1
|
Riviere C, Aljieli M, Mévélec MN, Lantier L, Boursin F, Lajoie L, Ducournau C, Germon S, Moiré N, Dimier-Poisson I, Aubrey N, di Tommaso A. Neospora caninum as delivery vehicle for anti-PD-L1 scFv-Fc: A novel approach for cancer immunotherapy. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200968. [PMID: 40236994 PMCID: PMC11999461 DOI: 10.1016/j.omton.2025.200968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/29/2024] [Accepted: 03/16/2025] [Indexed: 04/17/2025]
Abstract
Neospora caninum, a potential anticancer agent able to reactivate the immune response within the tumor microenvironment (TME), has recently shown enhanced immunomodulatory properties in different tumor models when armed with the cytokine, Il-15. In the current area of combination immunotherapy strategies designed to overcome treatment resistance, we engineered for the first time the protozoan Neospora caninum to vectorize and secrete a single-chain variable fragment fused to fragment crystallizable region (scFv-Fc) targeting human programmed cell death ligand 1 (PD-L1). Following validation of its secretion through the micronemes (protozoa secretory organelles), we demonstrated that the scFv-Fc could bind PD-L1 on mouse and human tumor cells, block the programmed cell death protein 1 (PD-1)/PD-L1 pathway leading to potentiate the T cell lymphocyte activity. Additionally, the scFv-Fc induced antibody-dependent cellular phagocytosis (ADCP) and antibody-dependent cellular cytotoxicity (ADCC). Those data demonstrate the feasibility of vectoring and secreting a functional antibody fragment by N. caninum, opening promising avenues for future research.
Collapse
Affiliation(s)
- Clément Riviere
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Muna Aljieli
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
- Faculty of Pharmacy, University of Gezira, Wad Madani, Sudan
| | | | - Louis Lantier
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Fanny Boursin
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Laurie Lajoie
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Céline Ducournau
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Stéphanie Germon
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Nathalie Moiré
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | | | - Nicolas Aubrey
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| | - Anne di Tommaso
- BioMAP, UMR ISP 1282 INRAe – Université de Tours, 37200 Tours, France
| |
Collapse
|
2
|
Shibutani Y, Kawanobe A, Suzuki S, Imaoka T, Tajiri K. Effects of Immune Checkpoint Inhibitor-induced Thyroid Dysfunction on Cardiac Troponin Levels. J Immunother 2025; 48:183-188. [PMID: 40333085 DOI: 10.1097/cji.0000000000000555] [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: 09/10/2024] [Accepted: 03/04/2025] [Indexed: 03/27/2025]
Abstract
Immune checkpoint inhibitor (ICI)-induced thyroid dysfunction is the most frequent endocrine immune-related adverse event (irAE). Thyroid hormones have various effects on the cardiovascular system; however, the impact of thyroid irAEs on the development of cardiovascular diseases is not fully understood. This retrospective study included 94 patients who received ICIs and had thyroid function and troponin T levels, markers of cardiac damage, measured at the National Cancer Center Hospital East between January 2017 and July 2022. Of the 94 patients, 36 (38%) showed elevated troponin levels after ICI treatment during the follow-up period. The median observation period was 249 days (interquartile range, 124-502 d). Thyroid irAEs [hypothyroidism (n=13) and hyperthyroidism (n=3)] associations were found in 16 (44%) of these 36 patients. None of the patients developed overt cardiovascular disease or died of heart disease, regardless of whether they experienced thyroid irAEs. The troponin levels increased with increasing thyroid stimulating hormone (TSH) levels. In particular, troponin levels were significantly elevated in patients with TSH >20 μIU/mL after ICI treatment ( P =0.009). In conclusion, thyroid irAEs may cause cardiac damage indicated by elevated troponin levels, necessitating special attention, particularly in cases of hypothyroidism where TSH exceeds 20 μIU/mL. Therefore, it is important to monitor cardiac markers along with thyroid function after ICI treatment.
Collapse
Affiliation(s)
- Yuma Shibutani
- Department of Pharmacy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Atsushi Kawanobe
- Department of Pharmacy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Shinya Suzuki
- Department of Pharmacy, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takuro Imaoka
- Department of Cardiology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Kazuko Tajiri
- Department of Cardiology, National Cancer Center Hospital East, Kashiwa, Japan
- Tsukuba Life Science Innovation Program (T-LSI), School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
3
|
Fankhauser RG, Johnson DB, Moslehi JJ, Balko JM. Preclinical mouse models of immune checkpoint inhibitor-associated myocarditis. NATURE CARDIOVASCULAR RESEARCH 2025:10.1038/s44161-025-00640-2. [PMID: 40335724 DOI: 10.1038/s44161-025-00640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/10/2025] [Indexed: 05/09/2025]
Abstract
In this Review, we present a comprehensive analysis of preclinical models used to study immune checkpoint inhibitor-associated myocarditis (hereafter ICI-myocarditis), a potentially lethal immune-related adverse event. We begin by providing an overview of immune checkpoint inhibitors, highlighting how their efficacy in cancer treatment is counterbalanced by their predisposition to cause immune-related adverse events. Next, we draw from human data to identify disease features that an effective mouse model should ideally mimic. After that, we present a critical evaluation of a wide variety of existing mouse models including genetic, pharmacological and humanized models. We summarize insights gathered about the underlying mechanisms of ICI-myocarditis and the role of mouse models in these discoveries. We conclude with a perspective on the future of preclinical models, highlighting potential model improvements and research directions that could strengthen our understanding of ICI-myocarditis, ultimately improving patient outcomes.
Collapse
Grants
- 5R01HL156021-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL155990-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL156021-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL155990-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL141466-05 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH P01 HL141084 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH R01 HL160688 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01CA227481-05 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 5P30CA068485-29 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 5R01CA227481-05 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- T32GM007347 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 25PRE1375723 American Heart Association (American Heart Association, Inc.)
Collapse
Affiliation(s)
- Reilly G Fankhauser
- Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Cardiovascular Research Institute, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Justin M Balko
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
Dia AK, Kolnohuz A, Yolchuyeva S, Tonneau M, Lamaze F, Orain M, Gagné A, Blais F, Coulombe F, Malo J, Belkaid W, Elkrief A, Williamson D, Routy B, Joubert P, Laplante M, Bilodeau S, Manem VS. Computational analysis of whole slide images predicts PD-L1 expression and progression-free survival in immunotherapy-treated non-small cell lung cancer patients. J Transl Med 2025; 23:510. [PMID: 40329352 PMCID: PMC12056990 DOI: 10.1186/s12967-025-06487-2] [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: 09/26/2024] [Accepted: 04/13/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by significantly improving the efficacy of treatments and tolerability for patients with non-small cell lung cancer (NSCLC). However, even after meticulous selection based on molecular criteria, only 20-30% of the patients respond to ICIs. This highlights the urgent clinical need to develop more precise biomarkers to better identify individuals who will benefit from these expensive therapies. METHODS Data from NSCLC patients treated with immunotherapy were collected from two institutions. From the histological images of tumors, pathomics features were extracted. We employed six machine learning models and seven feature selection methods to predict expression of the programmed death-ligand 1 (PD-L1), a current biomarker used to select patients for immunotherapy, and progression-free survival (PFS). The association between pathomics features and biological pathways was explored to validate pathomics-based signatures. We performed gene set enrichment analysis to identify the pathways enriched with the predictive signatures. RESULTS Handcrafted histological features were extracted from the whole slide images (WSI). The Support Vector Machines model with the SurfStar feature selection method, offered the best results, with an area under the curve (AUC) of around 0.66 for both the training and validation sets to predict PD-L1. For the prediction of PFS, the most effective model was linear discriminant analysis using the Multi Surf feature selection method with an AUC of 0.71 for the training set and 0.62 for the validation set. We found immune pathways to be upregulated in the high PD-L1 and high PFS groups, confirming the utility of image analysis for predicting clinical endpoints in patients treated with immunotherapy. CONCLUSION Our models, based on the analysis of histological images, can serve as predictive biomarkers for PD-L1 and PFS. This approach, focused on histological images, enables the distinction of patients based on treatment response, thus providing clinicians with a valuable tool for patient management. With further validation on external cohorts, these models could enhance clinical decision-making through analysis of routine medical images.
Collapse
Affiliation(s)
- Abdou Khadir Dia
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
| | - Alona Kolnohuz
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Université Laval, Québec, Canada
| | - Sevinj Yolchuyeva
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Marion Tonneau
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
- Université de médecine de Lille, Lille, France
| | - Fabien Lamaze
- Quebec Heart & Lung Institute Research Center, Québec, Canada
| | - Michele Orain
- Quebec Heart & Lung Institute Research Center, Québec, Canada
| | | | | | | | - Julie Malo
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Wiam Belkaid
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Arielle Elkrief
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Drew Williamson
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, USA
| | - Bertrand Routy
- Centre de Recherche du Centre Hospitalier Universitaire de Montréal, Montréal, Canada
| | - Philippe Joubert
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
| | - Mathieu Laplante
- Quebec Heart & Lung Institute Research Center, Québec, Canada
- Université Laval, Québec, Canada
| | - Steve Bilodeau
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada
- Cancer Research Center, Université Laval, Québec, Canada
- Big Data Research Center, Université Laval, Québec, Canada
| | - Venkata Sk Manem
- Centre de Recherche du CHU de Québec - Université Laval, Québec, Canada.
- Quebec Heart & Lung Institute Research Center, Québec, Canada.
- Department of Molecular Biology, Medical Biochemistry and Pathology, Université Laval, Québec, Canada.
- Cancer Research Center, Université Laval, Québec, Canada.
- Big Data Research Center, Université Laval, Québec, Canada.
| |
Collapse
|
5
|
Alghabban A, Corke L, Katzberg H, Bril V, Barnett-Tapia C, Mason W, Alothman R, Albiruni Ryan AR, Hogg D, Sridhar S, Dhani N, Spreafico A, Eng L, Sacher A, Bradbury P, Liu G, Leighl N, Shepherd FA. Myasthenia Gravis in Patients Treated With Immune Checkpoint Inhibitors. JTO Clin Res Rep 2025; 6:100772. [PMID: 40242666 PMCID: PMC12002810 DOI: 10.1016/j.jtocrr.2024.100772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 04/18/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have improved outcomes significantly for patients across multiple tumor types, and now are being used in combination with other therapies and in earlier settings where treatment intent is curative. Immune-related adverse events occur commonly and there are clear guidelines regarding management. Neurological toxicities such as myasthenia gravis (MG) with or without myositis are rare but are associated with high morbidity and mortality. Methods This single-centre study presents a series of patients treated with ICIs who subsequently developed immune-related MG. Presenting symptoms, treatments and outcomes were abstracted from retrospective chart review. Results We identified 16 patients (9 thoracic malignancies, 7 other tumor sites) who were diagnosed with MG after one or more cycles of ICI. Eleven had overlapping myositis. The median time from the first ICI treatment to the onset of symptoms was 49 days (range 17-361). All patients received steroids (prednisone 1-2 mg/kg); six required other immunosuppressive agents, and five underwent plasma exchange. Only two patients had complete resolution, eight improved with residual symptoms, two experienced initial improvement followed by deterioration, and four worsened despite treatment. Six patients died as a result of myasthenia-related complications (38%), three from progressive cancer (19%) and seven remain alive at the time of review (44%). Conclusion ICI-related MG is a rare and potentially fatal adverse event. Diagnosis and management remain a challenge, especially with negative serological markers and in the presence of overlapping syndromes with high mortality rates. Prompt recognition and multimodality treatment are key. Clinicians should have a low threshold for diagnosis and early management.
