164-025-00644-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized as a clonal proliferation of mature B lymphocytes with distinct immunophenotypic traits, predominantly affecting the middle-aged and elderly population. This condition is marked by an accumulation of lymphocytes within the peripheral blood, bone marrow, spleen, and lymph nodes. The associated immune dysregulation predisposes CLL patients to a higher risk of secondary malignancies and infections, which significantly contribute to morbidity and mortality rates. The advent of immunotherapy has revolutionized the prognosis of CLL, advancing treatment modalities and offering substantial benefits to patient outcomes. This review endeavors to synthesize and scrutinize the efficacy, merits, and limitations of the current immunotherapeutic strategies for CLL. The aim is to inform the selection of optimal treatment regimens tailored to individual patient needs. Furthermore, the review juxtaposes various therapeutic combinations to elucidate the comparative advantages of each approach, with the ultimate objective of enhancing patient prognosis and quality of life.
Collapse
Affiliation(s)
- Pan Gao
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Yang Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Jun Ma
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No.324, Jingwu Road, Jinan, Shandong, 250021, China.
| |
Collapse
|
5
|
Rendell M. Pharmacotherapy of type 1 diabetes - part 3: tomorrow. Expert Opin Pharmacother 2025; 26:535-550. [PMID: 40056035 DOI: 10.1080/14656566.2025.2468906] [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: 12/16/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 03/27/2025]
Abstract
INTRODUCTION The last 100 years have seen type 1 diabetes, a previously fatal disease, transformed by the administration of exogenous insulin. AREAS COVERED A standard literature search using the Google and Microsoft search engines and PubMed was performed. The development of synthetic insulins with varying onsets and duration of action improved glucose control, essential to mitigate the microvascular and macrovascular consequences of diabetes. Today insulin pumps guided by continuous glucose monitors are approaching the objective of normalized glucose levels. The area of greatest development is now in attempting to suppress the immune process which results in progressive destruction of the beta cell. It is possible to identify family members of patients with type 1 diabetes who may eventually develop the disease by measuring several beta cell antibodies. Very recently teplizumab, a CD3 inhibitor, has been approved to delay the onset of hyperglycemia in these individuals. EXPERT OPINION The future will see progress in immunosuppression, possibly using specific CAR-Treg cells directed at the beta cell antigens which trigger the immune process. In parallel, stem cell-derived beta cells may eventually make it possible to replace lost beta cells, resulting in a true cure for type 1 diabetes.
Collapse
Affiliation(s)
- Marc Rendell
- The Association of Diabetes Investigators, Omaha, NE, USA
- The Rose Salter Medical Research Foundation, Newport Coast, CA, USA
| |
Collapse
|
6
|
Trevisani GFM, Bento VFV, Salles Rosa Neto N. Late-Onset Neutropenia Induced by Rituximab in Rheumatic Diseases: A Report of Two Cases of Severe Presentation and a Literature Review. Cureus 2025; 17:e80074. [PMID: 40190867 PMCID: PMC11970875 DOI: 10.7759/cureus.80074] [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] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
Rituximab, a chimeric monoclonal antibody targeting the CD20 antigen on B cells, is widely used in oncological and immune-mediated diseases. However, late-onset neutropenia can occur, even in patients receiving concomitant immunosuppressants or chemotherapeutics, necessitating therapeutic adjustments. The development of neutropenia with rituximab monotherapy reinforces the relationship, but the exact pharmacological mechanism is still unknown. We report two cases of late-onset neutropenia after rituximab therapy: the first case is related to a woman with rheumatoid arthritis and anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis overlap and a history of alveolar hemorrhage; the second case concerns a woman with systemic lupus erythematosus and neurological manifestations. Both patients were hospitalized for febrile neutropenia, an unusual complication, and subsequently recovered after treatment with antibiotics and granulocyte colony-stimulating factor. Rituximab was discontinued. It is essential for rheumatologists to recognize and monitor for late-onset neutropenia during and after rituximab treatment, as early detection and intervention can prevent severe complications. The heterogeneity in clinical course observed in reported cases underscores the complexity of the condition and the impact on patient safety. The feasibility of resuming rituximab treatment after late-onset neutropenia requires careful evaluation.
Collapse
Affiliation(s)
| | | | - Nilton Salles Rosa Neto
- Center for Rare and Immune Disorders, Hospital Nove de Julho, São Paulo, BRA
- Rheumatology, Universidade Santo Amaro, São Paulo, BRA
| |
Collapse
|
7
|
Zhang J, Hu M, Wang C, Guo S. Successful sequential therapy with rituximab and telitacicept in refractory Anti-NMDA receptor encephalitis and MOG-associated demyelination: a case report and literature review. Front Immunol 2025; 16:1509143. [PMID: 39981240 PMCID: PMC11839722 DOI: 10.3389/fimmu.2025.1509143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/22/2025] [Indexed: 02/22/2025] Open
Abstract
Clinical management of the rare and complex overlapping syndrome of MOG-antibody disease and anti-NMDAR encephalitis (MNOS), which has an uncertain pathogenesis and a high risk of recurrence, is highly challenging. We describe the case of a 19 years-old female patient, who first complained of headache, fever, and irritability. After that, she experienced frequent seizures and mood disorders. The diagnosis of MNOS was verified through antibody tests and imaging. For the patient, intravenous immunoglobulin and high-dose methylprednisolone were effective as first-line immunotherapy. Long-term immunotherapy with oral prednisone and mycophenolate mofetil was used to prevent relapses. However, over six years, the patient had five relapses when the mycophenolate mofetil dosage was reduced. The patient's condition stabilized after taking rituximab as second-line immunotherapy, with less than 1% of total lymphocytes being CD19+ cells. Eleven months later, the plasmablast ratio increased, and patients experienced new symptoms such as bilateral optic neuritis. After that, the patient got telitacicept injections regularly for 13 months, during which time her symptoms subsided, and there were no adverse effects or relapses. This case suggests that telitacicept may be a viable adjunct or sequential therapy option for the depletion of B cells in MNOS.
Collapse
Affiliation(s)
| | | | - Chunjuan Wang
- Department of Neurology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| | - Shougang Guo
- Department of Neurology, Shandong Provincial Hospital Affiliated with Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
8
|
Nakanoh H, Tsuji K, Morimoto S, Fukushima K, Iwamuro M, Uchida HA, Wada J. A case of membranous nephropathy complicated by Cronkhite-Canada syndrome successfully treated with mizoribine. CEN Case Rep 2025; 14:72-80. [PMID: 38954395 PMCID: PMC11785862 DOI: 10.1007/s13730-024-00908-9] [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: 02/29/2024] [Accepted: 06/21/2024] [Indexed: 07/04/2024] Open
Abstract
Cronkhite-Canada syndrome (CCS) is a non-hereditary disorder characterized by non-neoplastic hamartomatous gastrointestinal polyposis, hair loss, nail atrophy, hyperpigmentation, and diarrhea. While the relationship between CCS and nephritis remains unclear, seven cases of nephritis complicated by CCS have been reported to date, all of which were membranous nephropathy (MN). A 57-year-old man presented with taste disturbance, hair loss, nail plate atrophy, skin pigmentation, and frequent diarrhea. Endoscopic findings showed multiple polyposis of the stomach and large intestine. Given the above, he was diagnosed with CCS. The symptoms gradually improved with prednisolone treatment, although urinary protein and hypoproteinemia appeared during the tapering of prednisolone. He was diagnosed with MN using a renal biopsy, and immunofluorescence microscopy with IgG subclass staining showed predominantly diffuse granular capillary wall staining of IgG4. The cause of secondary MN was not found, including malignant tumors. Nephrotic-range proteinuria persisted despite treatment with prednisolone and cyclosporine. Additional treatment with mizoribine resulted in incomplete remission type 1 of nephrotic syndrome, suggesting that mizoribine may be a treatment option for patients with CCS with steroid-resistant MN. Considering a high prevalence of hypoproteinemia due to chronic diarrhea and protein-losing enteropathy in patients with CCS, proteinuria might be overlooked; thus, follow-up urinalysis would be recommended in patients with CCS.
Collapse
Affiliation(s)
- Hiroyuki Nakanoh
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Kenji Tsuji
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan.
| | - Shiho Morimoto
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Kazuhiko Fukushima
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| | - Masaya Iwamuro
- Department of Gastroenterology and Hepatology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Haruhito A Uchida
- Department of Chronic Kidney Disease and Cardiovascular Disease, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology and Metabolism, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Okayama, 700-8558, Japan
| |
Collapse
|
9
|
Salah AN, Hashem AH, Zaki MB, Abulsoud AI, Atta AM, Elkalla WS, Moustafa HAM, El-Dakroury WA, El-Tokhy FS, ElBoghdady JA, Rizk NI, Abdel Mageed SS, Mohammed OA, Abdel-Reheim MA, Alghamdi HO, Doghish AS. Targeted Therapies: The Role of Monoclonal Antibodies in Disease Management. J Biochem Mol Toxicol 2025; 39:e70163. [PMID: 39887821 DOI: 10.1002/jbt.70163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 12/28/2024] [Accepted: 01/20/2025] [Indexed: 02/01/2025]
Abstract
Monoclonal antibodies (mAbs) are a key class of biotherapeutic medicines used to treat a wide range of diseases, such as cancer, infectious diseases, autoimmune disorders, cardiovascular diseases, and hemophilia. They can be engineered for greater effectiveness and specific applications while maintaining their structural elements for immune targeting. Traditional immunoglobulin treatments have limited therapeutic uses and various adverse effects. That makes mAbs show rapid growth in the pharmaceutical market, with over 250 mAbs in clinical studies. Although mAbs offer higher specificity, they are less effective against complex antigens. They have become essential in treating diseases with limited medical options, providing innovative solutions that improve patients' quality of life through increasing survival rates, shortening the length of stay in hospitals with an improved treatment outcome, and reducing side effects. This review outlines the mechanisms, applications, and advancements of mAbs, highlighting their transformative role in modern medicine and their potential to shape future therapeutic interventions.
Collapse
Affiliation(s)
- Akram N Salah
- Microbiology and Immunology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Amr H Hashem
- Botany and Microbiology Department, Faculty of Science, Al-Azhar University, Nasr City, 11884, Egypt
| | - Mohamed Bakr Zaki
- Department of Biochemistry, Faculty of Pharmacy, University of Sadat City, Menofia, 32897, Egypt
- Department of Biochemistry, Faculty of Pharmacy, Menoufia National University, km Cairo-Alexandria Agricultural Road, Menofia, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, 11785, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| | - Asmaa M Atta
- Pharmaceutical Chemistry Department, School of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Wagiha S Elkalla
- Microbiology and Immunology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Hebatallah Ahmed Mohamed Moustafa
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Walaa A El-Dakroury
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Fatma Sa'eed El-Tokhy
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Jasmine A ElBoghdady
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Nehal I Rizk
- Department of Biochemistry, Faculty of Pharmacy and Drug Technology, Egyptian Chinese University, Cairo, 11786, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Cairo, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | | | - Huda O Alghamdi
- College of Medicine, University of Bisha, Bisha, 61922, Saudi Arabia
| | - Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, 11829, Egypt
- Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, 11231, Cairo, Egypt
| |
Collapse
|
10
|
Peng H, Martens S, Uvyn A, Chen Y, Zhong Z, Louage B, De Geest BG. Exploration of Solid Phase Peptoid Synthesis for the Design of Trifunctional Hapten-Lipid-TLR7/8 Agonist Antibody-Recruiting Oligomers That Combine Innate Effector with Innate Activation Function. ACS APPLIED BIO MATERIALS 2025; 8:177-188. [PMID: 39288003 DOI: 10.1021/acsabm.4c00825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The strategic engagement of innate immunity is a promising avenue for cancer treatment. Antibody-recruiting molecules (ARMs) direct endogenous antibodies to target tumor sites, eliciting innate immune effector killing responses. In this study, we report the synthesis of ARMs by employing solid-phase peptoid synthesis to construct three libraries of antibody-recruiting oligomers. Using dinitrophenyl (DNP) as a model hapten and alkyl lipid chains for cell surface anchoring, we tailored oligomers with variations in valency and spatial configuration. Among these, an oligomer design featuring DNP connected to the oligomer backbone through an extended PEG linker and flanked by two lipid motifs emerged as the most effective in antibody recruitment in vitro. This oligomer was further functionalized to include an imidazoquinoline, creating a trifunctional hapten-lipid-TLR7/8 agonist oligomer, and a parallel variant was conjugated with rhodamine, resulting in a trifunctional hapten-lipid-dye oligomer. Upon intratumorally administration in a murine model, these oligomers induced localized immune activation within tumors. Subsequent ex vivo analysis of single-cell suspensions from excised tumors confirmed the enhanced binding of anti-DNP antibodies. These findings underscore the potential of custom-designed ARMs in orchestrating precise immune-mediated tumor targeting and highlight the adaptability of solid-phase synthesis in oligomer design for the design of multifunctional antibody recruiting molecules.