Collapse
Affiliation(s)
- Abdulrahman Alghabban
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lucy Corke
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Hans Katzberg
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Vera Bril
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Carolina Barnett-Tapia
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Warren Mason
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Raed Alothman
- Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Abdul Razak Albiruni Ryan
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - David Hogg
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Srikala Sridhar
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Neesha Dhani
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Lawson Eng
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Penelope Bradbury
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Natasha Leighl
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Frances A. Shepherd
- Princess Margaret Cancer Centre, University Health Network, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
6
|
Szabados BE, Guerrero-Ramos F, Grande E, Grivas P, Grünwald V, Miguel MC, Hussain SA, Kulkarni GS, Wilson AL, Shore ND, Sridhar SS, Hoyt M, Strumeier S, Sutton J, Brinkmann J, Teresi RE, Todenhöfer T. On the Horizon: A Global Multidisciplinary Perspective on Delivering Emerging Therapies for Patients with BCG-Naïve High-Risk NMIBC. Oncol Ther 2025:10.1007/s40487-025-00334-6. [PMID: 40246795 DOI: 10.1007/s40487-025-00334-6] [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: 01/29/2025] [Accepted: 03/20/2025] [Indexed: 04/19/2025] Open
Abstract
Patients with high-risk non-muscle invasive bladder cancer (NMIBC) are generally treated with transurethral resection of the bladder tumor followed by intravesical bacillus Calmette-Guérin (BCG), the current standard of care. However, recurrence or progression is common and may result in patients requiring radical cystectomy. Additionally, BCG continues to be in short supply worldwide. Therefore, there is an unmet need for new therapies that provide durable disease control and maintain quality of life. In the BCG-naïve high-risk NMIBC setting, potential new treatment options are emerging, with several regimens combining intravesical therapy with systemic PD-1 or PD-L1-directed immune checkpoint inhibitors (ICIs) currently under investigation in several Phase 3 trials. In routine clinical practice, NMIBC has traditionally been managed almost entirely by urologists. However, the introduction of systemic ICIs would likely require medical oncology expertise to help assess patients' fitness for these therapies and potentially for treatment administration and immune-related adverse event management. While multidisciplinary workflows are common practice for advanced bladder cancer, they would represent a paradigm shift in NMIBC. Based on current experience of managing patients with NMIBC across different countries and healthcare systems from our perspective as urologists, medical oncologists, and nurses, we discuss best practices for the potential integration of emerging therapies such as ICIs into the treatment of BCG-naïve high-risk NMIBC. We emphasize the need for multidisciplinary care, either through formalized multidisciplinary teams or cross-discipline collaborative workflows adapted to local needs, to ensure efficient coordination and sharing of responsibilities. Specialized nurses have the potential to play key roles across multiple aspects of patient care. We also highlight the crucial importance of effective communication across teams, increases in resourcing, and education for healthcare professionals, patients, and caregivers to enable eligible patients with high-risk NMIBC to benefit optimally from the introduction of these potential new treatment options. Supplementary file2 (MP4 407382 kb).
Collapse
Affiliation(s)
- Bernadett E Szabados
- Centre for Experimental Cancer Medicine, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
- University College London Hospital NHS Foundation Trust, London, UK.
| | | | - Enrique Grande
- Medical Oncology Department, MD Anderson Cancer Center Madrid, Madrid, Spain
| | - Petros Grivas
- Fred Hutchinson Cancer Center, University of Washington, Seattle, WA, USA
| | - Viktor Grünwald
- Department for Medical Oncology, Department of Urology, University Hospital Essen, Essen, Germany
| | | | - Syed A Hussain
- School of Medicine and Population Health, University of Sheffield, Sheffield, UK
| | - Girish S Kulkarni
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Ana Lisa Wilson
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Neal D Shore
- AUC Urology Specialists, Myrtle Beach, SC, USA
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | - Srikala S Sridhar
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Mary Hoyt
- Carolina Urologic Research Center, Myrtle Beach, SC, USA
| | | | | | | | | | | |
Collapse
|
7
|
Shires CB, Boughter JD, Golembeski CP, Sharp NM, Hall AT, Sebelik ME. Delayed regional metastasis from small face/scalp cutaneous squamous carcinoma: outcomes and predictors. Arch Dermatol Res 2025; 317:703. [PMID: 40220149 PMCID: PMC11993493 DOI: 10.1007/s00403-025-04224-9] [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: 01/10/2025] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025]
Abstract
OBJECTIVE CSCC (cutaneous squamous cell carcinoma) of the face/scalp generally has a favorable outcome. However, patients with delayed regional metastasis from scalp/facial CSCC do very poorly. Historically, prophylactic treatment of the N0 neck for the T1-T2 (as staged in the AJCC 7th edition) face/scalp cancers has not been recommended. This study aims to determine predictors of delayed regional metastases in < 4 cm CSCC of the face and scalp. METHODS We retrospectively analyzed medical records of patients diagnosed regional metastasis of CSCC of the scalp/face at least 6 months after the diagnosis of the primary CSCC at a tertiary academic head and neck clinic during a period between 2012 and 2018. RESULTS A total of 50 patients with CSCC of the face/scalp were diagnosed and treated for delayed cervical lymph node metastasis during a 6-year period. All patients were male. Areas affected were the scalp, cheeks, and forehead. No patient had neck lymph node metastases at initial presentation for their scalp/face cancer. The presence of perineural invasion in the primary tumor was the only observed predictor of delayed neck lymph node recurrence. We did not observe an association between diameter/depth of the primary skin tumor or lymphovascular invasion and the presence of regional lymph node recurrence. CONCLUSION In our review, perineural invasion (PNI) was the one factor predictive of delayed regional lymph node metastasis. This observation confirms the use of PNI to upstage even small CSCC to T3. Prophylactic treatment of the neck in patients with small scalp/face cancers with PNI should be strongly considered, especially in the age of immunotherapy.
Collapse
Affiliation(s)
| | - John D Boughter
- Department of Neuroanatomy, University of Tennessee, Memphis, TN, USA
| | | | - Nicholas M Sharp
- Baptist University College of Osteopathic Medicine, Memphis, TN, USA
| | | | - Merry E Sebelik
- Department of Otolaryngology, Emory University, Atlanta, GA, USA
| |
Collapse
|
8
|
Ton Nu QC, Deka G, Park PH. CD8 + T cell-based immunotherapy: Promising frontier in human diseases. Biochem Pharmacol 2025; 237:116909. [PMID: 40179991 DOI: 10.1016/j.bcp.2025.116909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 02/28/2025] [Accepted: 03/26/2025] [Indexed: 04/05/2025]
Abstract
The abundant cell components of the adaptive immune system called T lymphocytes (T cells) play important roles in mediating immune responses to eliminate the invaders and create the memory of the germs to form a new immunity for the next encounter. Among them, cytotoxic T cells expressing cell-surface CD8 are the most critical effector cells that directly eradicate the target infected cells by recognizing antigens presented by major histocompatibility complex class I molecules to protect our body from pathological threats. In the continuous evolution of immunotherapy, various CD8+ T cell-based therapeutic strategies have been developed based on the role and molecular concept of CD8+ T cells. The emergence of such remarkable therapies provides promising hope for multiple human disease treatments such as autoimmunity, infectious disease, cancer, and other non-infectious diseases. In this review, we aim to discuss the current knowledge on the utilization of CD8+ T cell-based immunotherapy for the treatment of various diseases, the molecular basis involved, and its limitations. Additionally, we summarize the recent advances in the use of CD8+ T cell-based immunotherapy and provide a comprehensive overview of CD8+ T cells, including their structure, underlying mechanism of function, and markers associated with CD8+ T cell exhaustion. Building upon these foundations, we delineate the advancement of CD8+ T cell-based immunotherapies with fundamental operating principles followed by research studies, and challenges, as well as illustrate human diseases involved in this development.
Collapse
Affiliation(s)
- Quynh Chau Ton Nu
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Gitima Deka
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea
| | - Pil-Hoon Park
- College of Pharmacy, Yeungnam University, Gyeongsan, Republic of Korea; Research institute of cell culture, Yeungnam University, Gyeongsan, Republic of Korea.
| |
Collapse
|
9
|
Gray R, Manisty C, Cheng R, Dastidar A, Mamas M, Ghosh A. Immune checkpoint inhibitors: Unravelling atherosclerotic cardiovascular risk. Atherosclerosis 2025; 403:119147. [PMID: 40037087 DOI: 10.1016/j.atherosclerosis.2025.119147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Affiliation(s)
| | | | - Richard Cheng
- Division of Cardiology, University of Washington, USA
| | - Amardeep Dastidar
- Bristol Heart Institute, NIHR Biomedical Research Centre Cardiovascular & North Bristol NHS Trust, Bristol, UK
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, UK
| | | |
Collapse
|
10
|
Jensen G, Wang X, Kuempel J, Palaskas N, Chen Z, Yu W, Chen Y, Mohammad H, Luo W, Chang J. Immune checkpoint inhibitor-associated myocarditis: a historical and comprehensive review. Am J Physiol Heart Circ Physiol 2025; 328:H734-H751. [PMID: 39925096 DOI: 10.1152/ajpheart.00687.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025]
Abstract
The most fatal side effect associated with revolutionary immune checkpoint inhibitor (ICI) cancer therapies is myocarditis, a rare and devastating complication with a mortality rate approaching 40%. This review comprehensively examines the limited knowledge surrounding this recently recognized condition, emphasizing the absence of evidence-based therapeutic strategies, diagnostic modalities, and reliable biomarkers that hinder effective management. It explores advancements in preclinical models that are uncovering disease mechanisms and enabling the identification of therapeutic targets. These efforts have informed the design of early clinical trials aimed at reducing mortality. With the growing prevalence of ICI therapies in oncology, addressing critical gaps, such as long-term outcomes and risk stratification, has become increasingly urgent. By synthesizing current evidence, this work seeks to enhance understanding and guide the development of strategies to improve patient outcomes and ensure the continued safe use of ICIs in cancer care.
Collapse
Affiliation(s)
- Garrett Jensen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Xinjie Wang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jacob Kuempel
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Nicolas Palaskas
- Department of Cardiology, MD Anderson Cancer Center, Houston, Texas, United States
| | - Zhishi Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Wei Yu
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Yanping Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Haseeb Mohammad
- Texas A&M University College of Medicine, Houston, Texas, United States
| | - Weijia Luo
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jiang Chang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| |
Collapse
|
11
|
Ravera F, Gilardi N, Ballestrero A, Zoppoli G. Applications, challenges and future directions of artificial intelligence in cardio-oncology. Eur J Clin Invest 2025; 55 Suppl 1:e14370. [PMID: 40191923 PMCID: PMC11973867 DOI: 10.1111/eci.14370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/28/2024] [Indexed: 04/09/2025]
Abstract
BACKGROUND The management of cardiotoxicity related to cancer therapies has emerged as a significant clinical challenge, prompting the rapid growth of cardio-oncology. As cancer treatments become more complex, there is an increasing need to enhance diagnostic and therapeutic strategies for managing their cardiovascular side effects. OBJECTIVE This review investigates the potential of artificial intelligence (AI) to revolutionize cardio-oncology by integrating diverse data sources to address the challenges of cardiotoxicity management. METHODS We explore applications of AI in cardio-oncology, focusing on its ability to leverage multiple data sources, including electronic health records, electrocardiograms, imaging modalities, wearable sensors, and circulating serum biomarkers. RESULTS AI has demonstrated significant potential in improving risk stratification and longitudinal monitoring of cardiotoxicity. By optimizing the use of electrocardiograms, non-invasive imaging, and circulating biomarkers, AI facilitates earlier detection, better prediction of outcomes, and more personalized therapeutic interventions. These advancements are poised to enhance patient outcomes and streamline clinical decision-making. CONCLUSIONS AI represents a transformative opportunity in cardio-oncology by advancing diagnostic and therapeutic capabilities. However, successful implementation requires addressing practical challenges such as data integration, model interpretability, and clinician training. Continued collaboration between clinicians and AI developers will be essential to fully integrate AI into routine clinical workflows.