Collapse
Affiliation(s)
- Haixia Peng
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Steven Martens
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Annemiek Uvyn
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Yong Chen
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Zifu Zhong
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Benoit Louage
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| | - Bruno G De Geest
- Department of Pharmaceutics, Ghent University, Ghent 9000, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Ghent 9000, Belgium
| |
Collapse
|
11
|
Zhang C, Liu H. Advancements and Future Directions of Dual-Target Chimeric Antigen Receptor T-Cell Therapy in Preclinical and Clinical Studies. J Immunol Res 2025; 2025:5845167. [PMID: 39844819 PMCID: PMC11753851 DOI: 10.1155/jimr/5845167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
In recent years, chimeric antigen receptor T-cell (CAR-T) therapy has made groundbreaking progress in the treatment of various cancer types, particularly hematological malignancies. In the meantime, various preclinical and clinical studies have extensively explored dual-target CAR-T therapies which can be designed to recognize two antigens simultaneously based on the immunophenotype of tumor cells. Compared with single-target CAR-T approach, dual-target CAR-T therapies demonstrate varying degrees of superior antitumor CAR effects, prevent antigen escape and relapse, reduce on-target off-tumor effects, and ensure durable responses in different types of cancer. These advantages highlight the potential future prospects in this field, showing varying degrees of advancement in preclinical and clinical studies. Herein, we aimed to review different dual-target CAR-T studies conducted on a wide range of tumor models, summarizing the selection of target combinations, the efficacy and safety demonstrated in preclinical and clinical settings, the existing limitations, and the potential future directions of this promising therapeutic strategy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/trends
- Immunotherapy, Adoptive/adverse effects
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Animals
- Neoplasms/therapy
- Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Antigens, Neoplasm/immunology
- Clinical Trials as Topic
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Treatment Outcome
Collapse
Affiliation(s)
- Chenyun Zhang
- School of Medicine, University of Tsinghua, Beijing, China
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Haizhou Liu
- Division of Hematology-Oncology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Kazama R, Ishikawa R, Sakai S. Development of Hemispherical 3D Models of Human Brain and B Cell Lymphomas Using On-Chip Cell Dome System. Bioengineering (Basel) 2024; 11:1303. [PMID: 39768123 PMCID: PMC11727638 DOI: 10.3390/bioengineering11121303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Lymphocytes are generally non-adherent. This makes it challenging to fabricate three-dimensional (3D) structures mimicking the three-dimensional lymphoma microenvironment in vivo. This study presents the fabrication of a hemispherical 3D lymphoma model using the on-chip Cell Dome system with a hemispherical cavity (1 mm in diameter and almost 300 µm in height). Both the human brain lymphoma cell line (TK) and human B cell lymphoma cell line (KML-1) proliferated and filled the cavities. Hypoxic regions were observed in the center of the hemispherical structures. CD19 expression did not change in either cell line, while CD20 expression was slightly upregulated in TK cells and downregulated in KML-1 cells cultured in the Cell Dome compared to those cultured in two-dimensional (2D) flasks. In addition, both TK and KML-1 cells in the hemispherical structures exhibited higher resistance to doxorubicin than those in 2D flasks. These results demonstrate the effectiveness of the on-chip Cell Dome for fabricating 3D lymphoma models and provide valuable insights into the study of lymphoma behavior and the development of new drugs for lymphoma treatment.
Collapse
Affiliation(s)
| | | | - Shinji Sakai
- Graduate School of Engineering Science, Osaka University, 1-3 Machikaneyama, Toyonaka 560-8531, Osaka, Japan; (R.K.); (R.I.)
| |
Collapse
|
13
|
Hu G, Zhao X, Wang Y, Zhu X, Sun Z, Yu X, Wang J, Liu Q, Zhang J, Zhang Y, Yang J, Chang T, Ruan Z, Lv J, Gao F. Advances in B Cell Targeting for Treating Muscle-Specific Tyrosine Kinase-Associated Myasthenia Gravis. Immunotargets Ther 2024; 13:707-720. [PMID: 39678139 PMCID: PMC11646387 DOI: 10.2147/itt.s492062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/30/2024] [Indexed: 12/17/2024] Open
Abstract
Myasthenia gravis (MG) is a typical autoimmune disease of the nervous system. It is characterized by skeletal muscle weakness and fatigue due to impaired neuromuscular junction transmission mediated by IgG autoantibodies. Muscle-specific receptor tyrosine kinase-associated MG (MuSK-MG), a rare and severe subtype of MG, is distinguished by the presence of anti-MuSK antibodies; it responds poorly to traditional therapies. Recent research on MuSK-MG treatment has focused on specific targeted therapies. Since B cells play a critical pathogenic role in producing autoantibodies and inflammatory mediators, they are often considered the preferred target for treating MuSK-MG. Currently, various B cell-targeted drugs have been developed to treat MuSK-MG; they have shown good therapeutic effects. This review explores the evolving landscape of B cell-targeted therapies in MuSK-MG, focusing on their mechanisms, efficacy, and safety, and the current limitations associated with their use. We discuss current B cell-targeted therapies aimed at depleting or modulating B cells via both direct and indirect approaches. Furthermore, we focus on novel and promising strategies such as Chimeric Autoantibody Receptor T cell therapy, which explicitly targets MuSK-specific B cells without compromising general humoral immunity. Finally, this review provides an outlook on the potential benefits and limitations of B cell-targeted therapy in developing new therapies for MuSK-MG. We conclude by discussing future research efforts needed to optimize these therapies, expand treatment options, and improve long-term outcomes in MuSK-MG management.
Collapse
Affiliation(s)
- Guanlian Hu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xue Zhao
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yiren Wang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiaoyan Zhu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Zhan Sun
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xiaoxiao Yu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
- BGI College, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jiahui Wang
- Department of Encephalopathy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Qian Liu
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Jing Zhang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yingna Zhang
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Junhong Yang
- Department of Encephalopathy, First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, People’s Republic of China
| | - Ting Chang
- Department of Neurology, Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Zhe Ruan
- Department of Neurology, Second Affiliated Hospital, Air Force Medical University, Xi’an, People’s Republic of China
| | - Jie Lv
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Feng Gao
- Department of Neuroimmunology, Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| |
Collapse
|
14
|
Nguyen OTP, Lara S, Ferro G, Peipp M, Kleinau S. Rituximab-IgG2 is a phagocytic enhancer in antibody-based immunotherapy of B-cell lymphoma by altering CD47 expression. Front Immunol 2024; 15:1483617. [PMID: 39712032 PMCID: PMC11659266 DOI: 10.3389/fimmu.2024.1483617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 11/14/2024] [Indexed: 12/24/2024] Open
Abstract
Antibody-dependent cellular phagocytosis (ADCP) by monocytes and macrophages contributes significantly to the efficacy of many therapeutic monoclonal antibodies (mAbs), including anti-CD20 rituximab (RTX) targeting CD20+ B-cell non-Hodgkin lymphomas (NHL). However, ADCP is constrained by various immune checkpoints, notably the anti-phagocytic CD47 molecule, necessitating strategies to overcome this resistance. We have previously shown that the IgG2 isotype of RTX induces CD20-mediated apoptosis in B-cell lymphoma cells and, when combined with RTX-IgG1 or RTX-IgG3 mAbs, can significantly enhance Fc receptor-mediated phagocytosis. Here, we report that the apoptotic effect of RTX-IgG2 on lymphoma cells contributes to changes in the tumor cell's CD47 profile by reducing its overall expression and altering its surface distribution. Furthermore, when RTX-IgG2 is combined with other lymphoma-targeting mAbs, such as anti-CD59 or anti-PD-L1, it significantly enhances the ADCP of lymphoma cells compared to single mAb treatment. In summary, RTX-IgG2 acts as a potent phagocytic enhancer by promoting Fc-receptor mediated phagocytosis through apoptosis and reduction of CD47 in CD20+ malignant B-cells. RTX-IgG2 represents a valuable therapeutic component in enhancing the effectiveness of different mAbs targeting B-cell NHL.
Collapse
Affiliation(s)
- Oanh T. P. Nguyen
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Sandra Lara
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Giovanni Ferro
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Matthias Peipp
- Division of Antibody-Based Immunotherapy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sandra Kleinau
- Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
15
|
Xiao R, Zhao W, Lin W, Xiao Y, Ren J, Zhou Y, Meng W, Bi E, Jiang L. Bendamustine-rituximab elicits dual tumoricidal and immunomodulatory responses via cGAS-STING activation in diffuse large B-cell lymphoma. J Immunother Cancer 2024; 12:e009212. [PMID: 39521616 PMCID: PMC11551994 DOI: 10.1136/jitc-2024-009212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/13/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Bendamustine-rituximab (BR) therapy stands out as a promising alternative for elderly patients with diffuse large B-cell lymphoma (DLBCL), demonstrating notable efficacy when conventional regimens pose challenges. Despite its clinical success, the intricate mechanisms underlying BR therapy have remained elusive. METHODS DLBCL cell lines were used to investigate the mechanism of BR therapy in vitro. RNA-seq and Western blot were used to explore the target pathways of BR therapy. STING was knocked out using Crispr-cas9 and inhibited using H-151 to investigate its role in BR therapy. Bulk RNA-seq and single-cell RNA-seq data from patients were analyzed to investigate the association between STING and pyroptosis pathways, validated using STING downregulated cells. Flow cytometry, transwell experiments and co-culture experiments were performed to investigate the inflammatory phenotype of DLBCL cells after BR treatment and its effect on T-cell recruitment and activation. RESULTS This study elucidates that BR elicits direct tumoricidal effects by promoting apoptosis and inducing cell cycle arrest. The synergistic impact with rituximab is further potentiated by complement addition, demonstrating the pivotal role of in vivo antibody-dependent cellular cytotoxicity. Moreover, our investigation reveals that, through a cGAS-STING-dependent pathway, prolonged exposure to BR induces pyroptosis in DLBCL cells. Activation of the cGAS-STING pathway by BR therapy triggers the release of inflammatory factors and upregulates major histocompatibility complex molecules, shaping an immunologically hot tumor microenvironment. CONCLUSIONS This unique dual influence not only directly targets DLBCL cells but also engages the patient's immune system, paving the way for innovative combination therapies. The study provides comprehensive insights into the multifaceted actions of BR in DLBCL, offering a foundation for refined and personalized treatment strategies in elderly patients.
Collapse
Affiliation(s)
- Ruipei Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenli Zhao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Yudian Xiao
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Jie Ren
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Yang Zhou
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
| | - Enguang Bi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, Guangdong, China
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Ling Jiang
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
16
|
Wu Y, Sun X, Kang K, Yang Y, Li H, Zhao A, Niu T. Hemophagocytic lymphohistiocytosis: current treatment advances, emerging targeted therapy and underlying mechanisms. J Hematol Oncol 2024; 17:106. [PMID: 39511607 PMCID: PMC11542428 DOI: 10.1186/s13045-024-01621-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a rapidly progressing, life-threatening syndrome characterized by excessive immune activation, often presenting as a complex cytokine storm. This hyperactive immune response can lead to multi-organ failure and systemic damage, resulting in an extremely short survival period if left untreated. Over the past decades, although HLH has garnered increasing attention from researchers, there have been few advancements in its treatment. The cytokine storm plays a crucial role in the treatment of HLH. Investigating the detailed mechanisms behind cytokine storms offers insights into targeted therapeutic approaches, potentially aiding in early intervention and improving the clinical outcome of HLH patients. To date, there is only one targeted therapy, emapalumab targeting interferon-γ, that has gained approval for primary HLH. This review aims to summarize the current treatment advances, emerging targeted therapeutics and underlying mechanisms of HLH, highlighting its newly discovered targets potentially involved in cytokine storms, which are expected to drive the development of novel treatments and offer fresh perspectives for future studies. Besides, multi-targeted combination therapy may be essential for disease control, but further trials are required to determine the optimal treatment mode for HLH.