Collapse
Affiliation(s)
- Francesco Ravera
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
| | - Nicolò Gilardi
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| | - Gabriele Zoppoli
- Department of Internal Medicine and Medical SpecialtiesUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San MartinoGenoaItaly
| |
Collapse
|
12
|
Jiao F, Shang Z, Lu H, Chen P, Chen S, Xiao J, Zhang F, Zhang D, Lv C, Han Y. A weakly supervised deep learning framework for automated PD-L1 expression analysis in lung cancer. Front Immunol 2025; 16:1540087. [PMID: 40230846 PMCID: PMC11994606 DOI: 10.3389/fimmu.2025.1540087] [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: 12/05/2024] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
The growing application of immune checkpoint inhibitors (ICIs) in cancer immunotherapy has underscored the critical need for reliable methods to identify patient populations likely to respond to ICI treatments, particularly in lung cancer treatment. Currently, the tumor proportion score (TPS), a crucial biomarker for patient selection, relies on manual interpretation by pathologists, which often shows substantial variability and inconsistency. To address these challenges, we innovatively developed multi-instance learning for TPS (MiLT), an innovative artificial intelligence (AI)-powered tool that predicts TPS from whole slide images. Our approach leverages multiple instance learning (MIL), which significantly reduces the need for labor-intensive cell-level annotations while maintaining high accuracy. In comprehensive validation studies, MiLT demonstrated remarkable consistency with pathologist assessments (intraclass correlation coefficient = 0.960, 95% confidence interval = 0.950-0.971) and robust performance across both internal and external cohorts. This tool not only standardizes TPS evaluation but also adapts to various clinical standards and provides time-efficient predictions, potentially transforming routine pathological practice. By offering a reliable, AI-assisted solution, MiLT could significantly improve patient selection for immunotherapy and reduce inter-observer variability among pathologists. These promising results warrant further exploration in prospective clinical trials and suggest new possibilities for integrating advanced AI in pathological diagnostics. MiLT represents a significant step toward more precise and efficient cancer immunotherapy decision-making.
Collapse
Affiliation(s)
- Feng Jiao
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zhanxian Shang
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Hongmin Lu
- Department of Oncology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peilin Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Shiting Chen
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Jiayi Xiao
- School of Life Science and Technology, Tongji University, Shanghai, China
| | - Fuchuang Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Dadong Zhang
- Department of Clinical and Translational Medicine, 3D Medicines Inc., Shanghai, China
| | - Chunxin Lv
- Department of Oncology, Shanghai Punan Hospital of Pudong New District, Shanghai, China
| | - Yuchen Han
- Department of Pathology, Shanghai Chest Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| |
Collapse
|
13
|
Guo Y, Wang X, Zhang C, Chen W, Fu Y, Yu Y, Chen Y, Shao T, Zhang J, Ding G. Tumor Immunotherapy Targeting B7-H3: From Mechanisms to Clinical Applications. Immunotargets Ther 2025; 14:291-320. [PMID: 40171330 PMCID: PMC11960814 DOI: 10.2147/itt.s507522] [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: 11/20/2024] [Accepted: 03/16/2025] [Indexed: 04/03/2025] Open
Abstract
B7-H3 (CD276) is an immune checkpoint from the B7 family of molecules and is abnormally expressed in tumor cells as a co-inhibitory molecule to promote tumor progression. Within the tumor microenvironment (TME), B7-H3 promotes tumor progression by impairing the T cell response, driving the polarization of tumor-associated macrophages (TAMs) to M2 phenotype, and inhibiting the function of other immune cells. In addition, B7-H3 promotes tumor cell proliferation, migration, invasion, metabolism disorder, angiogenesis, and resistance to treatment to promote tumor progression through its non-immunological functions. Immunotherapy targeting B7-H3, as well as combinations with other immune checkpoint therapies, have shown certain efficacy. In this review, we synthesizes the expression of B7-H3 and its mechanism to promote tumor progression through inducing immunomodulation and non-immunological functions, as well as its role of B7-H3 in tumor therapy, aiming to provide a reference for the clinical treatment of tumors.
Collapse
Affiliation(s)
- Yining Guo
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Xudong Wang
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Chen Zhang
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Weiwu Chen
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Yutian Fu
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Yanlan Yu
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Yicheng Chen
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Tiejuan Shao
- College of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, People’s Republic of China
| | - Jie Zhang
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
| | - Guoqing Ding
- Department of Urology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, People’s Republic of China
- National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310028, People’s Republic of China
| |
Collapse
|
14
|
Basak U, Mukherjee S, Chakraborty S, Sa G, Dastidar SG, Das T. In-silico analysis unveiling the role of cancer stem cells in immunotherapy resistance of immune checkpoint-high pancreatic adenocarcinoma. Sci Rep 2025; 15:10355. [PMID: 40133473 PMCID: PMC11937529 DOI: 10.1038/s41598-025-93924-3] [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: 11/06/2024] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Although immune checkpoint (IC) inhibition is a major treatment modality in cancer-immunotherapy, multiple cancers show low response. Our in-silico exploration by mining cancer datasets using R2, available clinical trial data, and Kaplan-Meier analysis from GEPIA depicted that unlike low-responder (LR) cancers, high-responder (HR) cancers furnish higher IC expression, that upon lowering may provide better prognosis. Contrastingly, pancreatic adenocarcinoma (PAAD) demonstrated high IC expression but low immunotherapy-response. Infiltration scores from TIMER2.0 revealed higher pro-tumor immune subsets and cancer-associated fibroblasts (CAFs) while depicting lower anti-tumor immune subsets in PAAD as compared to HR lung adenocarcinoma (LUAD). Additionally, bioinformatic tool cBioportal showed lesser tumor mutational burden, mismatch repair deficiency and greater percent of driver mutations in TP53, KRAS and CDKN2A in PAAD, supporting its higher immunotherapy-resistance than LUAD. Our search for the 'key' immunotherapy response-deciding factor(s) revealed cancer stem cells (CSCs), the known contributors of therapy-resistance and immuno-evasion, to be positively correlated with above-mentioned driver mutations, pro-tumor immune and CAF subsets; and that PAAD furnished higher expression of CSC genes than LUAD. UMAP/tSNE analyses revealed that high CSC signature is positively correlated with immunotherapy-resistance genes and pro-cancer immune cells, while negatively with cytotoxic-T cells in PAAD. Our in-silico study explains the low immunotherapy-response in high IC-expressing PAAD, wherein CSC plays a pivotal role. Further exploration portrayed correlation of CSCs with immunotherapy-resistance in other LR and HR cancers too, substantiating the need for personalized CSC evaluation and targeting for successful immunotherapy outcomes.
Collapse
Affiliation(s)
- Udit Basak
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sumon Mukherjee
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Sourio Chakraborty
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Gaurisankar Sa
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India
| | - Shubhra Ghosh Dastidar
- Bose Institute, Unified Academic Campus, EN 80, Sector V, Bidhannagar, Kolkata, 700091, India.
| | - Tanya Das
- Bose Institute, Centenary Campus, P-1/12, CIT Scheme VII M, Kolkata, 700054, India.
| |
Collapse
|
15
|
Palma M. Advancing Breast Cancer Treatment: The Role of Immunotherapy and Cancer Vaccines in Overcoming Therapeutic Challenges. Vaccines (Basel) 2025; 13:344. [PMID: 40333213 PMCID: PMC12030785 DOI: 10.3390/vaccines13040344] [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: 02/11/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 05/09/2025] Open
Abstract
Breast cancer (BC) remains a significant global health challenge due to its complex biology, which complicates both diagnosis and treatment. Immunotherapy and cancer vaccines have emerged as promising alternatives, harnessing the body's immune system to precisely target and eliminate cancer cells. However, several key factors influence the selection and effectiveness of these therapies, including BC subtype, tumor mutational burden (TMB), tumor-infiltrating lymphocytes (TILs), PD-L1 expression, HER2 resistance, and the tumor microenvironment (TME). BC subtypes play a critical role in shaping treatment responses. Triple-negative breast cancer (TNBC) exhibits the highest sensitivity to immunotherapy, while HER2-positive and hormone receptor-positive (HR+) subtypes often require combination strategies for optimal outcomes. High TMB enhances immune responses by generating neoantigens, making tumors more susceptible to immune checkpoint inhibitors (ICIs); whereas, low TMB may indicate resistance. Similarly, elevated TIL levels are associated with better immunotherapy efficacy, while PD-L1 expression serves as a key predictor of checkpoint inhibitor success. Meanwhile, HER2 resistance and an immunosuppressive TME contribute to immune evasion, highlighting the need for multi-faceted treatment approaches. Current breast cancer immunotherapies encompass a range of targeted treatments. HER2-directed therapies, such as trastuzumab and pertuzumab, block HER2 dimerization and enhance antibody-dependent cellular cytotoxicity (ADCC), while small-molecule inhibitors, like lapatinib and tucatinib, suppress HER2 signaling to curb tumor growth. Antibody-drug conjugates (ADCs) improve tumor targeting by coupling monoclonal antibodies with cytotoxic agents, minimizing off-target effects. Meanwhile, ICIs, including pembrolizumab, restore T-cell function, and CAR-macrophage (CAR-M) therapy leverages macrophages to reshape the TME and overcome immunotherapy resistance. While immunotherapy, particularly in TNBC, has demonstrated promise by eliciting durable immune responses, its efficacy varies across subtypes. Challenges such as immune-related adverse events, resistance mechanisms, high costs, and delayed responses remain barriers to widespread success. Breast cancer vaccines-including protein-based, whole-cell, mRNA, dendritic cell, and epitope-based vaccines-aim to stimulate tumor-specific immunity. Though clinical success has been limited, ongoing research is refining vaccine formulations, integrating combination therapies, and identifying biomarkers for improved patient stratification. Future advancements in BC treatment will depend on optimizing immunotherapy through biomarker-driven approaches, addressing tumor heterogeneity, and developing innovative combination therapies to overcome resistance. By leveraging these strategies, researchers aim to enhance treatment efficacy and ultimately improve patient outcomes.
Collapse
Affiliation(s)
- Marco Palma
- Institute for Globally Distributed Open Research and Education (IGDORE), 03181 Torrevieja, Spain
| |
Collapse
|
16
|
Camilli M, Maggio L, Tinti L, Torre I, Viscovo M, Viscovo M, Tamburrini G, Lombardo A, Cardinale DM, Minotti G, Rocca B. Cardio-oncology: Emerging Concepts in Cardiovascular Sequelae of Cancer Therapies, Translational Research and Reverse Cardio-oncology. Eur Cardiol 2025; 20:e05. [PMID: 40170756 PMCID: PMC11959581 DOI: 10.15420/ecr.2024.49] [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: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 04/03/2025] Open
Abstract
Cardio-oncology was established with the aim of defining primary and secondary prevention approaches through surveillance and the use of tools to stratify and diminish the cardiovascular risk to cancer patients. This branch of medicine also contributes to establishing a new field in translational medicine for cardiovascular disease by focusing on the interplay between cancer and heart disease. In this first article in the new cardio-oncology section of the journal, we explore the main concepts of emerging anti-cancer therapies and their plausible cardiotoxic effects and we will describe advances and gaps in knowledge, highlighting how cardio-oncology is contributing to translational cardiology. We will speculate on the complex interplay between cancer and heart failure and discuss an emerging concept known as reverse cardio-oncology. We also present the perspective that cardio-oncology represents a promising platform area of research, allowing the discovery of novel pathways involved in cardiovascular disease through the identification of toxicities induced by targeted cancer therapies.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Luca Maggio
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Ilaria Torre
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Giulia Tamburrini
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | | | - Giorgio Minotti
- Unit of Drug Sciences, Fondazione Policlinico Universitario Campus Bio-MedicoRome, Italy
| | - Bianca Rocca
- Department of Medicine and Surgery, Libera Università MediterraneaBari, Italy
- Department of Safety and Bioethics, Università Cattolica del Sacro CuoreRome, Italy
| |
Collapse
|
17
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
18
|
Xu Y, Zhang J, Pan D, Yan J, Chen C, Wang L, Wang X, Yang M, Xu Y. Development of Novel Peptide-Based Radiotracers for Detecting FGL1 Expression in Tumors. Mol Pharm 2025; 22:1605-1614. [PMID: 39893698 DOI: 10.1021/acs.molpharmaceut.4c01293] [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] [Indexed: 02/04/2025]
Abstract
A novel immune checkpoint, FGL1, is a potentially viable target for tumor immunotherapy. The development of FGL1-targeted PET probes could provide significant insights into the immune system's status and the evaluation of treatment efficacy. A ClusPro 2.0 server was used to analyze the interaction between FGL1 and LAG3, and the candidate peptides were identified by using the Rosetta peptide derivate protocol. Three candidate peptides targeting FGL1, named FGLP21, FGLP22, and FGLP23, with a simulated affinity of -9.56, -8.55, and -8.71 kcal/mol, respectively, were identified. The peptides were readily conjugated with p-NCS-benzyl-NODA-GA, and the resulting compounds were successfully labeled with 68Ga in approximately 70% yields and radiochemical purity greater than 95%. In vitro competitive cell-binding assay demonstrated that all probes bound to FGL1 with IC50 ranging from 100 nM to 160 nM. Among the probes, PET imaging revealed that 68Ga-NODA-FGLP21 exhibited the best tumor imaging performance in mice bearing FGL1 positive Huh7 tumor. At 60 min p.i., the tumor uptake of 68Ga-NODA-FGLP21 was significantly higher than those of 68Ga-NODA-FGLP22 and 68Ga-NODA-FGLP23, respectively (2.51 ± 0.11% ID/g vs 1.00 ± 0.16% ID/g and 1.49 ± 0.05% ID/g). Simultaneously, the tumor-to-muscle uptake ratios of the former were also higher than those of the latter, respectively (19.40 ± 2.30 vs 9.65 ± 0.62 and 12.45 ± 0.72). In the presence of unlabeled FGLP21, the uptake of 68Ga-NODA-FGLP21 in Huh7 xenograft decreased to 0.81 ± 0.09% ID/g at 60 min p.i., which is similar to that observed in the FGL1 negative U87 MG tumor (0.46 ± 0.03% ID/g). The results were consistent with the immunohistochemical analysis and ex vivo autoradiography. No significant radioactivity was accumulated in normal organs, except for kidneys. In summary, a preclinical study confirmed that the tracer 68Ga-NODA-FGLP21 has the potential to specifically detect FGL1 expression in tumors with good contrast to the background.
Collapse
Affiliation(s)
- Yue Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jinyuan Zhang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junjie Yan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Chongyang Chen
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xinyu Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Min Yang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuping Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
19
|
De Lucia A, Mazzotti L, Gaimari A, Zurlo M, Maltoni R, Cerchione C, Bravaccini S, Delmonte A, Crinò L, Borges de Souza P, Pasini L, Nicolini F, Bianchi F, Juan M, Calderon H, Magnoni C, Gazzola L, Ulivi P, Mazza M. Non-small cell lung cancer and the tumor microenvironment: making headway from targeted therapies to advanced immunotherapy. Front Immunol 2025; 16:1515748. [PMID: 39995659 PMCID: PMC11847692 DOI: 10.3389/fimmu.2025.1515748] [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: 10/23/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
Over the past decades, significant progress has been made in the understanding of non-small cell lung cancer (NSCLC) biology and tumor progression mechanisms, resulting in the development of novel strategies for early detection and wide-ranging care approaches. Since their introduction, over 20 years ago, targeted therapies with tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for NSCLC. Nowadays, targeted therapies remain the gold standard for many patients, but still they suffer from many adverse effects, including unexpected toxicity and intrinsic acquired resistance mutations, which lead to relapse. The adoption of immune checkpoint inhibitors (ICIs) in 2015, has offered exceptional survival benefits for patients without targetable alterations. Despite this notable progress, challenges remain, as not all patients respond favorably to ICIs, and resistance to therapy can develop over time. A crucial factor influencing clinical response to immunotherapy is the tumor microenvironment (TME). The TME is pivotal in orchestrating the interactions between neoplastic cells and the immune system, influencing tumor growth and treatment outcomes. In this review, we discuss how the understanding of this intricate relationship is crucial for the success of immunotherapy and survey the current state of immunotherapy intervention, with a focus on forthcoming and promising chimeric antigen receptor (CAR) T cell therapies in NSCLC. The TME sets major obstacles for CAR-T therapies, creating conditions that suppress the immune response, inducing T cell exhaustion. To enhance treatment efficacy, specific efforts associated with CAR-T cell therapy in NSCLC, should definitely focus TME-related immunosuppression and antigen escape mechanisms, by combining CAR-T cells with immune checkpoint blockades.
Collapse
Affiliation(s)
- Anna De Lucia
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucia Mazzotti
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Gaimari
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Matteo Zurlo
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Roberta Maltoni
- Healthcare Administration, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Claudio Cerchione
- Hematology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Sara Bravaccini
- Department of Medicine and Surgery, “Kore” University of Enna, Enna, Italy
| | - Angelo Delmonte
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Lucio Crinò
- Medical Oncology Department, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Patricia Borges de Souza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Luigi Pasini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabio Nicolini
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Fabrizio Bianchi
- Unit of Cancer Biomarker, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Manel Juan
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Hugo Calderon
- Department of Immunology, Institut D’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Chiara Magnoni
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Luca Gazzola
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Paola Ulivi
- Translational Oncology Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| | - Massimiliano Mazza
- Advanced Cellular Therapies and Rare Tumors Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy
| |
Collapse
|
20
|
Crespo E, Loureiro LR, Stammberger A, Hoffmann L, Berndt N, Hoffmann A, Dagostino C, Soto KEG, Rupp L, Arndt C, Schneider M, Ball CR, Bachmann M, Schmitz M, Feldmann A. RevCAR-mediated T-cell response against PD-L1-expressing cells turns suppression into activation. NPJ Precis Oncol 2025; 9:42. [PMID: 39924591 PMCID: PMC11808103 DOI: 10.1038/s41698-025-00828-6] [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: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
Applying CAR T-cell therapy to treat solid tumors is especially challenging due to the immunosuppressive tumor microenvironment (TME). While our modular RevCAR system enhances the safety and controllability of CAR T-cell therapy, effectively targeting solid tumors remains difficult. Since PD-L1 is an immune checkpoint frequently upregulated by cancer cells and their microenvironment, it is a relevant target for solid tumors. Here, we introduce a novel PD-L1 RevTM capable of redirecting RevCAR T-cells to specifically target and kill PD-L1-expressing tumor cells, becoming activated and secreting pro-inflammatory cytokines. This is shown in vitro with monolayer and 3D models, including patient-derived cultures, and in vivo. Furthermore, we demonstrate in vitro and in vivo an AND-gated targeting of cells simultaneously expressing PD-L1 and another tumor-associated antigen by the Dual RevCAR system. Our findings suggest that RevCAR-mediated targeting of PD-L1 could be a promising therapeutic approach for modulating the TME and improving solid tumor treatment.
Collapse
Affiliation(s)
- Eugenia Crespo
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Liliana R Loureiro
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Antonia Stammberger
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Lydia Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Nicole Berndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Anja Hoffmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Claudia Dagostino
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Karla E G Soto
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Claudia Arndt
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Mildred Scheel Early Career Center, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Martin Schneider
- Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Claudia R Ball
- Department for Translational Medical Oncology, National Center for Tumor Diseases Dresden (NCT/UCC), a partnership between DKFZ, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, and Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Translational Medical Oncology, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Faculty of Biology, TUD Dresden University of Technology, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany.
| | - Marc Schmitz
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany
| | - Anja Feldmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany.
- German Cancer Consortium (DKTK), partner site Dresden, Dresden, Germany.
- National Center for Tumor Diseases (NCT), partner site Dresden, Dresden, Germany.
| |
Collapse
|
21
|
Alnajjar NF, Tahan SR, Iriarte C. Immunotherapy-associated bullous and oral erosive lichenoid eruption successfully treated with hydroxychloroquine. JAAD Case Rep 2025; 56:63-66. [PMID: 39886602 PMCID: PMC11779660 DOI: 10.1016/j.jdcr.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2025] Open
Affiliation(s)
- Nadean F. Alnajjar
- Harvard Medical School, Boston, Massachusetts
- Department of Dermatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Steven R. Tahan
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - Christopher Iriarte
- Department of Dermatology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
- Department of Dermatology, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
22
|
Walmsley CS, Schoepflin Z, De Brabandt C, Rangachari D, Berwick S, Patell R. Hemophagocytic lymphohistiocytosis associated with immune checkpoint inhibitor use: A review of the current knowledge and future directions. Blood Cells Mol Dis 2025; 110:102896. [PMID: 39366077 DOI: 10.1016/j.bcmd.2024.102896] [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: 08/10/2024] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/06/2024]
Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a severe and often lethal inflammatory syndrome characterized by excessive immune activation leading to fever, cytopenias, and multiorgan involvement. Immune checkpoint inhibitors (ICIs) are central to many contemporary cancer regimens, but their use is associated with immune-related adverse events. Here, we report a case of ICI-induced HLH successfully treated with single agent dexamethasone and provide a scoping review of the literature for cases of ICI-induced HLH with a focus on treatment strategies and outcomes. Using the Medline database, we searched for cases of ICI-associated HLH, with a total of 51 cases reported between 2017 and 2023. Our results underscore the severe nature of this disease, with a 13.7 % mortality rate across 51 case reports. Treatment strategies for ICI-induced HLH were variable: steroids alone (56.9 %), steroids with etoposide (17.6 %), steroids with tociluzumab (11.8 %), among other combinations. Our literature review indicates that steroids alone may be sufficient treatment in some cases of ICI-HLH, with comparable mortality with steroids alone (n = 29) (13.8 %) to that of cases treated with both steroids and immunomodulators (n = 15, 13.3 %). Moreover, all patients treated with steroids and tocilizumab survived (n = 6), suggesting that tocilizumab may be a reasonable next line of therapy when steroid monotherapy proves inadequate. We propose an outline for investigation and treatment of this rare complication of ICI use. Finally, we discuss possible future approaches to develop evidence-based strategies for the diagnosis and management of ICI-induced HLH including the importance of integrating the role of patient community involvement.