Collapse
Affiliation(s)
- Yijun Wu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xu Sun
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Kai Kang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuqi Yang
- West China School of Medicine, Sichuan University, Chengdu, Sichuan, China
| | - He Li
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Zhao
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Ting Niu
- Department of Hematology, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- State Key Laboratory of Biotherapy, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
17
|
Miyamoto K, Ohmoto A, Yoneoka D, Rahman MO, Ota E. First-line therapy for high-risk people with chronic lymphocytic leukemia: a network meta-analysis. Cochrane Database Syst Rev 2024; 10:CD015169. [PMID: 39474946 PMCID: PMC11523224 DOI: 10.1002/14651858.cd015169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
OBJECTIVES This is a protocol for a Cochrane Review (intervention). The objectives are as follows: Primary objective: to assess the benefits and harms of currently recommended regimens as the first-line therapy in high-risk people with chronic lymphocytic leukemia, using network meta-analysis Secondary objectives: to assess whether the benefits and harms of the recommended regimens differ according to sex, Rai stage, or genetic mutation status to estimate the ranking of treatments for overall survival, progression-free survival, objective response rate, complete response rate, minimal residual disease, and serious adverse events to estimate the overall rate of adverse events and serious adverse events.
Collapse
MESH Headings
- Female
- Humans
- Male
- Adenine/analogs & derivatives
- Adenine/therapeutic use
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Neoplasm, Residual
- Randomized Controlled Trials as Topic
Collapse
Affiliation(s)
- Kenichi Miyamoto
- Department of Health Policy and Informatics, Graduate School of Medicine, Institute of Science Tokyo, Tokyo, Japan
| | - Akihiro Ohmoto
- Department of Medical Oncology, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Daisuke Yoneoka
- Division of Biostatistics and Bioinformatics, Graduate School of Public Health, St. Luke's International University, Tokyo, Japan
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
| | - Md Obaidur Rahman
- Center for Surveillance, Immunization, and Epidemiologic Research, National Institute of Infectious Diseases, Tokyo, Japan
- Department of Global Health Nursing, Graduate School of Nursing Science, St. Luke's International University, Tokyo, Japan
- Center for Evidence-Based Medicine and Clinical Research, Dhaka, Bangladesh
| | - Erika Ota
- Department of Global Health Nursing, Graduate School of Nursing Science, St. Luke's International University, Tokyo, Japan
| |
Collapse
|
18
|
Lee I, Byun BH, Kim BI, Choi CW, Kang HJ, Kang CS, Woo SK, Lee KC, Kang JH, Lim I. Evaluating 64 Cu-DOTA-rituximab as a PET agent in patients with B-cell lymphoma: a head-to-head comparison with 18 F-fluorodeoxyglucose PET/computed tomography. Nucl Med Commun 2024; 45:865-873. [PMID: 39155810 DOI: 10.1097/mnm.0000000000001889] [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: 08/20/2024]
Abstract
BACKGROUND This study aimed to evaluate the biodistribution of 64 Cu-DOTA-rituximab and its diagnostic feasibility for lymphoma using CD20-targeted 64 Cu-DOTA-rituximab PET/computed tomography (PET/CT). METHODS A prospective study involving six patients diagnosed with lymphoma was conducted between January 2022 and January 2023. These patients underwent 18 F-fluorodeoxyglucose ( 18 F-FDG) and 64 Cu-DOTA-rituximab PET/CT scans. 64 Cu-DOTA-rituximab PET/CT images were acquired at 1, 24, and 48 h after administering 64 Cu-DOTA-rituximab to assess the biodistribution and dosimetry over time. The observed lymph nodes were categorized into specific regions, including cervical and supraclavicular, axillary and infraclavicular, mediastinal, hilar, abdominal paraaortic and retroperitoneal, iliac, mesenteric, and inguinal regions, to compare the diagnostic ability of 18 F-FDG and 64 Cu-DOTA-rituximab PET/CT in detecting lymphoma lesions. Furthermore, the tumor-to-background ratio was calculated and compared with the maximum standardized uptake (SUV max ) of the tumors and the mean standardized uptake (SUV mean ) of normal organs. Internal radiation dosimetry was determined using the OLINDA/EXM software. RESULTS 64 Cu-DOTA-rituximab uptake in lymph nodes associated with lymphoma progressively increased from 1 to 48 h after injection. In contrast, 64 Cu-DOTA-rituximab uptake in normal organs, such as blood, lung, kidney, bladder, muscle, bone, and brain, decreased over time, whereas it increased in the liver and spleen. When it comes to the comparison between 64 Cu-DOTA-rituximab and 18 F-FDG, the SUV max of tumors was higher on 64 Cu-DOTA-rituximab PET/CT (18.1 ± 8.3) than on 18 F-FDG PET/CT (5.2 ± 1.5). Additionally, the tumor-to-background ratio, measured using the SUV mean of normal muscles, was higher on 64 Cu-DOTA-rituximab PET/CT (55.7 ± 31.0) than on 18 F-FDG PET/CT (8.6 ± 2.8). No adverse events related to 64 Cu-DOTA-rituximab injection were reported. CONCLUSION The results of this study demonstrate the feasibility of using 64 Cu-DOTA-rituximab PET/CT to evaluate the CD20 expression. The increased 64 Cu-DOTA-rituximab uptake in lymph nodes associated with tumors, higher SUV max , and tumor-to-muscle ratios observed with 64 Cu-DOTA-rituximab PET/CT compared with 18 F-FDG PET/CT, highlight the diagnostic potential of this imaging modality.
Collapse
Affiliation(s)
| | | | | | | | - Hye Jin Kang
- Division of Hematology/Oncology, Department of Internal Medicine, Korea Cancer Center Hospital and
| | - Chi Soo Kang
- Division of Applied RI, Research Institute of Radiological and Medical Sciences, Korea Institutes of Radiological and Medical Sciences, Seoul, Korea
| | - Sang-Keun Woo
- Division of Applied RI, Research Institute of Radiological and Medical Sciences, Korea Institutes of Radiological and Medical Sciences, Seoul, Korea
| | - Kyo Chul Lee
- Division of Applied RI, Research Institute of Radiological and Medical Sciences, Korea Institutes of Radiological and Medical Sciences, Seoul, Korea
| | - Joo Hyun Kang
- Division of Applied RI, Research Institute of Radiological and Medical Sciences, Korea Institutes of Radiological and Medical Sciences, Seoul, Korea
| | | |
Collapse
|
19
|
Li Q, Zhang K, Yu Y, Yu Z, Xu J, Shen W, Zhang L, Qu A, Liang H. TFAB002s, novel CD20-targeting T cell-dependent bispecific Fab-FabCH3 antibodies, exhibit potent antitumor efficacy against malignant B-cell lymphoma. PLoS One 2024; 19:e0310889. [PMID: 39321199 PMCID: PMC11423992 DOI: 10.1371/journal.pone.0310889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/09/2024] [Indexed: 09/27/2024] Open
Abstract
B-cell lymphoma, clinically, comprises a heterogeneous group of malignancies that encompass various subtypes. CD20 is an optimal target for therapeutic antibodies in B-cell lymphoma immunotherapy since approximately 90% of B-cell malignancies typically exhibit CD20 expression on their surface, while its presence is limited in normal tissues. In this study, we have developed a series of novel non-IgG-like T cell-dependent bispecific antibodies by constructing Fab-FabCH3, referred to as Tandem Antigen-binding Fragment 002 (TFAB002), which specifically target CD20 for the treatment of malignant B-cell lymphoma. TFAB002s display strong binding affinity with CD20 and moderate binding affinity with CD3, thereby triggering target-specific T-cell activation, cytokine release, and tumor cell lysis in vitro. Furthermore, TFAB002s exhibit potent cytotoxicity against B-cell malignancies that express varying levels of CD20. Besides, the TFAB002s show potent pharmacodynamic activity in vivo in the WIL2-S cells CDX mouse model. Collectively, these results underscore the potential of TFAB002s as a highly promising therapeutic approach for selectively depleting CD20-positive B cells, thereby warranting further clinical evaluation as a viable treatment option for CD20-expressing B-cell malignancies.
Collapse
Affiliation(s)
- Qinghong Li
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Kunming Zhang
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Yao Yu
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Zeng Yu
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Jingyi Xu
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Wenyan Shen
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Lin Zhang
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Aidong Qu
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| | - Hongyuan Liang
- No.1 Research Laboratory, Shanghai Institute of Biological Products Co., Ltd., Shanghai, China
| |
Collapse
|
20
|
Campagnoli LIM, Ahmad L, Marchesi N, Greco G, Boschi F, Masi F, Mallucci G, Bergamaschi R, Colombo E, Pascale A. Disclosing the Novel Protective Mechanisms of Ocrelizumab in Multiple Sclerosis: The Role of PKC Beta and Its Down-Stream Targets. Int J Mol Sci 2024; 25:8923. [PMID: 39201609 PMCID: PMC11354964 DOI: 10.3390/ijms25168923] [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: 07/12/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/02/2024] Open
Abstract
Ocrelizumab (OCR) is a humanized anti-CD20 monoclonal antibody approved for both Relapsing and Primary Progressive forms of Multiple Sclerosis (MS) treatment. OCR is postulated to act via rapid B cell depletion; however, by analogy with other anti-CD20 agents, additional effects can be envisaged, such as on Protein Kinase C (PKC). Hence, this work aims to explore novel potential mechanisms of action of OCR in peripheral blood mononuclear cells from MS patients before and after 12 months of OCR treatment. We first assessed, up-stream, PKCβII and subsequently explored two down-stream pathways: hypoxia-inducible factor 1 alpha (HIF-1α)/vascular endothelial growth factor (VEGF), and human antigen R (HuR)/manganese-dependent superoxide dismutase (MnSOD) and heat shock proteins 70 (HSP70). At baseline, higher levels of PKCβII, HIF-1α, and VEGF were found in MS patients compared to healthy controls (HC); interestingly, the overexpression of this inflammatory cascade was counteracted by OCR treatment. Conversely, at baseline, the content of HuR, MnSOD, and HSP70 was significantly lower in MS patients compared to HC, while OCR administration induced the up-regulation of these neuroprotective pathways. These results enable us to disclose the dual positive action of OCR: anti-inflammatory and neuroprotective. Therefore, in addition to B cell depletion, the effect of OCR on these molecular cascades can contribute to counteracting disease progression.
Collapse
Affiliation(s)
| | - Lara Ahmad
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
| | - Nicoletta Marchesi
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy; (L.I.M.C.); (N.M.); (F.B.)
| | - Giacomo Greco
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | - Federica Boschi
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy; (L.I.M.C.); (N.M.); (F.B.)
| | - Francesco Masi
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
- Department of Brain and Behavioural Sciences, University of Pavia, 27100 Pavia, Italy
| | - Giulia Mallucci
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
- Neurocenter of Southern Switzerland, Ente Ospedaliero Cantonale (EOC), 6900 Lugano, Switzerland
| | - Roberto Bergamaschi
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
| | - Elena Colombo
- Multiple Sclerosis Center, IRCCS Mondino Foundation, 27100 Pavia, Italy; (L.A.); (G.G.); (F.M.); (R.B.); (E.C.)
| | - Alessia Pascale
- Department of Drug Sciences, Section of Pharmacology, University of Pavia, 27100 Pavia, Italy; (L.I.M.C.); (N.M.); (F.B.)
| |
Collapse
|
21
|
Varco-Merth B, Chaunzwa M, Duell DM, Marenco A, Goodwin W, Dannay R, Nekorchuk M, Shao D, Busman-Sahay K, Fennessey CM, Silipino L, Hull M, Bosche WJ, Fast R, Oswald K, Shoemaker R, Bochart R, MacAllister R, Labriola CS, Smedley JV, Axthelm MK, Davenport MP, Edlefsen PT, Estes JD, Keele BF, Lifson JD, Lewin SR, Picker LJ, Okoye AA. Impact of alemtuzumab-mediated lymphocyte depletion on SIV reservoir establishment and persistence. PLoS Pathog 2024; 20:e1012496. [PMID: 39173097 PMCID: PMC11373844 DOI: 10.1371/journal.ppat.1012496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 09/04/2024] [Accepted: 08/09/2024] [Indexed: 08/24/2024] Open
Abstract
Persistence of the rebound-competent viral reservoir (RCVR) within the CD4+ T cell compartment of people living with HIV remains a major barrier to HIV cure. Here, we determined the effects of the pan-lymphocyte-depleting monoclonal antibody (mAb) alemtuzumab on the RCVR in SIVmac239-infected rhesus macaques (RM) receiving antiretroviral therapy (ART). Alemtuzumab administered during chronic ART or at the time of ART initiation induced >95% depletion of circulating CD4+ T cells in peripheral blood and substantial CD4+ T cell depletion in lymph nodes. However, treatment was followed by proliferation and reconstitution of CD4+ T cells in blood, and despite ongoing ART, levels of cell-associated SIV DNA in blood and lymphoid tissues were not substantially different between alemtuzumab-treated and control RM after immune cell reconstitution, irrespective of the time of alemtuzumab treatment. Upon ART cessation, 19 of 22 alemtuzumab-treated RM and 13 of 13 controls rebounded with no difference in the time to rebound between treatment groups. Time to rebound and reactivation rate was associated with plasma viral loads (pVLs) at time of ART initiation, suggesting lymphocyte depletion had no durable impact on the RCVR. However, 3 alemtuzumab-treated RM that had lowest levels of pre-ART viremia, failed to rebound after ART withdrawal, in contrast to controls with similar levels of SIV replication. These observations suggest that alemtuzumab therapy has little to no ability to reduce well-established RCVRs but may facilitate RCVR destabilization when pre-ART virus levels are particularly low.
Collapse
Affiliation(s)
- Benjamin Varco-Merth
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Morgan Chaunzwa
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Derick M. Duell
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Alejandra Marenco
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - William Goodwin
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Rachel Dannay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Michael Nekorchuk
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Danica Shao
- Fred Hutchinson Cancer Research Center, Seattle, Washington State, United States of America
| | - Kathleen Busman-Sahay
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Christine M. Fennessey
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Lorna Silipino
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Michael Hull
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - William J. Bosche
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Randy Fast
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Kelli Oswald
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Rebecca Shoemaker
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Rachele Bochart
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Rhonda MacAllister
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Caralyn S. Labriola
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Jeremy V. Smedley
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Michael K. Axthelm
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Paul T. Edlefsen
- Fred Hutchinson Cancer Research Center, Seattle, Washington State, United States of America
| | - Jacob D. Estes
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Brandon F. Keele
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Jeffrey D. Lifson
- AIDS and Cancer Virus Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Sharon R. Lewin
- Department of Infectious Diseases, The University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, Australia
| | - Louis J. Picker
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| | - Afam A. Okoye
- Vaccine and Gene Therapy Institute, Oregon Health & Science University, Beaverton, Oregon, United States of America
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, Oregon, United States of America
| |
Collapse
|
22
|
Perricone C, Castellucci A, Cafaro G, Calvacchi S, Bruno L, Dal Pozzolo R, Tromby F, Colangelo A, Gerli R, Bartoloni E. Rational approach to the prescription of anti-rheumatic drugs in rheumatoid arthritis: a product leaflet-based strategy in Italy. Front Immunol 2024; 15:1398314. [PMID: 38979406 PMCID: PMC11228816 DOI: 10.3389/fimmu.2024.1398314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/06/2024] [Indexed: 07/10/2024] Open
Abstract
The treatment of patients with rheumatoid arthritis (RA) has dramatically changed in the past 30 years. Currently, numerous conventional, biologic, and targeted synthetic DMARDs have been licensed and used following recommendations provided by international and national scientific societies. However, the availability of biosimilars and the increasing necessity of savings impacted on the local/national prescription of these drugs. The information provided by data sheet of every single drug is a decisive factor on the choice of a certain treatment merged with the patient's profile. Thus, our purpose was to construct a rational algorithm for the treatment strategy in RA according to costs and the product leaflet of the biologic and targeted-synthetic DMARDs currently licensed in Italy. We used the most recent available recommendations and then we performed a review of the literature considering all the factors that are known to influence drug safety/effectiveness. All these factors were considered in the context of the data sheets of currently available originators and biosimilars.
Collapse
Affiliation(s)
- Carlo Perricone
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Andrea Castellucci
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giacomo Cafaro
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Santina Calvacchi
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Lorenza Bruno
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Dal Pozzolo
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Tromby
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Anna Colangelo
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Roberto Gerli
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Bartoloni
- Rheumatology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
Lin HH, Wang CH, Huang SH, Lin SY, Kato T, Namba K, Hosogi N, Song C, Murata K, Yen CH, Hsu TL, Wong CH, Wu YM, Tu IP, Chang WH. Use of phase plate cryo-EM reveals conformation diversity of therapeutic IgG with 50 kDa Fab fragment resolved below 6 Å. Sci Rep 2024; 14:14079. [PMID: 38890341 PMCID: PMC11189423 DOI: 10.1038/s41598-024-62045-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 05/13/2024] [Indexed: 06/20/2024] Open
Abstract
While cryogenic electron microscopy (cryo-EM) is fruitfully used for harvesting high-resolution structures of sizable macromolecules, its application to small or flexible proteins composed of small domains like immunoglobulin (IgG) remain challenging. Here, we applied single particle cryo-EM to Rituximab, a therapeutic IgG mediating anti-tumor toxicity, to explore its solution conformations. We found Rituximab molecules exhibited aggregates in cryo-EM specimens contrary to its solution behavior, and utilized a non-ionic detergent to successfully disperse them as isolated particles amenable to single particle analysis. As the detergent adversely reduced the protein-to-solvent contrast, we employed phase plate contrast to mitigate the impaired protein visibility. Assisted by phase plate imaging, we obtained a canonical three-arm IgG structure with other structures displaying variable arm densities co-existing in solution, affirming high flexibility of arm-connecting linkers. Furthermore, we showed phase plate imaging enables reliable structure determination of Fab to sub-nanometer resolution from ab initio, yielding a characteristic two-lobe structure that could be unambiguously docked with crystal structure. Our findings revealed conformation diversity of IgG and demonstrated phase plate was viable for cryo-EM analysis of small proteins without symmetry. This work helps extend cryo-EM boundaries, providing a valuable imaging and structural analysis framework for macromolecules with similar challenging features.
Collapse
Affiliation(s)
- Hsin-Hung Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Chun-Hsiung Wang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Academia Sinica Cryo-EM Facility, Academia Sinica, Taipei, Taiwan
| | - Shih-Hsin Huang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Institute of Preventive Medicine, National Defense Medical Center, New Taipei City, Taiwan
| | - Sung-Yao Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Takayuki Kato
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, Japan
- Institute of Protein Research, Osaka University, Suita, Osaka, Japan
| | - Keiichi Namba
- Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka, Japan
| | - Naoki Hosogi
- JEOL Ltd., 1-2 Musashino 3-chome, Akishima, Tokyo, Japan
| | - Chihong Song
- Exploratory Research Center on Life and Living Systems (ExCELLS) and National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, Japan
| | - Kazuyoshi Murata
- Exploratory Research Center on Life and Living Systems (ExCELLS) and National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, 38 Nishigonaka Myodaiji, Okazaki, Aichi, Japan
| | | | - Tsui-Ling Hsu
- Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Chi-Huey Wong
- Genomic Research Center, Academia Sinica, Taipei, Taiwan
| | - Yi-Min Wu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Cryo-EM Facility, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - I-Ping Tu
- Institute of Statistical Science, Academia Sinica, Taipei, Taiwan
| | - Wei-Hau Chang
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan.
- Genomic Research Center, Academia Sinica, Taipei, Taiwan.
- Institute of Physics, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
24
|
Mundisugih J, Ravindran D, Kizana E. Exploring the Therapeutic Potential of Gene Therapy in Arrhythmogenic Right Ventricular Cardiomyopathy. Biomedicines 2024; 12:1351. [PMID: 38927558 PMCID: PMC11201581 DOI: 10.3390/biomedicines12061351] [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: 05/17/2024] [Revised: 06/07/2024] [Accepted: 06/13/2024] [Indexed: 06/28/2024] Open
Abstract
Right dominant arrhythmogenic cardiomyopathy, commonly known as Arrhythmogenic Right Ventricular Cardiomyopathy (ARVC), represents a formidable challenge in cardiovascular medicine, as conventional therapies are commonly ineffective in impeding disease progression and the development of end-stage heart failure. Recombinant adeno-associated virus (AAV)-mediated gene therapy presents a promising avenue for targeted therapeutic interventions, potentially revolutionising treatment approaches for ARVC patients. Encouraging results from preclinical studies have sparked optimism about the possibility of curing specific subtypes of ARVC in the near future. This narrative review delves into the dynamic landscape of genetic therapy for ARVC, elucidating its underlying mechanisms and developmental stages, and providing updates on forthcoming trials. Additionally, it examines the hurdles and complexities impeding the successful translation of ARVC genetic therapies into clinical practice. Despite notable scientific advancements, the journey towards implementing genetic therapies for ARVC patients in real-world clinical settings is still in its early phases.
Collapse
Affiliation(s)
- Juan Mundisugih
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Dhanya Ravindran
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| | - Eddy Kizana
- Centre for Heart Research, Westmead Institute for Medical Research, Westmead, NSW 2145, Australia
- Department of Cardiology, Westmead Hospital, Westmead, NSW 2145, Australia
- Sydney Medical School, The University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
25
|
Zhang C, Lin Y, Kuang Q, Li H, Jiang Q, Yang X. Case report: A highly active refractory myasthenia gravis with treatment of telitacicept combined with efgartigimod. Front Immunol 2024; 15:1400459. [PMID: 38799457 PMCID: PMC11116603 DOI: 10.3389/fimmu.2024.1400459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 04/22/2024] [Indexed: 05/29/2024] Open
Abstract
There is always a lack of effective treatment for highly active refractory generalized myasthenia gravis (GMG). Recently, telitacicept combined with efgartigimod significantly reduces circulating B cells, plasma cells, and immunoglobulin G, which brings promising therapeutic strategies. We report a case of a 37-year-old female patient with refractory GMG, whose condition got significant improvement and control with this latest treatment after multiple unsuccessful therapies of immunosuppressants. The new combination deserves further attention in the therapeutic application of myasthenia gravis.
Collapse
Affiliation(s)
| | | | | | | | - Qilong Jiang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaojun Yang
- The First Affiliated Hospital of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
26
|
McLinden GP, Avery AC, Gardner HL, Hughes K, Rodday AM, Liang K, London CA. Safety and biologic activity of a canine anti-CD20 monoclonal antibody in dogs with diffuse large B-cell lymphoma. J Vet Intern Med 2024; 38:1666-1674. [PMID: 38662527 PMCID: PMC11099711 DOI: 10.1111/jvim.17080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 04/05/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND To explore the safety and utility of combining low dose single-agent doxorubicin with a canine specific anti-CD20 monoclonal antibody (1E4-cIgGB) in client owned dogs with untreated B-cell lymphoma. ANIMALS Forty-two client-owned dogs with untreated B-cell lymphoma. METHODS A prospective, single arm, open label clinical trial of dogs with B-cell lymphoma were enrolled to receive 1E4-cIgGB and doxorubicin in addition to 1 of 3 immunomodulatory regimens. B-cell depletion was monitored by flow cytometry performed on peripheral blood samples at each visit. RESULTS Dogs demonstrated a statistically significant depletion in CD21+ B-cells 7 days following the first antibody infusion (median fraction of baseline at 7 days = 0.04, P < .01) that persisted throughout treatment (median fraction of baseline at 21 days = 0.01, P < .01) whereas CD5+ T-cells remained unchanged (median fraction of baseline at 7 days = 1.05, P = .88; median fraction of baselie at 7 days = 0.79, P = .42; Figure 1; Supplemental Table 3). Recovery of B-cells was delayed, with at Day 196, only 6/17 dogs (35%) remaining on the study had CD21+ counts >0.5 of baseline, indicating sustained B cell depletion at 4+ months after the final treatment. 1E4-cIgGB was well tolerated with only 1 dog exhibiting a hypersensitivity event within minutes of the last antibody infusion. CONCLUSIONS The canine 1E4-cIgGB anti-CD20 monoclonal antibody is apparently safe when administered with doxorubicin and effectively depletes B-cells in dogs with DLBCL.