Collapse
Affiliation(s)
- Charlotte S Walmsley
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States of America
| | - Zachary Schoepflin
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Charlotte De Brabandt
- Breast Cancer Center, Beth Israel Deaconess Medical Center, Boston, MA, United States of America; Patient Ambassador Program, American Cancer Society, Boston, MA, United States of America
| | - Deepa Rangachari
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Shana Berwick
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America
| | - Rushad Patell
- Division of Hematology/Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, United States of America.
| |
Collapse
|
23
|
Park SJ, Yang S, Lee S, Joo SH, Park T, Kim DH, Kim H, Park S, Kim JT, Kwack WG, Kang SW, Song YK, Cha JM, Rhee SY, Chung EK. Machine-Learning Parsimonious Prediction Model for Diagnostic Screening of Severe Hematological Adverse Events in Cancer Patients Treated with PD-1/PD-L1 Inhibitors: Retrospective Observational Study by Using the Common Data Model. Diagnostics (Basel) 2025; 15:226. [PMID: 39857110 PMCID: PMC11763827 DOI: 10.3390/diagnostics15020226] [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: 10/04/2024] [Revised: 01/08/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: Earlier detection of severe immune-related hematological adverse events (irHAEs) in cancer patients treated with a PD-1 or PD-L1 inhibitor is critical to improving treatment outcomes. The study aimed to develop a simple machine learning (ML) model for predicting irHAEs associated with PD-1/PD-L1 inhibitors. Methods: We utilized the Observational Medical Outcomes Partnership-Common Data Model based on electronic medical records from a tertiary (KHMC) and a secondary (KHNMC) hospital in South Korea. Severe irHAEs were defined as Grades 3-5 by the Common Terminology Criteria for Adverse Events (version 5.0). The predictive model was developed using the KHMC dataset, and then cross-validated against an independent cohort (KHNMC). The full ML models were then simplified by selecting critical features based on the feature importance values (FIVs). Results: Overall, 397 and 255 patients were included in the primary (KHMC) and cross-validation (KHNMC) cohort, respectively. Among the tested ML algorithms, random forest achieved the highest accuracy (area under the receiver operating characteristic curve [AUROC] 0.88 for both cohorts). Parsimonious models reduced to 50% FIVs of the full models showed comparable performance to the full models (AUROC 0.83-0.86, p > 0.05). The KHMC and KHNMC parsimonious models shared common predictive features including furosemide, oxygen gas, piperacillin/tazobactam, and acetylcysteine. Conclusions: Considering the simplicity and adequate predictive performance, our simplified ML models might be easily implemented in clinical practice with broad applicability. Our model might enhance early diagnostic screening of irHAEs induced by PD-1/PD-L1 inhibitors, contributing to minimizing the risk of severe irHAEs and improving the effectiveness of cancer immunotherapy.
Collapse
Affiliation(s)
- Seok Jun Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Seungwon Yang
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Suhyun Lee
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Sung Hwan Joo
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
| | - Taemin Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Dong Hyun Kim
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Hyeonji Kim
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Soyun Park
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Jung-Tae Kim
- Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Won Gun Kwack
- Division of Pulmonary, Allergy and Critical Care Medicine, Kyung Hee University Hospital, Seoul 02447, Republic of Korea;
| | - Sung Wook Kang
- Department of Pulmonary and Critical Care Medicine, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| | - Yun-Kyoung Song
- College of Pharmacy, The Catholic University of Korea-Sungsim Campus, Bucheon 14662, Gyeonggi-do, Republic of Korea;
| | - Jae Myung Cha
- Division of Gastroenterology, Department of Internal Medicine, Kyung Hee University Hospital at Gangdong, Kyung Hee University School of Medicine, Seoul 05278, Republic of Korea
| | - Sang Youl Rhee
- Center for Digital Health, Medical Science Research Institute, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Endocrinology and Metabolism, Kyung Hee University School of Medicine, Seoul 02447, Republic of Korea
| | - Eun Kyoung Chung
- Department of Regulatory Science, College of Pharmacy, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea; (S.J.P.); (S.Y.); (S.H.J.); (T.P.); (D.H.K.); (H.K.); (S.P.)
- Institute of Regulatory Innovation Through Science (IRIS), Kyung Hee University, Seoul 02447, Republic of Korea
- Department of Pharmacy, College of Pharmacy, Kyung Hee University, Seoul 02447, Republic of Korea;
- Department of Pharmacy, Kyung Hee University Hospital at Gangdong, Seoul 05278, Republic of Korea;
| |
Collapse
|
24
|
Lindsay AC, Walker AM, Schneeweiss S. Myocarditis in Patients Starting Combination Checkpoint Inhibitor Therapy: Analysis of a Commercial Claims Database. J Am Heart Assoc 2025; 14:e035689. [PMID: 39719418 PMCID: PMC12054407 DOI: 10.1161/jaha.124.035689] [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: 04/08/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors have improved the clinical outcomes of several cancers but have also been associated with a greater risk of immune-related adverse effects, especially when combined. The objective of this study was to investigate the incidence of myocarditis in relation to the use of dual concurrent versus single immune checkpoint inhibitors therapies. METHODS AND RESULTS A cohort study was conducted using medical and pharmacy claims data (2011-2022) from a large US commercial insurer. Cox regression quantified the comparative risks of myocarditis or heart failure in patients with cancer receiving treatment with combination therapy (nivolumab and ipilimumab) in comparison to taking a single immune checkpoint inhibitor only. Mean follow-up time in 53 018 patients was 226 days (interquartile range, 93-495 days). There were 148 cases of myocarditis (0.3%), 33 (0.7%) in patients on combination therapy, and 115 (0.2%) in patients on monotherapy. The risk of myocarditis per 1000 patients was 7.40 in the combination therapy group and 2.37 in the monotherapy group (risk ratio, 3.12 [95% CI, 2.12-4.60]). Using multivariable regression analysis, the hazard ratio for myocarditis in the combination therapy group was 2.38 (1.57-3.63). No difference in the risk of heart failure was found between combination and single therapy. CONCLUSIONS Therapy with 2 immune checkpoint inhibitors was associated with an increased risk of myocarditis compared with monotherapy, with most cases occurring in the first 6 months of therapy.
Collapse
Affiliation(s)
- Alistair C. Lindsay
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| | - Alexander M. Walker
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMA
| | - Sebastian Schneeweiss
- Division of Pharmacoepidemiology and Pharmacoeconomics, Department of MedicineBrigham and Women’s Hospital, Harvard Medical SchoolBostonMA
| |
Collapse
|
25
|
Du Y, Zhang H, Liu J, Duan X, Chen S, Jiang W. HK3: A potential prognostic biomarker with metastasis inhibition capabilities in hepatocellular carcinoma. Biochem Biophys Res Commun 2024; 741:151057. [PMID: 39615209 DOI: 10.1016/j.bbrc.2024.151057] [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: 09/11/2024] [Revised: 11/06/2024] [Accepted: 11/22/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) stands as one of the prevalent malignant tumors worldwide. The effectiveness of immunotherapy frequently depends on the intricate dynamics of immunomodulation within the tumor microenvironment (TME). The current study aims to identify prognostically relevant genes and their functional roles in HCC. This is achieved by utilizing immune scores and mutations as the basis, through the application of bioinformatics and molecular biological analysis. METHODS Differentially expressed genes (DEGs) analysis was conducted using the "clusterProfiler" package for functional enrichment. Cox regression analysis and LASSO regression analysis were performed for prognostic gene screening. Kaplan-Meier curve were further utilized to verify the prognostic value of these genes. The relationship between selected genes and immune cells was analyzed using ssGSEA algorithm and TIMER. The HK3 expression in HCC cells was tested by Western blot. Additionally, wound healing and transwell assays were utilized to detect the impact of HK3 on HCC metastasis. RESULTS Patients who had higher ESTIMATE, stromal, and immune scores exhibited more favorable overall survival rates. There are 17 genes that overlap among the DEGs related to the immune-stromal-ESTIMATE scores, mutated genes, and DEGs in HCC tissues compared to normal tissues. Among the DEGs, three genes (STAB1, COL15A1 and HK3) emerged with the most profound association concerning survival outcomes. Notably, the HK3 genes displayed a pronounced correlation with immune infiltration. Concurrently, diminished expression levels of HK3 were observed in HCC tissues and upregulation of HK3 resulted in a significant reduction in HCC cell metastasis in vitro and in vivo. CONCLUSIONS HK3 emerges as a novel prognostic biomarker for HCC, exerting regulatory influence over cellular proliferation, metastasis, and invasiveness. These findings indicate that HK3 holds promise as a potential candidate for treatment and prognosis of HCC.
Collapse
Affiliation(s)
- Yexiang Du
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Hongchuan Zhang
- Department of Oncology, Dianjiang People's Hospital of Chongqing, Chongqing, 408300, China
| | - Jialong Liu
- 65136, Troops Hospital of PLA, Dalian, Liaoning, 116300, China
| | - Xiaodong Duan
- Department of Anatomy, Basic Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Suhua Chen
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guizhou, 550002, China
| | - Wenbin Jiang
- Department of Clinical Laboratory, Guizhou Provincial People's Hospital, Guizhou, 550002, China.
| |
Collapse
|
26
|
Jung W, Cho IY, Jung J, Cho MH, Koo HY, Park YMM, Han K, Shin DW. Changes in Physical Activity and Cardiovascular Disease Risk in Cancer Survivors: A Nationwide Cohort Study. JACC CardioOncol 2024; 6:879-889. [PMID: 39801643 PMCID: PMC11711819 DOI: 10.1016/j.jaccao.2024.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 09/11/2024] [Accepted: 09/23/2024] [Indexed: 01/16/2025] Open
Abstract
Background Cancer survivors face an elevated risk of cardiovascular disease, with physical inactivity after cancer treatment potentially worsening this risk. Objectives The aim of this study was to investigate the association between physical activity before and after a cancer diagnosis and the risk for heart disease. Methods A nationwide cohort of 269,943 cancer survivors (mean age 56.3, 45.7% men) was evaluated for physical activity adherence 2 years before and after diagnosis. The primary outcomes were the incidence of myocardial infarction (MI), heart failure (HF), and atrial fibrillation. Subdistribution HRs (sHRs) and 95% CIs were calculated using Gray's method, accounting for death as a competing risk. Results Over a follow-up period of 1,111,329.28 person-years, compared with those who remained inactive, persistent physical activity was associated with a 20% reduction in MI risk (sHR: 0.80; 95% CI: 0.70-0.91) and a 16% reduction risk in HF risk (sHR: 0.84; 95% CI: 0.78-0.90). Initiating physical activity after a cancer diagnosis was linked to an 11% lower risk for MI (sHR: 0.89; 95% CI: 0.79-0.99) and a 13% lower risk for HF (sHR: 0.87; 95% CI: 0.82-0.93). Being active only before diagnosis was associated with a 20% lower risk for MI (sHR: 0.80; 95% CI: 0.71-0.91) and a 6% lower risk for HF (sHR: 0.94; 95% CI: 0.88-1.00). No association was observed between physical activity and atrial fibrillation risk. Associations varied by primary cancer site. Conclusions These findings underscore the importance of maintaining physical activity for cardiovascular health in cancer survivors and suggest that physical activity before a diagnosis may offer enduring protection against ischemic heart disease and cardiac dysfunction.
Collapse
Affiliation(s)
- Wonyoung Jung
- Department of Family Medicine/Obesity and Metabolic Health Center, Kangdong Sacred Heart Hospital, Hallym University, Seoul, Republic of Korea
- Department of Medicine, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - In Young Cho
- Department of Family Medicine and Supportive Care Center, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jinhyung Jung
- Department of Biostatistics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Mi Hee Cho
- Samsung C&T Medical Clinic, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hye Yeon Koo
- Department of Family Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Yong-Moon Mark Park
- Department of Epidemiology, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kyungdo Han
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, Republic of Korea
| | - Dong Wook Shin
- Department of Clinical Research Design and Evaluation, Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Seoul, Republic of Korea
- Center for Trend Sensing-Risk Modeling, Institute of Quality of Life in Cancer, Samsung Medical Center, Seoul, Republic of Korea
| |
Collapse
|
27
|
Hoshi K, Hashim Y, Togo S, Saiwaki S, Motomura H, Sumiyoshi I, Nakazawa S, Ochi Y, Miyoshi C, Heo R, Tabe Y, Abe K, Urata Y, Takahashi K. Design of buffer property for the new enrichment method of circulating tumor cell based on immunomagnetic-negative separation. Comput Struct Biotechnol J 2024; 25:281-289. [PMID: 39720309 PMCID: PMC11667567 DOI: 10.1016/j.csbj.2024.11.033] [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: 08/14/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 12/26/2024] Open
Abstract
Metastasis is a significant contributor to cancer-related mortality and a critical issue in cancer. Monitoring the changes in circulating tumor cells (CTCs) with metastatic potential is a valuable prognostic and predictive biomarker. CTCs are a rare population in the peripheral blood of patients with cancer. The enrichment process is extremely important for the isolation of clinically significant CTC subpopulations, which can then be used for further analysis. The present study postulates that the buffer serves as an essential field for immunomagnetic separation, thereby enhancing the efficacy of CTC enrichment in peripheral blood. This, in turn, facilitates CTC detection. Here, we describe the design of buffers for developing a novel immunomagnetic-negative separation method for CTC enrichment. During the design process, the buffer properties of the floating and cell coatings had a synergistic effect on the efficiency of cell enrichment in blood samples. The efficacy of the method was evaluated using peripheral blood samples from patients with non-small cell lung cancer (NSCLC) and small cell lung cancer (SCLC). The developed method enriched clinically relevant CTC subpopulations that expressed the epithelial-mesenchymal transition (EMT)-related molecule vimentin and/or the cancer immune checkpoint marker programmed death ligand 1 (PD-L1). Furthermore, it was applicable as a part of the enrichment process in a TelomeScan® (OBP-401)-based CTC detection assay with high sensitivity and specificity. From the perspective of methodological approaches, the design of buffer properties can be useful for developing a highly versatile enrichment method for handling CTC heterogeneity.
Collapse
Affiliation(s)
- Kazuaki Hoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yasinjan Hashim
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Leading Center for the Development and Research of Cancer Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shinsaku Togo
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shoko Saiwaki
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Hiroaki Motomura
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Issei Sumiyoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Shun Nakazawa
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yusuke Ochi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Chieko Miyoshi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Rihyang Heo
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Yoko Tabe
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Clinical Laboratory Medicine, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| | - Kanae Abe
- Oncolys BioPharma, Inc., 4-1-28 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| | - Yasuo Urata
- Oncolys BioPharma, Inc., 4-1-28 Toranomon, Minato-ku, Tokyo 105-0001, Japan
| | - Kazuhisa Takahashi
- Division of Respiratory Medicine, Juntendo University Faculty of Medicine & Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Department of Minimally Invasive Next-Generation Cancer Diagnosis by TelomeScan, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
| |
Collapse
|
28
|
Villatore A, Bosi C, Pomaranzi C, Cigliola A, Tateo V, Mercinelli C, Vignale D, Rizzo S, Necchi A, Peretto G. Myocarditis Following Pembrolizumab Plus Axitinib, and Belzutifan Plus Lenvatinib for Renal Cell Carcinoma: A Case Report. Cardiovasc Toxicol 2024; 24:1168-1173. [PMID: 39085529 DOI: 10.1007/s12012-024-09906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Cardiac toxicity is an adverse event of several classes of anti-cancer drugs. Herein, we present the case of a 52-year-old woman with metastatic renal cell carcinoma (RCC), previously treated with debulking surgery, pembrolizumab (immune checkpoint inhibitor) in combination with axitinib (tyrosine kinase inhibitor (TKI)), followed by lenvatinib (TKI) and belzutifan (HIF-2α inhibitor), who developed myocarditis proven by cardiac magnetic resonance and endomyocardial biopsy. The case was notable for reporting a not-yet described adverse event during treatment with belzutifan plus lenvatinib, the etiology of which was of unobvious determination given the pre-exposure to pembrolizumab, a known cause of drug-related myocarditis. We surmise that myocarditis was a delayed adverse event related to pembrolizumab (8 months after treatment interruption), although we emphasize that only attentive monitoring of cardiac adverse events of patients exposed to belzutifan and lenvatinib in the context of large clinical trials may rule out any causal implication of these drugs.
Collapse
Affiliation(s)
- Andrea Villatore
- Vita-Salute San Raffaele University, Milan, Italy.
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy.
| | - Carlo Bosi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Antonio Cigliola
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valentina Tateo
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Mercinelli
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Davide Vignale
- Experimental Imaging Center, Radiology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Stefania Rizzo
- Cardiovascular Pathology Unit, Azienda Ospedaliera Di Padova, Padua, Italy
| | - Andrea Necchi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giovanni Peretto
- Vita-Salute San Raffaele University, Milan, Italy
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
29
|
Oyakawa T, Muraoka N, Iida K, Fujita A, Yokoyama K, Ishikawa H, Murakami H. Relevance of surveillance manual for the early detection of immune checkpoint inhibitor-induced myocarditis: A case series. Asia Pac J Oncol Nurs 2024; 11:100598. [PMID: 39582548 PMCID: PMC11582373 DOI: 10.1016/j.apjon.2024.100598] [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: 07/30/2024] [Accepted: 09/18/2024] [Indexed: 11/26/2024] Open
Abstract
Objective The European Cardio-Oncology Guidelines recommend regular electrocardiography and troponin testing during immune checkpoint inhibitors (ICIs) treatment, but their efficacy for monitoring ICI treatment remains unclear. This study aimed to evaluate the effectiveness of a surveillance protocol for early detection of ICI-induced myocarditis. Methods Between May 2014 and May 2024, patients who began treatment with ICIs at our hospital and developed ICI-induced myocarditis were included in this study. We created a straightforward management flowchart for myocarditis. The protocol was based on monitoring troponin T levels. We confirmed the efficacy of our surveillance protocol using a case series of ICI-induced myocarditis. Results During the observation period, 3481 patients were newly started on ICIs at our hospital. Five patients were previously diagnosed with myocarditis, and five patients were diagnosed with myocarditis after the implementation of the surveillance protocol. The manual enabled the early detection of myocarditis, and the mortality rate for myocarditis at our hospital improved from 60% to 0%. The incidence of conduction system disorders significantly reduced from 100% to 0% (P < 0.01). After the surveillance protocol was initiated, there were no cases of myocarditis requiring immunosuppressive drugs beyond steroids. Conclusions This study confirmed the relevance of a troponin-based surveillance protocol for the early detection of ICI-induced myocarditis. The implementation of the surveillance protocol reduced mortality from myocarditis and significantly reduced serious complications of conduction system disorders. Although this study is a small case series of patients who developed myocarditis, we confirm the effectiveness of surveillance for myocarditis.
Collapse
Affiliation(s)
- Takuya Oyakawa
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Nao Muraoka
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kei Iida
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
- Mishimatoukai Hospital, Shizuoka, Japan
| | - Ayano Fujita
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | | | - Haruyasu Murakami
- Division of Advanced Medical Development, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
30
|
Kalinoski H, Daoud A, Rusinkevich V, Jurčová I, Talor MV, Welsh RA, Hughes D, Zemanová K, Stříž I, Hooper JE, Kautzner J, Peichl P, Melenovský V, Won T, Čiháková D. Injury-induced myosin-specific tissue-resident memory T cells drive immune checkpoint inhibitor myocarditis. Proc Natl Acad Sci U S A 2024; 121:e2323052121. [PMID: 39378095 PMCID: PMC11494310 DOI: 10.1073/pnas.2323052121] [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: 01/21/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Cardiac myosin-specific (MyHC) T cells drive the disease pathogenesis of immune checkpoint inhibitor-associated myocarditis (ICI-myocarditis). To determine whether MyHC T cells are tissue-resident memory T (TRM) cells, we characterized cardiac TRM cells in naive mice and established that they have a distinct phenotypic and transcriptional profile that can be defined by their upregulation of CD69, PD-1, and CXCR6. We then investigated the effects of cardiac injury through a modified experimental autoimmune myocarditis mouse model and an ischemia-reperfusion injury mouse model and determined that cardiac inflammation induces the recruitment of autoreactive MyHC TRM cells, which coexpress PD-1 and CD69. To investigate whether the recruited MyHC TRM cells could increase susceptibility to ICI-myocarditis, we developed a two-hit ICI-myocarditis mouse model where cardiac injury was induced, mice were allowed to recover, and then were treated with anti-PD-1 antibodies. We determined that mice who recover from cardiac injury are more susceptible to ICI-myocarditis development. We found that murine and human TRM cells share a similar location in the heart and aggregate along the perimyocardium. We phenotyped cells obtained from pericardial fluid from patients diagnosed with dilated cardiomyopathy and ischemic cardiomyopathy and established that pericardial T cells are predominantly CD69+ TRM cells that up-regulate PD-1. Finally, we determined that human pericardial macrophages produce IL-15, which supports and maintains pericardial TRM cells.
Collapse
Affiliation(s)
- Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Vitali Rusinkevich
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Ivana Jurčová
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Monica V. Talor
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Robin A. Welsh
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - David Hughes
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD21205
| | - Kateřina Zemanová
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Ilja Stříž
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Jody E. Hooper
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Taejoon Won
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| |
Collapse
|
31
|
Barac A, Wadlow RC, Deeken JF, deFilippi C. Cardiac Troponin I and T in ICI Myocarditis Screening, Diagnosis, and Prognosis. JACC CardioOncol 2024; 6:804-807. [PMID: 39479330 PMCID: PMC11520212 DOI: 10.1016/j.jaccao.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Ana Barac
- Inova Schar Heart and Vascular, Fairfax, Virginia, USA
- Inova Schar Cancer, Fairfax, Virginia, USA
| | | | | | | |
Collapse
|
32
|
Srinivasan S, Armitage J, Nilsson J, Waithman J. Transcriptional rewiring in CD8 + T cells: implications for CAR-T cell therapy against solid tumours. Front Immunol 2024; 15:1412731. [PMID: 39399500 PMCID: PMC11466849 DOI: 10.3389/fimmu.2024.1412731] [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: 04/05/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024] Open
Abstract
T cells engineered to express chimeric-antigen receptors (CAR-T cells) can effectively control relapsed and refractory haematological malignancies in the clinic. However, the successes of CAR-T cell therapy have not been recapitulated in solid tumours due to a range of barriers such as immunosuppression, poor infiltration, and tumour heterogeneity. Numerous strategies are being developed to overcome these barriers, which include improving culture conditions and manufacturing protocols, implementing novel CAR designs, and novel approaches to engineering the T cell phenotype. In this review, we describe the various emerging strategies to improve CAR T cell therapy for solid tumours. We specifically focus on new strategies to modulate cell function and fate that have precipitated from the growing knowledge of transcriptional circuits driving T cell differentiation, with the ultimate goal of driving more productive anti-tumour T cell immunity. Evidence shows that enrichment of particular phenotypic subsets of T cells in the initial cell product correlates to improved therapeutic responses and clinical outcomes. Furthermore, T cell exhaustion and poor persistence are major factors limiting therapeutic efficacy. The latest preclinical work shows that targeting specific master regulators and transcription factors can overcome these key barriers, resulting in superior T cell therapeutic products. This can be achieved by targeting key transcriptional circuits promoting memory-like phenotypes or sustaining key effector functions within the hostile tumour microenvironment. Additional discussion points include emerging considerations for the field such as (i) targeting permutations of transcription factors, (ii) transient expression systems, (iii) tissue specificity, and (iv) expanding this strategy beyond CAR-T cell therapy and cancer.