Collapse
MESH Headings
- Animals
- Dogs
- Dog Diseases/drug therapy
- Dog Diseases/immunology
- Doxorubicin/therapeutic use
- Doxorubicin/pharmacology
- Doxorubicin/administration & dosage
- Female
- Male
- Lymphoma, Large B-Cell, Diffuse/veterinary
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/adverse effects
- Prospective Studies
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- Antigens, CD20/immunology
Collapse
Affiliation(s)
- Gretchen P. McLinden
- Cummings School of Veterinary MedicineTufts UniversityNorth GraftonMassachusettsUSA
| | - Anne C. Avery
- College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Heather L. Gardner
- Cummings School of Veterinary MedicineTufts UniversityNorth GraftonMassachusettsUSA
| | - Kelley Hughes
- College of Veterinary Medicine and Biomedical SciencesColorado State UniversityFort CollinsColoradoUSA
| | - Angie M. Rodday
- Clinical Translational Science InstituteTufts UniversityNorth GraftonMassachusettsUSA
| | - Kexuan Liang
- Clinical Translational Science InstituteTufts UniversityNorth GraftonMassachusettsUSA
| | - Cheryl A. London
- Cummings School of Veterinary MedicineTufts UniversityNorth GraftonMassachusettsUSA
| |
Collapse
|
27
|
Pang Y, Ghosh N. Novel and multiple targets for chimeric antigen receptor-based therapies in lymphoma. Front Oncol 2024; 14:1396395. [PMID: 38711850 PMCID: PMC11070555 DOI: 10.3389/fonc.2024.1396395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy targeting CD19 in B-cell non-Hodgkin lymphoma (NHL) validates the utility of CAR-based therapy for lymphomatous malignancies. Despite the success, treatment failure due to CD19 antigen loss, mutation, or down-regulation remains the main obstacle to cure. On-target, off-tumor effect of CD19-CAR T leads to side effects such as prolonged B-cell aplasia, limiting the application of therapy in indolent diseases such as chronic lymphocytic leukemia (CLL). Alternative CAR targets and multi-specific CAR are potential solutions to improving cellular therapy outcomes in B-NHL. For Hodgkin lymphoma and T-cell lymphoma, several cell surface antigens have been studied as CAR targets, some of which already showed promising results in clinical trials. Some antigens are expressed by different lymphomas and could be used for designing tumor-agnostic CAR. Here, we reviewed the antigens that have been studied for novel CAR-based therapies, as well as CARs designed to target two or more antigens in the treatment of lymphoma.
Collapse
Affiliation(s)
- Yifan Pang
- Department of Hematologic Oncology and Blood Disorders, Atrium Health Levine Cancer Institute, Wake Forest School of Medicine, Charlotte, NC, United States
| | | |
Collapse
|
28
|
Azzam T, Du JJ, Flowers MW, Ali AV, Hunn JC, Vijayvargiya N, Knagaram R, Bogacz M, Maravillas KE, Sastre DE, Fields JK, Mirzaei A, Pierce BG, Sundberg EJ. Combinatorially restricted computational design of protein-protein interfaces to produce IgG heterodimers. SCIENCE ADVANCES 2024; 10:eadk8157. [PMID: 38598628 PMCID: PMC11006224 DOI: 10.1126/sciadv.adk8157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 03/08/2024] [Indexed: 04/12/2024]
Abstract
Redesigning protein-protein interfaces is an important tool for developing therapeutic strategies. Interfaces can be redesigned by in silico screening, which allows for efficient sampling of a large protein space before experimental validation. However, computational costs limit the number of combinations that can be reasonably sampled. Here, we present combinatorial tyrosine (Y)/serine (S) selection (combYSelect), a computational approach combining in silico determination of the change in binding free energy (ΔΔG) of an interface with a highly restricted library composed of just two amino acids, tyrosine and serine. We used combYSelect to design two immunoglobulin G (IgG) heterodimers-combYSelect1 (L368S/D399Y-K409S/T411Y) and combYSelect2 (D399Y/K447S-K409S/T411Y)-that exhibit near-optimal heterodimerization, without affecting IgG stability or function. We solved the crystal structures of these heterodimers and found that dynamic π-stacking interactions and polar contacts drive preferential heterodimeric interactions. Finally, we demonstrated the utility of our combYSelect heterodimers by engineering both a bispecific antibody and a cytokine trap for two unique therapeutic applications.
Collapse
Affiliation(s)
- Tala Azzam
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jonathan J. Du
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Maria W. Flowers
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Adeela V. Ali
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jeremy C. Hunn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nina Vijayvargiya
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Rushil Knagaram
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Marek Bogacz
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kino E. Maravillas
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Diego E. Sastre
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - James K. Fields
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Ardalan Mirzaei
- Sydney Pharmacy School, Faculty of Medicine and Health, The University of Sydney, Camperdown, NSW, Australia
| | - Brian G. Pierce
- University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, MD 20850, USA
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20850, USA
| | - Eric J. Sundberg
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322, USA
| |
Collapse
|
29
|
Potter RA, Peterson EL, Griffin D, Cooper Olson G, Lewis S, Cochran K, Mendell JR, Rodino-Klapac LR. Use of plasmapheresis to lower anti-AAV antibodies in nonhuman primates with pre-existing immunity to AAVrh74. Mol Ther Methods Clin Dev 2024; 32:101195. [PMID: 38327805 PMCID: PMC10847772 DOI: 10.1016/j.omtm.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 01/18/2024] [Indexed: 02/09/2024]
Abstract
Patients with pre-existing immunity to adeno-associated virus (AAV) are currently unable to receive systemic gene transfer therapies. In this nonhuman primate study, we investigated the impact of immunosuppression strategies on gene transfer therapy safety and efficacy and analyzed plasmapheresis as a potential pretreatment for circumvention of pre-existing immunity or redosing. In part 1, animals received delandistrogene moxeparvovec (SRP-9001), an AAVrh74-based gene transfer therapy for Duchenne muscular dystrophy. Cohort 1 (control, n = 2) received no immunosuppression; cohorts 2-4 (n = 3 per cohort) received prednisone at different time points; and cohort 5 (n = 3) received rituximab, sirolimus, and prednisone before and after dosing. In part 2, cohorts 2-4 underwent plasmapheresis before redosing; cohort 5 was redosed without plasmapheresis. We analyzed safety, immune response (humoral and cell-mediated responses and complement activation), and vector genome distribution. After 2 or 3 plasmapheresis exchanges, circulating anti-AAVrh74 antibodies were reduced, and animals were redosed. Plasmapheresis was well tolerated, with no abnormal clinical or immunological observations. Cohort 5 (redosed with high anti-AAVrh74 antibody titers) had hypersensitivity reactions, which were controlled with treatment. These findings suggest that plasmapheresis is a safe and effective method to reduce anti-AAV antibody levels in nonhuman primates prior to gene transfer therapy. The results may inform human studies involving redosing or circumvention of pre-existing immunity.
Collapse
Affiliation(s)
| | | | | | | | - Sarah Lewis
- Sarepta Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Kyle Cochran
- Sarepta Therapeutics, Inc., Cambridge, MA 02142, USA
| | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43210, USA
| | - Louise R. Rodino-Klapac
- Sarepta Therapeutics, Inc., Cambridge, MA 02142, USA
- Center for Gene Therapy, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
30
|
Jeong E, Hong H, Lee YA, Kim KS. Potential Rheumatoid Arthritis-Associated Interstitial Lung Disease Treatment and Computational Approach for Future Drug Development. Int J Mol Sci 2024; 25:2682. [PMID: 38473928 PMCID: PMC11154459 DOI: 10.3390/ijms25052682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disease characterized by swelling in at least one joint. Owing to an overactive immune response, extra-articular manifestations are observed in certain cases, with interstitial lung disease (ILD) being the most common. Rheumatoid arthritis-associated interstitial lung disease (RA-ILD) is characterized by chronic inflammation of the interstitial space, which causes fibrosis and the scarring of lung tissue. Controlling inflammation and pulmonary fibrosis in RA-ILD is important because they are associated with high morbidity and mortality. Pirfenidone and nintedanib are specific drugs against idiopathic pulmonary fibrosis and showed efficacy against RA-ILD in several clinical trials. Immunosuppressants and disease-modifying antirheumatic drugs (DMARDs) with anti-fibrotic effects have also been used to treat RA-ILD. Immunosuppressants moderate the overexpression of cytokines and immune cells to reduce pulmonary damage and slow the progression of fibrosis. DMARDs with mild anti-fibrotic effects target specific fibrotic pathways to regulate fibrogenic cellular activity, extracellular matrix homeostasis, and oxidative stress levels. Therefore, specific medications are required to effectively treat RA-ILD. In this review, the commonly used RA-ILD treatments are discussed based on their molecular mechanisms and clinical trial results. In addition, a computational approach is proposed to develop specific drugs for RA-ILD.
Collapse
Affiliation(s)
- Eunji Jeong
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Hyunseok Hong
- Yale College, Yale University, New Haven, CT 06520, USA;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Yeon-Ah Lee
- Division of Rheumatology, Department of Internal Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Kyoung-Soo Kim
- Department of Medicine, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Clinical Pharmacology and Therapeutics, School of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- East-West Bone & Joint Disease Research Institute, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea
| |
Collapse
|
31
|
Huang W, Wang B, Ou Q, Zhang X, He Y, Mao X, Wei X, Kou X. ASC-expressing pyroptotic extracellular vesicles alleviate sepsis by protecting B cells. Mol Ther 2024; 32:395-410. [PMID: 38093517 PMCID: PMC10861962 DOI: 10.1016/j.ymthe.2023.12.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 11/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Pyroptosis is an inflammatory programmed cell death process characterized by membrane rupture. Interestingly, pyroptotic cells can generate plenty of nanosized vesicles. Non-inflammatory apoptotic cell death-derived apoptotic vesicles (apoVs) were systemically characterized and displayed multiple physiological functions and therapeutic potentials. However, the characteristics of pyroptotic cell-generated extracellular vesicles (EVs) are largely unknown. Here, we identified a group of pyroptotic EVs (pyroEVs) from in vitro cultured pyroptotic mesenchymal stem cells (MSCs), as well as from septic mouse blood. Compared with apoVs, pyroEVs express similar levels of annexin V, calreticulin, and common EV markers, but express a decreased level of apoptotic marker cleave caspase-3. PyroEVs, but not apoVs and exosomes, specifically express pyroptotic maker apoptosis-associated speck-like protein containing CARD (ASC). More importantly, MSC-derived pyroEVs protect B cells in the spleen and bone marrow to relieve inflammatory responses and enhance the survival rate of the septic mice. Mechanistically, pyroEV membrane-expressed ASC binds to B cells to repress cell death by repressing Toll-like receptor 4. This study uncovered the characteristics of pyroEVs and their therapeutic role in sepsis and B cell-mediated immune response.
Collapse
Affiliation(s)
- Weiying Huang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Ben Wang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Qianmin Ou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xiao Zhang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Department of Prosthodontics, Peking University School and Hospital of Stomatology and National Clinical Research Center for Oral Diseases, National Engineering Laboratory for Digital and Material Technology of Stomatology, and Beijing Key Laboratory of Digital Stomatology, 22 Zhongguancun South Avenue, Haidian District, Beijing 100081, China
| | - Yifan He
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xueli Mao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong 510055, China.
| | - Xiaoxing Kou
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, South China Center of Craniofacial Stem Cell Research, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China; Key Laboratory of Stem Cells and Tissue Engineering (Sun Yat-Sen University), Ministry of Education, Guangzhou 510080, China.
| |
Collapse
|
32
|
Vecchio E, Marino R, Mimmi S, Canale C, Caiazza C, Arcucci A, Ruocco MR, Schiavone M, Santamaria G, Palmieri C, Iaccino E, Mallardo M, Quinto I, Fiume G. Enhanced pro-apoptotic activity of rituximab through IBTK silencing in non-Hodgkin lymphoma B-cells. Front Oncol 2024; 14:1339584. [PMID: 38371626 PMCID: PMC10869532 DOI: 10.3389/fonc.2024.1339584] [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: 11/17/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Rituximab is a commonly used chemotherapeutic drug for patients with aggressive lymphomas, such as non-Hodgkin's lymphoma (NHL). Currently, the combination of Rituximab and chemotherapy (R-CHOP) stands as the most prevalent first-line therapy for NHL. Nevertheless, the development of new therapeutic approaches remains imperative. An increasing body of evidence highlights a novel role for IBTK in tumorigenesis and cancer growth. In this study, we aim to broaden our understanding of IBTK's function in B-lymphoma, with a particular focus on its impact on the expression of the oncogene MYC. Here, we assessed the effects of combining Rituximab with IBTK silencing on cell viability through cell cycle analysis and Annexin V assays in vitro. Furthermore, we leveraged the transplantability of Eμ-myc lymphomas to investigate whether the inhibition of IBTK could elicit anti-tumor effects in the treatment of lymphomas in vivo. Our data suggests that IBTK silencing may serve as an effective anti-tumor agent for aggressive B-Lymphomas, underscoring its role in promoting apoptosis when used in combination with Rituximab, both in in vitro and in vivo settings.
Collapse
Affiliation(s)
- Eleonora Vecchio
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Rossana Marino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Selena Mimmi
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Camilla Canale
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Carmen Caiazza
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Alessandro Arcucci
- Department of Public Health, University of Naples “Federico II”, Naples, Italy
| | - Maria Rosaria Ruocco
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Marco Schiavone
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Gianluca Santamaria
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Camillo Palmieri
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Massimo Mallardo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Ileana Quinto
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| | - Giuseppe Fiume
- Department of Experimental and Clinical Medicine, University Magna Græcia of Catanzaro, Catanzaro, Italy
| |
Collapse
|
33
|
Panebianco M, Ciccarese C, Strusi A, Beccia V, Carbone C, Agostini A, Piro G, Tortora G, Iacovelli R. The Role of the Complement in Clear Cell Renal Carcinoma (ccRCC)-What Future Prospects Are There for Its Use in Clinical Practice? Cancers (Basel) 2024; 16:490. [PMID: 38339243 PMCID: PMC10854780 DOI: 10.3390/cancers16030490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
In recent years, the first-line available therapeutic options for metastatic renal cell carcinoma (mRCC) have radically changed with the introduction into clinical practice of new immune checkpoint inhibitor (ICI)-based combinations. Many efforts are focusing on identifying novel prognostic and predictive markers in this setting. The complement system (CS) plays a central role in promoting the growth and progression of mRCC. In particular, mRCC has been defined as an "aggressive complement tumor", which encompasses a group of malignancies with poor prognosie and highly expressed complement components. Several preclinical and retrospective studies have demonstrated the negative prognostic role of the complement in mRCC; however, there is little evidence on its possible role as a predictor of the response to ICIs. The purpose of this review is to explore more deeply the physio-pathological role of the complement in the development of RCC and its possible future use in clinical practice as a prognostic and predictive factor.
Collapse
Affiliation(s)
- Martina Panebianco
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Chiara Ciccarese
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Alessandro Strusi
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Viria Beccia
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Carmine Carbone
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Antonio Agostini
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Geny Piro
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
| | - Giampaolo Tortora
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| | - Roberto Iacovelli
- Medical Oncology, Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (M.P.); (C.C.); (C.C.); (A.A.); (G.P.); (G.T.)
- Medical Oncology, Department of Translational Medicine and Surgery, Catholic University of the Sacred Heart, 00168 Rome, Italy; (A.S.); (V.B.)
| |
Collapse
|
34
|
Campbell E, Adamson H, Luxton T, Tiede C, Wälti C, Tomlinson DC, Jeuken LJC. Therapeutic drug monitoring of immunotherapies with novel Affimer-NanoBiT sensor construct. SENSORS & DIAGNOSTICS 2024; 3:104-111. [PMID: 38249540 PMCID: PMC10795742 DOI: 10.1039/d3sd00126a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Concentration-therapeutic efficacy relationships have been observed for several therapeutic monoclonal antibodies (TmAb), where low circulating levels can result in ineffective treatment and high concentrations can cause adverse reactions. Rapid therapeutic drug monitoring (TDM) of TmAb drugs would provide the opportunity to adjust an individual patient's dosing regimen to improve treatment results. However, TDM for immunotherapies is currently limited to centralised testing methods with long sample-collection to result timeframes. Here, we show four point-of-care (PoC) TmAb biosensors by combining anti-idiotypic Affimer proteins and NanoBiT split luciferase technology at a molecular level to provide a platform for rapid quantification (<10 minutes) for four clinically relevant TmAb (rituximab, adalimumab, ipilimumab and trastuzumab). The rituximab sensor performed best with 4 pM limit of detection (LoD) and a quantifiable range between 8 pM-2 nM with neglectable matrix effects in serum up to 1%. After dilution of serum samples, the resulting quantifiable range for all four sensors falls within the clinically relevant range and compares favourably with the sensitivity and/or time-to-result of current ELISA standards. Further development of these sensors into a PoC test may improve treatment outcome and quality of life for patients receiving immunotherapy.
Collapse
Affiliation(s)
- Emma Campbell
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Hope Adamson
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Timothy Luxton
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
| | - Christian Tiede
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- School of Molecular and Cellular Biology, University of Leeds Leeds LS2 9JT UK
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds LS2 9JT UK
| | - Darren C Tomlinson
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- School of Molecular and Cellular Biology, University of Leeds Leeds LS2 9JT UK
| | - Lars J C Jeuken
- School of Biomedical Science, University of Leeds Leeds LS2 9JT UK
- Astbury Centre for Structural Molecular Biology, University of Leeds LS2 9JT UK
- Leiden Institute of Chemistry, Leiden University PO Box 9502 2300 RA Leiden The Netherlands
| |
Collapse
|
35
|
Wemlinger SM, Cambier JC. Therapeutic tactics for targeting B lymphocytes in autoimmunity and cancer. Eur J Immunol 2024; 54:e2249947. [PMID: 37816494 DOI: 10.1002/eji.202249947] [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: 01/13/2023] [Revised: 10/05/2023] [Accepted: 10/09/2023] [Indexed: 10/12/2023]
Abstract
B lymphocytes have become a very popular therapeutic target in a number of autoimmune indications due to their newly appreciated roles, and approachability, in these diseases. Many of the therapies now applied in autoimmunity were initially developed to deplete malignant B cells. These strategies have also been found to benefit patients suffering from such autoimmune diseases as multiple sclerosis, type I diabetes, systemic lupus erythematosus, and rheumatoid arthritis, to name a few. These observations have supported the expansion of research addressing the mechanistic contributions of B cells in these diseases, as well as blossoming of therapeutics that target them. This review seeks to summarize cutting-edge modalities for targeting B cells, including monoclonal antibodies, bispecific antibodies, antibody-drug conjugates, chimeric antigen receptor-T cells, and small molecule inhibitors. Efforts to refine B-cell targeted therapy to eliminate only pathogenic autoreactive cells will be addressed as well as the potential for future B-cell-based cellular therapeutics. Finally, we also address approaches that seek to silence B-cell function without depletion.
Collapse
Affiliation(s)
- Scott M Wemlinger
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - John C Cambier
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
36
|
Park SJ, Greer PL, Lee N. From odor to oncology: non-canonical odorant receptors in cancer. Oncogene 2024; 43:304-318. [PMID: 38087050 DOI: 10.1038/s41388-023-02908-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/16/2023] [Accepted: 11/23/2023] [Indexed: 01/31/2024]
Abstract
Odorant receptors, traditionally associated with olfaction as chemoreceptors, have been increasingly recognized for their presence and diverse functions in various non-nasal tissues throughout the body. Beyond their roles in sensory perception, emerging evidence suggests a compelling interplay between odorant receptors and cancer progression as well. Alongside the canonical GPCR odorant receptors, dysregulation of non-canonical odorant receptors such as trace amine-associated receptors (TAARs), formyl peptide receptors (FPRs), and membrane-spanning 4A family (MS4As) has been observed in various cancer types, suggesting their contributions to cancer progression. The roles of these non-canonical chemoreceptors in cancer are complex, with some receptors promoting tumorigenesis and others acting as tumor-suppressing factors upon activation, depending on the cancer type. These findings shed light on the potential of non-canonical odorant receptors as therapeutic targets and prognostic markers in cancer, inviting further exploration to unravel their precise mechanisms of action and implications in cancer biology. In this review, we provide a comprehensive overview of the intricate relationships between these chemoreceptors and various types of cancer, potentially paving the way for innovative odor-based therapeutics. Ultimately, this review discusses the potential development of novel therapeutic strategies targeting these non-canonical chemoreceptors.
Collapse
Affiliation(s)
- Sung Jin Park
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Paul L Greer
- Program in Molecular Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Namgyu Lee
- Department of Biomedical Science and Engineering, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
37
|
Bak YG, Choi HJ, Kim YE, Oh SH, Kim KM. Successful Use of Bortezomib for Recurrent Progressive Familial Intrahepatic Cholestasis Type II After Liver Transplantation: A Pediatric Case with a 9-Year Follow-Up. Pediatr Gastroenterol Hepatol Nutr 2024; 27:71-76. [PMID: 38249643 PMCID: PMC10796264 DOI: 10.5223/pghn.2024.27.1.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 01/23/2024] Open
Abstract
Recurrence of progressive familial intrahepatic cholestasis (PFIC) type II poses challenges during postoperative liver transplant care. Posttransplant patients with PFIC type II risk developing recurrent cholestasis with normal gamma-glutamyl transferase activity, which mimics the original bile salt export pump (BSEP) protein deficiency and is related to a form of immunoglobulin G antibody (anti-BSEP)-mediated rejection. Bortezomib effectively induces apoptosis of actively antibody-producing plasma cells that may have a role in antibody-mediated rejection. In this case, we used bortezomib to treat PFIC type II recurrence after liver transplantation in a child.
Collapse
Affiliation(s)
- Yu Gyoung Bak
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, Korea
| | - Ho Jung Choi
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, Korea
| | - Yeong Eun Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, Korea
| | - Seak Hee Oh
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, Korea
| | - Kyung Mo Kim
- Department of Pediatrics, University of Ulsan College of Medicine, Asan Medical Center Children’s Hospital, Seoul, Korea
| |
Collapse
|
38
|
Sayer M, Chapman GB, Thomas M, Dhaun N. Cardiovascular Disease in Anti-neutrophil Cytoplasm Antibody-Associated Vasculitis. Curr Rheumatol Rep 2024; 26:12-23. [PMID: 38015334 PMCID: PMC10776689 DOI: 10.1007/s11926-023-01123-8] [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] [Accepted: 11/06/2023] [Indexed: 11/29/2023]
Abstract
PURPOSE OF REVIEW Anti-neutrophil cytoplasm antibody (ANCA)-associated vasculitis (AAV) is a rare, multisystem, autoimmune disease characterised by microvascular inflammation. Over the past 20 years, advances in immunological management have improved short-term patient outcomes. Longer-term patient outcomes remain poor with cardiovascular disease now the leading cause of death in AAV. Here, we examine the potential pathways that contribute to the increased risk of cardiovascular disease in AAV and the current evidence to manage this risk. RECENT FINDINGS The incidence of cardiovascular disease in AAV exceeds that expected by traditional risk factors alone, suggesting a contribution from disease-specific factors. Similarly, it is unclear how different immunosuppressive therapies contribute to and modify cardiovascular risk, and there is a paucity of data examining the efficacy of traditional cardioprotective medications in AAV. There is a lack of evidence-based cardiovascular risk assessment tools and cardioprotective therapies in patients with AAV which should be addressed to improve long-term outcomes.
Collapse
Affiliation(s)
- Matthew Sayer
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Gavin B Chapman
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Matthew Thomas
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Neeraj Dhaun
- Edinburgh Kidney, University/BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, UK.