Collapse
Affiliation(s)
- Shamini Srinivasan
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
| | - Jesse Armitage
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| | - Jonas Nilsson
- Melanoma Discovery Lab, Harry Perkins Institute of Medical Research, Centre of Medical Research, The University of Western Australia, Perth, WA, Australia
- Sahlgrenska Center for Cancer Research, Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Jason Waithman
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia
- Telethon Kids Cancer Centre, Telethon Kids Institute, Perth, WA, Australia
| |
Collapse
|
33
|
Chiang CH, Xu XH, Song J, Xanthavanij N, Chi KY, Chang YC, Chang Y, Hsiao CL, Hsia YP, Chiang CH, Lin S. The incidence and risk of cardiovascular events associated with pembrolizumab in patients with breast cancer. Cancer Lett 2024; 611:217277. [PMID: 39332590 DOI: 10.1016/j.canlet.2024.217277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 09/21/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Affiliation(s)
- Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA.
| | - Xiaocao Haze Xu
- Division of Hematology and Oncology, Department of Medicine, University of Vermont Medical Center, Burlington, VT, USA
| | - Junmin Song
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nutchapon Xanthavanij
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Cambridge, MA, USA
| | - Kuan-Yu Chi
- Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yu-Cheng Chang
- Department of Medicine, Danbury Hospital, Danbury, CT, USA
| | - Yu Chang
- Section of Neurosurgery, Department of Surgery, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chieh-Lien Hsiao
- School of International and Public Affairs, Columbia University, New York, NY, USA
| | - Yuan Ping Hsia
- Department of Family Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan.
| | - Cho-Hung Chiang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan.
| | - Shuwen Lin
- Department of Oncology, Montefiore Medical Center, Montefiore Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
34
|
Xie W, Liu H, Lin Q, Lian L, Liang B. Association of non-high-density lipoprotein to high-density lipoprotein ratio (NHHR) with prognosis in cancer survivors: a population-based study in the United States. Front Nutr 2024; 11:1430835. [PMID: 39296499 PMCID: PMC11409846 DOI: 10.3389/fnut.2024.1430835] [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: 05/10/2024] [Accepted: 08/08/2024] [Indexed: 09/21/2024] Open
Abstract
Background Patients with cancer frequently exhibit alterations in serum lipid profiles associated with chemotherapy. It has been reported that lipid distribution in cancer correlates with tumor progression. However, the prognostic value of serum lipid biomarkers in cancer survivors remains a subject of debate. We aim to explore the relationship between non-high-density lipoprotein to high-density lipoprotein ratio (NHHR) and the prognosis of cancer survivors. Methods In this study, we analyzed cancer survivor data from the National Health and Nutrition Examination Survey (NHANES) from 1999-2000 to 2017-2018. The study included prospective cohorts that included total cholesterol (TC) and high-density lipoprotein cholesterol (HDL-C) levels as well as mortality data. Weighted multivariate cox regression models, competing risk models and restricted cubic spline (RCS) models were applied to investigate the association between NHHR and cancer survival. Subgroup and sensitivity analyses were performed to test the robustness of the results. Results This study involved 4,177 participants, representing about 19.6 million U.S. adults. After adjustment for various factors, the lower NHHR group (≤1.64) had a 31% (HR 1.31; 95% CI [1.11,1.54], p = 0.001) higher risk of death from any cause compared to the higher NHHR group. The link between NHHR and mortality remained stable across most subgroups, with notable interactions for smoking (p = 0.006) and diabetes status (p = 0.046). A J-shaped pattern was observed between NHHR and all-cause mortality, significantly among obesity-related cancer survivors (overall association test p-value = 0.0068, non-linear association test p-value = 0.0016). However, a non-significant negative correlation was observed for cancer-specific mortality (overall association test p-value = 0.48, non-linear association test p-value = 0.66). Considering the competitive risk of heart disease and cancer-specific mortality, there is no difference between the high and low NHHR groups, while the low NHHR group showed an increased risk of non-specific causes of death (p < 0.001). Conclusion The results of this study suggest that NHHR is an important indicator that is strongly associated with all-cause mortality in cancer survivors, and that this relationship may be influenced by the interaction of diabetes and smoking status. This finding may provide important information for future research and patient management.
Collapse
Affiliation(s)
- Wenxia Xie
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huizhuo Liu
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiaoxin Lin
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liyou Lian
- Department of Internal Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Bin Liang
- Department of Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
35
|
Rus Bakarurraini NAA, Kamarudin AA, Jamal R, Abu N. Engineered T cells for Colorectal Cancer. Immunotherapy 2024; 16:987-998. [PMID: 39229803 PMCID: PMC11485792 DOI: 10.1080/1750743x.2024.2391733] [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: 07/25/2023] [Accepted: 08/06/2024] [Indexed: 09/05/2024] Open
Abstract
Colorectal cancer (CRC) is a major contributor to global cancer incidence and mortality. Conventional treatments have limitations; hence, innovative approaches are imperative. Recent advancements in cancer research have led to the development of personalized targeted therapies and immunotherapies. Immunotherapy, in particular, T cell-based therapies, exhibited to be promising in enhancing cancer treatment outcomes. This review focuses on the landscape of engineered T cells as a potential option for the treatment of CRC. It highlights the approaches, challenges and current advancements in this field. As the understanding of molecular mechanisms increases, engineered T cells hold great potential in revolutionizing cancer treatment. To fully explore their safety efficacy in improving patient outcomes, further research and clinical trials are necessary.
Collapse
Affiliation(s)
| | - Ammar Akram Kamarudin
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Rahman Jamal
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| | - Nadiah Abu
- UKM Medical Molecular Biology Institute (UMBI), Universiti Kebangsaan Malaysia (UKM), Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Wilcox NS, Amit U, Reibel JB, Berlin E, Howell K, Ky B. Cardiovascular disease and cancer: shared risk factors and mechanisms. Nat Rev Cardiol 2024; 21:617-631. [PMID: 38600368 PMCID: PMC11324377 DOI: 10.1038/s41569-024-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Cardiovascular disease (CVD) and cancer are among the leading causes of morbidity and mortality globally, and these conditions are increasingly recognized to be fundamentally interconnected. In this Review, we present the current epidemiological data for each of the modifiable risk factors shared by the two diseases, including hypertension, hyperlipidaemia, diabetes mellitus, obesity, smoking, diet, physical activity and the social determinants of health. We then review the epidemiological data demonstrating the increased risk of CVD in patients with cancer, as well as the increased risk of cancer in patients with CVD. We also discuss the shared mechanisms implicated in the development of these conditions, highlighting their inherent bidirectional relationship. We conclude with a perspective on future research directions for the field of cardio-oncology to advance the care of patients with CVD and cancer.
Collapse
Affiliation(s)
- Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob B Reibel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eva Berlin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendyl Howell
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
37
|
Nobin H, Garvin S, Hagman H, Nodin B, Jirström K, Brunnström H. The prognostic value of programmed death-ligand 1 (PD-L1) expression in resected colorectal cancer without neoadjuvant therapy - differences between antibody clones and cell types. BMC Cancer 2024; 24:1051. [PMID: 39187798 PMCID: PMC11346183 DOI: 10.1186/s12885-024-12812-7] [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: 06/14/2024] [Accepted: 08/14/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND Programmed death-ligand 1 (PD-L1) expression on tumor cells is associated with poor prognosis in several malignancies, while partly contradictory and inconclusive results have been presented for colorectal cancer (CRC). This study aimed to evaluate PD-L1 as a prognostic biomarker in CRC by comparing three different antibody clones. METHODS Patients surgically treated for CRC between January 1st, 2007, and December 31st, 2015, in Kalmar County, Sweden, were retrospectively included. Tissue microarrays from 862 primary tumors without neoadjuvant treatment were assessed for immunohistochemical expression of PD-L1 in tumor cells (TC) and immune cells (IC) using clones 73-10, SP263, and 22C3. Cox regression proportional hazard models were used to estimate hazard ratios for overall survival (OS) and disease-free interval (DFI) in univariable and multivariable analyses, with 1% and 5% set as cut-offs for positive expression in TC and IC respectively. RESULTS PD-L1 expression in TC was found in 89 (10%) cases for clone 73-10, 76 (9%) for clone SP263, and 38 (4%) for clone 22C3, while the numbers for IC were 317 (37%) cases for clone 73-10, 264 (31%) for clone SP263, and 89 (10%) for clone 22C3. PD-L1 expression in IC was associated with prolonged OS and DFI in univariable analysis for all three clones. The link to prolonged DFI remained in multivariable analysis for 73-10 and SP263, but only for 73-10 regarding OS. PD-L1 expression in TC was not prognostic of OS in any analysis, while it was associated with prolonged DFI for SP263, and a trend was seen for 73-10. The link to prolonged DFI remained for SP263 and was strengthened for 73-10 in multivariable analysis. CONCLUSIONS The prognostic value of PD-L1 expression in both IC and TC differs between antibody clones, with 73-10 and SP263 being more reliable for prognostic information than 22C3 in resected CRC.
Collapse
Affiliation(s)
- Hampus Nobin
- Department of Pathology, Region Kalmar, Kalmar County Hospital, Kalmar, Sweden.
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, Lund, Sweden.
| | - Stina Garvin
- Department of Clinical Pathology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Helga Hagman
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Björn Nodin
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
| | - Karin Jirström
- Department of Clinical Sciences Lund, Division of Oncology and Therapeutic Pathology, Lund University, Lund, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Regional University Laboratories, Skåne University Hospital, Lund, Sweden
| | - Hans Brunnström
- Department of Clinical Sciences Lund, Division of Pathology, Lund University, Lund, Sweden
- Department of Genetics, Pathology, and Molecular Diagnostics, Regional University Laboratories, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
38
|
Beaulieu C, Wu KY, Füzéry AK, Raizman JE, Tsui AKY, Ye C, Basappa NS, Gyenes GT, Koshman SL. Elevated Troponin T in Immune-Checkpoint Inhibitor Myositis: A Case of Mistaken Myocarditis. JACC Case Rep 2024; 29:102462. [PMID: 39295815 PMCID: PMC11405950 DOI: 10.1016/j.jaccas.2024.102462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/30/2024] [Accepted: 06/18/2024] [Indexed: 09/21/2024]
Abstract
A patient with metastatic renal cell carcinoma on axitinib and pembrolizumab had elevated high-sensitivity cardiac troponin T and normal high-sensitivity cardiac troponin I with unremarkable cardiac investigations. A noncardiac cause (myositis) was the likely cause for cardiac troponin T elevation. Cardiac troponin I may be a more appropriate marker to support a myocarditis diagnosis with concurrent myositis.