- Department of Renal Medicine, Royal Infirmary of Edinburgh, Edinburgh, UK.
| |
Collapse
|
39
|
Guo Y, Zhao H, Ren M, Wang Y, Wang L, Tang L. Efficacy and safety of rituximab in elderly patients with membranous nephropathy. Front Pharmacol 2023; 14:1323334. [PMID: 38186651 PMCID: PMC10771833 DOI: 10.3389/fphar.2023.1323334] [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: 10/17/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Objectives: Advancing age is a risk factor for treatment-related side effects and mortality in membranous nephropathy (MN) patients treated with traditional immunosuppressive regimens. This study aimed to determine the efficacy and safety of rituximab (RTX) in the treatment of elderly patients with MN. Methods: We performed a single center retrospective review of 37 consecutive MN patients aged 70 and older at the time of RTX infusion. We also enrolled 76 young patients (<70 years old) with MN as the control group. We assessed clinical and laboratory indices, remission rates, and adverse events at RTX infusion, 3 months, and last visit. Results: A total of 37 elderly patients with MN were included, with a median follow-up period of 15.50 (10.00, 24.40) months. Of the 37 patients, 75.68% were male, and mean age was 71.89 ± 2.47 years. At last visit, 7 (18.92%) patients achieved complete remission, and 26 (70.27%) patients achieved complete or partial remission. There were no differences in the complete remission rate and complete or partial remission rate at last visit compared to young patients (26.32% vs. 18.92%, p = 0.387; 85.53% vs. 70.27%, p = 0.055). After RTX treatment, three of 6 elderly patients with pneumonia died due to ineffective treatment of the infection in RTX therapy courses. The results of multivariant regression analysis showed that elderly patients have an increased risk of serious infection, compared with patients younger than 70 years (OR = 32.874, 95% CI 1.300-831.490, p = 0.034). For each increase of 1 g/L in serum albumin, the risk of serious infection would decrease by 43.2% (OR = 0.568, 95% CI 0.334-0.969, p = 0.038). Conclusion: This study demonstrates that RTX is effective in the treatment of elderly patients with MN. However, we also observed a high incidence of infectious complications. Our experience was limited by its retrospective design and relatively small sample size, and further randomized controlled studies with large sample size are needed to confirm our preliminary findings.
Collapse
Affiliation(s)
- Yanhong Guo
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Huayan Zhao
- Department of Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingjing Ren
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yulin Wang
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liuwei Wang
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lin Tang
- Department of Nephropathy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Chen X, Qiu J, Gao Z, Liu B, Zhang C, Yu W, Yang J, Shen Y, Qi L, Yao X, Sun H, Yang X. Myasthenia gravis: Molecular mechanisms and promising therapeutic strategies. Biochem Pharmacol 2023; 218:115872. [PMID: 37865142 DOI: 10.1016/j.bcp.2023.115872] [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: 09/03/2023] [Revised: 10/16/2023] [Accepted: 10/18/2023] [Indexed: 10/23/2023]
Abstract
Myasthenia gravis (MG) is a type of autoimmune disease caused by the blockage of neuromuscular junction transmission owing to the attack of autoantibodies on transmission-related proteins. Related antibodies, such as anti-AChR, anti-MuSK and anti-LRP4 antibodies, can be detected in most patients with MG. Although traditional therapies can control most symptoms, several challenges remain to be addressed, necessitating the development of more effective and safe treatment strategies for MG. With the in-depth exploration on the mechanism and immune targets of MG, effective therapies, especially therapies using biologicals, have been reported recently. Given the important roles of immune cells, cytokines and intercellular interactions in the pathological process of MG, B-cell targeted therapy, T-cell targeted therapy, proteasome inhibitors targeting plasma cell, complement inhibitors, FcRn inhibitors have been developed for the treatment of MG. Although these novel therapies exert good therapeutic effects, they may weaken the immunity and increase the risk of infection in MG patients. This review elaborates on the pathogenesis of MG and discusses the advantages and disadvantages of the strategies of traditional treatment and biologicals. In addition, this review emphasises that combined therapy may have better therapeutic effects and reducing the risk of side effects of treatments, which has great prospects for the treatment of MG. With the deepening of research on immunotherapy targets in MG, novel opportunities and challenges in the treatment of MG will be introduced.
Collapse
Affiliation(s)
- Xin Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Jiayi Qiu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Zihui Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Boya Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Chen Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Weiran Yu
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Jiawen Yang
- Department of Clinical Medicine, Medical College, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Lei Qi
- Department of Emergency Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, PR China
| | - Xinlei Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Department of Neurology, Affiliated Hospital of Nantong University, Nantong University, Nantong, Jiangsu Province 226001, PR China.
| |
Collapse
|
41
|
Lee SM, Min SW, Kwon HS, Bae GD, Jung JH, Park HI, Lee SH, Lim CS, Ko BJ, Lee JC, Jung ST. Effective clearance of rituximab-resistant tumor cells by breaking the mirror-symmetry of immunoglobulin G and simultaneous binding to CD55 and CD20. Sci Rep 2023; 13:18275. [PMID: 37880350 PMCID: PMC10600224 DOI: 10.1038/s41598-023-45491-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 10/19/2023] [Indexed: 10/27/2023] Open
Abstract
Complement-dependent cytotoxicity (CDC), which eliminates aberrant target cells through the assembly and complex formation of serum complement molecules, is one of the major effector functions of anticancer therapeutic antibodies. In this study, we discovered that breaking the symmetry of natural immunoglobulin G (IgG) antibodies significantly increased the CDC activity of anti-CD20 antibodies. In addition, the expression of CD55 (a checkpoint inhibitor in the CDC cascade) was significantly increased in a rituximab-resistant cell line generated in-house, suggesting that CD55 overexpression might be a mechanism by which cancer cells acquire rituximab resistance. Based on these findings, we developed an asymmetric bispecific antibody (SBU-CD55 × CD20) that simultaneously targets both CD55 and CD20 to effectively eliminate rituximab-resistant cancer cells. In various cancer cell lines, including rituximab-resistant lymphoma cells, the SBU-CD55 × CD20 antibody showed significantly higher CDC activity than either anti-CD20 IgG antibody alone or a combination of anti-CD20 IgG antibody and anti-CD55 IgG antibody. Furthermore, the asymmetric bispecific antibody (SBU-CD55 × CD20) exhibited significantly higher CDC activity against rituximab-resistant cancer cells compared to other bispecific antibodies with symmetric features. These results demonstrate that enhancing CDC with an asymmetric CD55-binding bispecific antibody could be a new strategy for developing therapeutics to treat patients with relapsed or refractory cancers.
Collapse
Affiliation(s)
- Sang Min Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
- Department of Applied Chemistry, Kookmin University, 77, Jeongneung-ro, Seongbuk-gu, Seoul, 02707, Republic of Korea
| | - Sung-Won Min
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hyeong Sun Kwon
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Gong-Deuk Bae
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Ji Hae Jung
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Hye In Park
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea
| | - Seung Hyeon Lee
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Chung Su Lim
- New Drug Development Center, Osong Medical Innovation Foundation 123, Cheongju, Chungcheongbuk-do, 28160, Republic of Korea
| | - Byoung Joon Ko
- School of Biopharmaceutical and Medical Science, Sungshin Women's University, 55, Dobonng-Ro 76ga-gil, Gangbuk, Seoul, 01133, Republic of Korea
| | - Ji Chul Lee
- SG Medical, 3-11, Ogeum-ro 13-gil, Songpa-gu, Seoul, 05548, Republic of Korea.
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School of Medicine, Korea University, 73 Goryeodae-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea.
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Republic of Korea.
- Institute of Human Genetics, Korea University College of Medicine, Seoul, 02841, Republic of Korea.
- Biomedical Research Center, Korea University Anam Hospital, Seoul, 02841, Republic of Korea.
| |
Collapse
|
42
|
Campbell E, Adamson H, Kohl D, Tiede C, Wälti C, Tomlinson DC, Jeuken LJC. Enzyme - Switch sensors for therapeutic drug monitoring of immunotherapies. Biosens Bioelectron 2023; 237:115488. [PMID: 37419072 PMCID: PMC10427837 DOI: 10.1016/j.bios.2023.115488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/19/2023] [Accepted: 06/16/2023] [Indexed: 07/09/2023]
Abstract
Therapeutic monoclonal antibodies (TmAb) have emerged as effective treatments for a number of cancers and autoimmune diseases. However, large interpatient disparities in the pharmacokinetics of TmAb treatment requires close therapeutic drug monitoring (TDM) to optimise dosage for individual patients. Here we demonstrate an approach for achieving rapid, sensitive quantification of two monoclonal antibody therapies using a previously described enzyme switch sensor platform. The enzyme switch sensor consists of a β-lactamase - β-lactamase inhibitor protein (BLA-BLIP) complex with two anti-idiotype binding proteins (Affimer proteins) as recognition elements. The BLA-BLIP sensor was engineered to detect two TmAbs (trastuzumab and ipilimumab) by developing constructs incorporating novel synthetic binding reagents to each of these mAbs. Trastuzumab and ipilimumab were successfully monitored with sub nM sensitivity in up to 1% serum, thus covering the relevant therapeutic range. Despite the modular design, the BLA-BLIP sensor was unsuccessful in detecting two further TmAbs (rituximab and adalimumab), an explanation for which was explored. In conclusion, the BLA-BLIP sensors provide a rapid biosensor for TDM of trastuzumab and ipilimumab with the potential to improve therapy. The sensitivity of this platform alongside its rapid action would be suitable for bedside monitoring in a point-of-care (PoC) setting.
Collapse
Affiliation(s)
- Emma Campbell
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Hope Adamson
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Declan Kohl
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom
| | - Christian Tiede
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Christoph Wälti
- School of Electronic and Electrical Engineering, University of Leeds, LS2 9JT, United Kingdom
| | - Darren C Tomlinson
- Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Lars J C Jeuken
- School of Biomedical Science, University of Leeds, Leeds, LS2 9JT, United Kingdom; Astbury Centre for Structural Molecular Biology, University of Leeds, LS2 9JT, United Kingdom; Leiden Institute of Chemistry, Leiden University, PO Box 9502, 2300 RA, Leiden, the Netherlands.
| |
Collapse
|
43
|
Kittleson MM. Management of the sensitized heart transplant candidate. Curr Opin Organ Transplant 2023; 28:362-369. [PMID: 37678171 DOI: 10.1097/mot.0000000000001096] [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: 09/09/2023]
Abstract
PURPOSE OF REVIEW For sensitized heart transplant candidates who have antibodies to human leukocyte antigens (HLA), finding a suitable donor can be challenging and can lead to adverse waitlist outcomes. In recent years, the number of sensitized patients awaiting heart transplantation has increased likely due to the use of durable and mechanical circulatory support as well as increasing number of candidates with underlying congenital heart disease. RECENT FINDINGS Advances in the assessment of HLA antibodies allow for identification of heart transplant candidates who may benefit from desensitization strategies to widen the donor pool and mitigate the risk of adverse posttransplant outcomes. SUMMARY Antibody sensitization is a barrier to successful heart transplantation and strategies to identify sensitized patients, stratify their risk, and mitigate this risk through desensitization is crucial to optimize the quality of life and survival of HT recipients.
Collapse
Affiliation(s)
- Michelle M Kittleson
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
44
|
Rajoub N, Gerard CJJ, Pantuso E, Fontananova E, Caliandro R, Belviso BD, Curcio E, Nicoletta FP, Pullen J, Chen W, Heng JYY, Ruane S, Liddell J, Alvey N, Ter Horst JH, Di Profio G. A workflow for the development of template-assisted membrane crystallization downstream processing for monoclonal antibody purification. Nat Protoc 2023; 18:2998-3049. [PMID: 37697106 DOI: 10.1038/s41596-023-00869-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2023] [Accepted: 06/06/2023] [Indexed: 09/13/2023]
Abstract
Monoclonal antibodies (mAbs) are commonly used biologic drugs for the treatment of diseases such as rheumatoid arthritis, multiple sclerosis, COVID-19 and various cancers. They are produced in Chinese hamster ovary cell lines and are purified via a number of complex and expensive chromatography-based steps, operated in batch mode, that rely heavily on protein A resin. The major drawback of conventional procedures is the high cost of the adsorption media and the extensive use of chemicals for the regeneration of the chromatographic columns, with an environmental cost. We have shown that conventional protein A chromatography can be replaced with a single crystallization step and gram-scale production can be achieved in continuous flow using the template-assisted membrane crystallization process. The templates are embedded in a membrane (e.g., porous polyvinylidene fluoride with a layer of polymerized polyvinyl alcohol) and serve as nucleants for crystallization. mAbs are flexible proteins that are difficult to crystallize, so it can be challenging to determine the optimal conditions for crystallization. The objective of this protocol is to establish a systematic and flexible approach for the design of a robust, economic and sustainable mAb purification platform to replace at least the protein A affinity stage in traditional chromatography-based purification platforms. The procedure provides details on how to establish the optimal parameters for separation (crystallization conditions, choice of templates, choice of membrane) and advice on analytical and characterization methods.
Collapse
Affiliation(s)
- Nazer Rajoub
- CMAC Future Manufacturing Research Hub, c/o Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Technology and Innovation Centre, Glasgow, UK
| | - Charline J J Gerard
- CMAC Future Manufacturing Research Hub, c/o Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Technology and Innovation Centre, Glasgow, UK
| | - Elvira Pantuso
- Consiglio Nazionale delle Ricerche (CNR), Istituto per la Tecnologia delle Membrane (ITM), Rende, Italy
| | - Enrica Fontananova
- Consiglio Nazionale delle Ricerche (CNR), Istituto per la Tecnologia delle Membrane (ITM), Rende, Italy
| | - Rocco Caliandro
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia (IC), Bari, Italy
| | - Benny D Belviso
- Consiglio Nazionale delle Ricerche (CNR), Istituto di Cristallografia (IC), Bari, Italy
| | - Efrem Curcio
- Department of Environmental Engineering, University of Calabria, Rende, Italy
| | - Fiore P Nicoletta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Edificio Polifunzionale, Rende, Italy
| | - James Pullen
- FUJIFILM Diosynth Biotechnologies, Billingham, UK
| | - Wenqian Chen
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Jerry Y Y Heng
- Department of Chemical Engineering, Imperial College London, London, UK
| | - Sean Ruane
- Center for Process Innovation (CPI), Darlington, UK
| | - John Liddell
- Center for Process Innovation (CPI), Darlington, UK
| | | | - Joop H Ter Horst
- CMAC Future Manufacturing Research Hub, c/o Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Technology and Innovation Centre, Glasgow, UK
| | - Gianluca Di Profio
- Consiglio Nazionale delle Ricerche (CNR), Istituto per la Tecnologia delle Membrane (ITM), Rende, Italy.
| |
Collapse
|
45
|
Lambi AG, Popoff SN, Benhaim P, Barbe MF. Pharmacotherapies in Dupuytren Disease: Current and Novel Strategies. J Hand Surg Am 2023; 48:810-821. [PMID: 36935324 PMCID: PMC10440226 DOI: 10.1016/j.jhsa.2023.02.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/27/2022] [Accepted: 02/06/2023] [Indexed: 03/21/2023]
Abstract
Dupuytren disease is a benign, progressive fibroproliferative disorder of the hands. To date, only one pharmacotherapy (clostridial collagenase) has been approved for use in Dupuytren disease. There is a great need for additional nonsurgical methods that can be used to either avoid the risks of invasive treatments or help minimize recurrence rates following treatment. A number of nonsurgical modalities have been discussed in the past and continue to appear in discussions among hand surgeons, despite highly variable and often poor or no long-term clinical data. This article reviews many of the pharmacotherapies discussed in the treatment of Dupuytren disease and novel therapies used in inflammation and fibrosis that offer potential treatment options.
Collapse
Affiliation(s)
- Alex G Lambi
- Department of Orthopedics and Rehabilitation, University of New Mexico School of Medicine, Albuquerque, NM.
| | - Steven N Popoff
- Department of Orthopaedic Surgery, Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | - Prosper Benhaim
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA
| | - Mary F Barbe
- Department of Biomedical Education and Data Science, Lewis Katz School of Medicine at Temple University, Philadelphia, PA; Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| |
Collapse
|
46
|
Xiao Z, Murakhovskaya I. Rituximab resistance in ITP and beyond. Front Immunol 2023; 14:1215216. [PMID: 37575230 PMCID: PMC10422042 DOI: 10.3389/fimmu.2023.1215216] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/10/2023] [Indexed: 08/15/2023] Open
Abstract
The pathophysiology of immune thrombocytopenia (ITP) is complex and encompasses innate and adaptive immune responses, as well as megakaryocyte dysfunction. Rituximab is administered in relapsed cases and has the added benefit of inducing treatment-free remission in over 50% of patients. Nevertheless, the responses to this therapy are not long-lasting, and resistance development is frequent. B cells, T cells, and plasma cells play a role in developing resistance. To overcome this resistance, targeting these pathways through splenectomy and novel therapies that target FcγR pathway, FcRn, complement, B cells, plasma cells, and T cells can be useful. This review will summarize the pathogenetic mechanisms implicated in rituximab resistance and examine the potential therapeutic interventions to overcome it. This review will explore the efficacy of established therapies, as well as novel therapeutic approaches and agents currently in development.
Collapse
Affiliation(s)
| | - Irina Murakhovskaya
- Division of Hematology, Department of Hematology-Oncology, Montefiore Medical Center, Albert Einstein College of Medicine, New York City, NY, United States
| |
Collapse
|
47
|
Athni TS, Barmettler S. Hypogammaglobulinemia, late-onset neutropenia, and infections following rituximab. Ann Allergy Asthma Immunol 2023; 130:699-712. [PMID: 36706910 PMCID: PMC10247428 DOI: 10.1016/j.anai.2023.01.018] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/23/2022] [Accepted: 01/16/2023] [Indexed: 01/26/2023]
Abstract
Rituximab is a chimeric anti-CD20 monoclonal antibody that targets CD20-expressing B lymphocytes, has a well-defined efficacy and safety profile, and is broadly used to treat a wide array of diseases. In this review, we cover the mechanism of action of rituximab and focus on hypogammaglobulinemia and late-onset neutropenia-2 immune effects secondary to rituximab-and subsequent infection. We review risk factors and highlight key considerations for immunologic monitoring and clinical management of rituximab-induced secondary immune deficiencies. In patients treated with rituximab, monitoring for hypogammaglobulinemia and infections may help to identify the subset of patients at high risk for developing poor B cell reconstitution, subsequent infections, and adverse complications. These patients may benefit from early interventions such as vaccination, antibacterial prophylaxis, and immunoglobulin replacement therapy. Systematic evaluation of immunoglobulin levels and peripheral B cell counts by flow cytometry, both at baseline and periodically after therapy, is recommended for monitoring. In addition, in those patients with prolonged hypogammaglobulinemia and increased infections after rituximab use, immunologic evaluation for inborn errors of immunity may be warranted to further risk stratification, increase monitoring, and assist in therapeutic decision-making. As the immunologic effects of rituximab are further elucidated, personalized approaches to minimize the risk of adverse reactions while maximizing benefit will allow for improved care of patients with decreased morbidity and mortality.
Collapse
Affiliation(s)
| | - Sara Barmettler
- Allergy and Clinical Immunology Unit, Division of Rheumatology, Allergy, and Immunology, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
48
|
Deng L, Xu G. Update on the Application of Monoclonal Antibody Therapy in Primary Membranous Nephropathy. Drugs 2023; 83:507-530. [PMID: 37017915 DOI: 10.1007/s40265-023-01855-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 04/06/2023]
Abstract
When first introduced, rituximab (RTX), a chimeric anti-CD20 monoclonal antibody, brought about an alternative therapeutic paradigm for primary membranous nephropathy (PMN). Rituximab was shown to be effective and safe in PMN patients with kidney dysfunction, with. patients receiving second-line rituximab therapy achieving remission as effectively as those patients who had not previously received immunotherapy. No safety issues were reported. The B cell-driven protocol seems to be as efficient as the 375 mg/m2 × 4 regimen or 1 g × 2 regimen in achieving B cell depletion and remission, but patients with high M-type phospholipase A2 receptor (PLA2R) antibody levels may benefit from a higher dose of rituximab. While rituximab added another therapeutic option to the treatment regimen, it does have limitations as 20 to 40% of patients do not respond. Not all patients respond to RTX therapy for lymphoproliferative disorders either, therefore further novel anti-CD20 monoclonal antibodies have been developed and these may provide alternative therapeutic options for PMN. Ofatumumab, a fully human monoclonal antibody, specifically recognizes an epitope encompassing both the small and large extracellular loops of the CD20 molecule, resulting in increased complement-dependent cytotoxic activity. Ocrelizumab binds an alternative but overlapping epitope region to rituximab and displays enhanced antibody-dependent cellular cytotoxic (ADCC) activities. Obinutuzumab is designed to have a modified elbow-hinge amino acid sequence, leading to increased direct cell death induction and ADCC activities. In PMN clinical studies, ocrelizumab and obinutuzumab showed promising results, while ofatumumab displayed mixed results. However, there is a lack of randomized controlled trials with large samples, especially direct head-to-head comparisons. Alternative molecular mechanisms have been suggested in this context to explore novel therapeutic strategies. B cell activator-targeted, plasma cell-targeted and complement-directed treatments may lead to novel therapy paradigms for PMN. Exploratory strategies for the use of drugs with different mechanisms, such as a combination of rituximab and cyclophosphamide and a steroid, a combination of rituximab and a calcineurin inhibitor, may provide more rapid and efficient remission, but the combination of standard immunosuppression with rituximab could increase infection risk.
Collapse
Affiliation(s)
- Le Deng
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Gaosi Xu
- Department of Nephrology, The Second Affiliated Hospital of Nanchang University, No. 1, Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
49
|
DeFilippis EM, Kransdorf EP, Jaiswal A, Zhang X, Patel J, Kobashigawa JA, Baran DA, Kittleson MM. Detection and management of HLA sensitization in candidates for adult heart transplantation. J Heart Lung Transplant 2023; 42:409-422. [PMID: 36631340 DOI: 10.1016/j.healun.2022.12.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/13/2022] [Accepted: 12/17/2022] [Indexed: 12/28/2022] Open
Abstract
Heart transplantation (HT) remains the preferred therapy for patients with advanced heart failure. However, for sensitized HT candidates who have antibodies to human leukocyte antigens , finding a suitable donor can be challenging and can lead to adverse waitlist outcomes. In recent years, the number of sensitized patients awaiting HT has increased likely due to the use of durable and mechanical circulatory support as well as increasing number of candidates with underlying congenital heart disease. This State-of-the-Art review discusses the assessment of human leukocyte antigens antibodies, potential desensitization strategies including mechanisms of action and specific protocols, the approach to a potential donor including the use of complement-dependent cytotoxicity, flow cytometry, and virtual crossmatches, and peritransplant induction management.
Collapse
Affiliation(s)
- Ersilia M DeFilippis
- Center for Advanced Cardiac Care, Division of Cardiology, Columbia University Irving Medical Center, New York, New York
| | - Evan P Kransdorf
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Abhishek Jaiswal
- Hartford HealthCare Heart and Vascular Institute, Hartford Hospital, Hartford, Connecticut
| | - Xiaohai Zhang
- HLA and Immunogenetics Laboratory, Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jignesh Patel
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jon A Kobashigawa
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - David A Baran
- Cleveland Clinic, Heart Vascular and Thoracic Institute, Weston, Florida
| | | |
Collapse
|
50
|
Brzezicka KA, Paulson JC. Impact of Siglecs on autoimmune diseases. Mol Aspects Med 2023; 90:101140. [PMID: 36055802 PMCID: PMC9905255 DOI: 10.1016/j.mam.2022.101140] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/19/2022] [Accepted: 08/21/2022] [Indexed: 02/08/2023]
Abstract
Autoimmune diseases affect tens of millions of people just in the United States alone. Most of the available treatment options are aimed at reducing symptoms but do not lead to cures. Individuals affected with autoimmune diseases suffer from the imbalance between tolerogenic and immunogenic functions of their immune system. Often pathogenesis is mediated by autoreactive B and T cells that escape central tolerance and react against self-antigens attacking healthy tissues in the body. In recent years Siglecs, sialic-acid-binding immunoglobulin (Ig)-like lectins, have gained attention as immune checkpoints for therapeutic interventions to dampen excessive immune responses and to restore immune tolerance in autoimmune diseases. Many Siglecs function as inhibitory receptors suppressing activation signals in various immune cells through binding to sialic acid ligands as signatures of self. In this review, we highlight potential of Siglecs in suppressing immune responses causing autoimmune diseases. In particular, we cover the roles of CD22 and Siglec-G/Siglec-10 in regulating autoreactive B cell responses. We discuss several functions of Siglec-10 in the immune modulation of other immune cells, and the potential of therapeutic strategies for restoring immune tolerance by targeting Siglecs and expanding regulatory T cells. Finally, we briefly review efforts evaluating Siglec-based biomarkers to monitor autoimmune diseases.
Collapse
Affiliation(s)
- Katarzyna Alicja Brzezicka
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - James C Paulson
- Department of Molecular Medicine, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA; Department of Immunology and Microbiology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA.
| |
Collapse
|