Collapse
Affiliation(s)
- Carissa Beaulieu
- Division of Medical Oncology, Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Kai Yi Wu
- Division of Cardiology, Department of Medicine, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Anna K Füzéry
- Department of Laboratory of Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Public Laboratories Ltd, Edmonton, Alberta, Canada
| | - Joshua E Raizman
- Department of Laboratory of Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Public Laboratories Ltd, Edmonton, Alberta, Canada
| | - Albert K Y Tsui
- Department of Laboratory of Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
- Alberta Public Laboratories Ltd, Edmonton, Alberta, Canada
| | - Carrie Ye
- Division of Rheumatology, Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Naveen S Basappa
- Division of Medical Oncology, Department of Oncology, University of Alberta, Cross Cancer Institute, Edmonton, Alberta, Canada
| | - Gabor T Gyenes
- Division of Cardiology, Department of Medicine, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| | - Sheri L Koshman
- Division of Cardiology, Department of Medicine, University of Alberta, Mazankowski Alberta Heart Institute, Edmonton, Alberta, Canada
| |
Collapse
|
39
|
Kusuma F, Glenardi G, Mangkuliguna G, Winarto H, Purwoto G, Utami TW, Anggraeni TD. Efficacy, safety, and patient-reported outcome of immune checkpoint inhibitor in gynecologic cancers: A systematic review and meta-analysis of randomized controlled trials. PLoS One 2024; 19:e0307800. [PMID: 39133693 PMCID: PMC11318932 DOI: 10.1371/journal.pone.0307800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/11/2024] [Indexed: 08/15/2024] Open
Abstract
Over the past decades, immune checkpoint inhibitors (ICIs) have shown dramatic efficacy in improving survival rates in multiple malignancies. Recently, gynecological cancer patients also showed to respond favorably to ICI treatment. This study aimed to evaluate the efficacy, safety, and patient-reported outcomes of ICI therapy in gynecological cancers. We conducted a systematic review and meta-analysis by retrieving literature from multiple electronic databases, such as MEDLINE, ScienceDirect, EBSCO, ProQuest, and Google Scholar. The protocol used in this study has been registered in PROSPERO (CRD42022369529). We included a total of 12 trials involving 8 therapies and 8,034 patients. ICI group demonstrated a longer OS (HR: 0.807; 95% CI: 0.719, 0.907; p = 0.000) and greater PFS improvement (HR: 0.809; 95% CI: 0.673, 0.973; p = 0.024) compared to the control group. There was no significant difference in the incidence of treatment-related adverse events [RR: 0.968; 95%CI: 0.936, 1.001; p = 0.061], but a higher incidence of immune-related adverse events (IRAEs) was observed in the ICI group (RR: 3.093; 95%CI: 1.933, 4.798; p = 0.000). Although the mean changes of QOL score from baseline was not significantly different between both groups (SMD: 0.048; 95% CI: -0.106, 0.202; p = 0.542), the time to definitive QOL deterioration was longer in the ICI group (HR: 0.508; 95% CI: 0.461, 0.560; p = 0.000). Despite having a higher incidence of IRAE, ICI was shown to improve survival rates and QOL of patients. Thus, it should be considered as a new standard of care for gynecologic cancers, especially in advanced stages.
Collapse
Affiliation(s)
- Fitriyadi Kusuma
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| | - Glenardi Glenardi
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
- School of Medicine and Health Sciences, Department of Medicine, Atma Jaya Catholic University of Indonesia, North Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
- Lewoleba General Hospital, Lembata Island, East Nusa Tenggara, Indonesia
| | - Ghea Mangkuliguna
- School of Medicine and Health Sciences, Department of Medicine, Atma Jaya Catholic University of Indonesia, North Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| | - Hariyono Winarto
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| | - Gatot Purwoto
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| | - Tofan Widya Utami
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| | - Tricia Dewi Anggraeni
- Division of Oncology Gynecology, Department of Obstetrics and Gynecology, Dr. Cipto Mangunkusumo Hospital, Greater Jakarta, Daerah Khusus Ibukota Jakarta, Indonesia
| |
Collapse
|
40
|
Fenech M, Ajanaku A, Hsuan J, McCormick A, Shamas S, Ghadiri N. Immune checkpoint inhibitors and the orbit; two cases of reactive dacryoadenitis. Orbit 2024:1-5. [PMID: 39109960 DOI: 10.1080/01676830.2024.2385010] [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: 06/20/2024] [Accepted: 07/17/2024] [Indexed: 01/11/2025]
Abstract
Immune checkpoints refer to mechanisms entrusted with the modulation of immune responses in peripheral tissues and are required for minimising collateral damage. Immune checkpoint inhibitors (ICPi) work through numerous pathways, including the anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), anti-PD-1 (programmed cell death protein 1) and the PD-L1 (protein cell death protein-ligand-1) pathways. They are proving to be an exciting therapeutic avenue in the attempt to activate anti-tumour activity. Ipilimumab is a fully human monoclonal antibody working on the anti-CTLA-4 pathway, while nivolumab and pembrolizumab are humanised monoclonal IgG4 antibodies that work on the PD-1 pathway. Despite a growing body of research pertinent to these novel therapies, early indications show that they are limited by their side effect profile. Furthermore, their efficacy appears to be greater in cancers with a high mutational burden. We present two female patients with bilateral reactive dacryoadenitis secondary to ICPi therapy, a finding that to the best of our knowledge was not previously described in the literature.
Collapse
Affiliation(s)
- Matthew Fenech
- Department of Ophthalmology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Ayodeji Ajanaku
- Department of Ophthalmology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - James Hsuan
- Department of Ophthalmology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Austin McCormick
- Department of Ophthalmology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Simon Shamas
- Department of Oncology, The Clatterbridge Cancer Centre NHS Foundation Trust, Bebington, UK
| | - Nima Ghadiri
- Department of Ophthalmology, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
41
|
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, Georgakilas AG. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy. Pharmacol Ther 2024; 260:108670. [PMID: 38823489 DOI: 10.1016/j.pharmthera.2024.108670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece; Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | - Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Yağmur Kafalı
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris I Giotakis
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197 Berlin, Germany
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany; German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany; Genomics Core Facility, Philipps-University, 35043 Marburg, Germany; Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany; Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece.
| |
Collapse
|
42
|
Sunyur AM, Alkhayat D, Mohammad HA, Alahmadi HA, Alharbi LA, Khawaji ZY, Badawi AS. Clinical Patterns and Factors Contributing to Ophthalmic and Otologic Events Associated With Immune Checkpoint Inhibitors: A Narrative Review. Cureus 2024; 16:e66611. [PMID: 39258067 PMCID: PMC11386091 DOI: 10.7759/cureus.66611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2024] [Indexed: 09/12/2024] Open
Abstract
Immune checkpoint inhibitors, which are a type of cancer immunotherapy, have been associated with the development of adverse events related to an overactive immune system caused by the effect of this type of therapy. It affects a wide range of organs, including the ear and eye. Ophthalmic toxicity related to immune checkpoint inhibitors usually occurs bilaterally. Corneal toxicity (mainly dry eye disease) and uveitis are the most commonly reported patterns of toxicity. Other patterns of involvement include optic neuritis, serous retinal detachment, keratitis, ophthalmoplegia, and ocular myasthenia, but are not limited to them. Potential factors contributing to the development of toxicity are age, previous history of ocular immune disease, type, doses, and duration of treatment, and race. Ototoxicity is also reported in the literature, usually manifesting as bilateral, symmetrical/asymmetrical hearing loss. Ear toxicity presenting as ear fullness, tinnitus, and vertigo has also been mentioned in the literature. Hearing loss is often associated with word/speech recognition. An audiogram usually shows a pattern of sensorineural hearing loss. Otitis media has also been reported to be a potential cause of ear toxicity. Immune checkpoint inhibitor toxicity was present more commonly when used along with other anti-neoplastic agents. Ear toxicity, which presumably results from damage to the melanocytes in the ear, often presents with other melanocytotic manifestations, like uveitis and vitiligo. According to the literature, some agents (ipilimumab, nivolumab, atezolizumab, and pembrolizumab) were more commonly associated with toxic effects on the eye and ear and more when combined with each other.
Collapse
Affiliation(s)
- Amal M Sunyur
- Medicine and Surgery, Taibah University, Medina, SAU
| | - Duaa Alkhayat
- Department of Clinical Oncology, Taibah University, Medina, SAU
| | | | | | | | | | | |
Collapse
|
43
|
Cho I, You SC, Cha MJ, Hwang HJ, Cho EJ, Kim HJ, Park SM, Kim SE, Lee YG, Youn JC, Park CS, Shim CY, Chung WB, Sohn IS. Cancer therapy-related cardiac dysfunction and the role of cardiovascular imaging: systemic review and opinion paper from the Working Group on Cardio-Oncology of the Korean Society of Cardiology. J Cardiovasc Imaging 2024; 32:13. [PMID: 39075626 PMCID: PMC11288116 DOI: 10.1186/s44348-024-00014-5] [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: 10/15/2023] [Accepted: 01/14/2024] [Indexed: 07/31/2024] Open
Abstract
Cardio-oncology is a critical field due to the escalating significance of cardiovascular toxicity as a side effect of anticancer treatments. Cancer therapy-related cardiac dysfunction (CTRCD) is a prevalent condition associated with cardiovascular toxicity, necessitating effective strategies for prediction, monitoring, management, and tracking. This comprehensive review examines the definition and risk stratification of CTRCD, explores monitoring approaches during anticancer therapy, and highlights specific cardiovascular toxicities linked to various cancer treatments. These include anthracyclines, HER2-targeted agents, vascular endothelial growth factor inhibitors, immune checkpoint inhibitors, chimeric antigen receptor T-cell therapies, and tumor-infiltrating lymphocytes therapies. Incorporating the Korean data, this review offers insights into the regional nuances in managing CTRCD. Using systematic follow-up incorporating cardiovascular imaging and biomarkers, a better understanding and management of CTRCD can be achieved, optimizing the cardiovascular health of both cancer patients and survivors.
Collapse
Affiliation(s)
- Iksung Cho
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seng-Chan You
- Department of Biomedical Systems Informatics, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Min-Jae Cha
- Department of Radiology, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul, Republic of Korea
| | - Hui-Jeong Hwang
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea
| | - Eun Jeong Cho
- Division of Cardiology, Department of Internal Medicine, Heart and Brain Hospital, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong, Republic of Korea
| | - Hee Jun Kim
- Division of Medical Oncology, Department of Internal Medicine, Chung-Ang University Hospital, Seoul, Republic of Korea
| | - Seong-Mi Park
- Division of Cardiology, Department of Internal Medicine, Korea University College of Medicine, Seoul, Republic of Korea
| | - Sung-Eun Kim
- Department of Cardiovascular Medicine, Kangdong Sacred Heart Hospital, Seoul, Republic of Korea
| | - Yun-Gyoo Lee
- Division of Hematology & Medical Oncology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jong-Chan Youn
- Division of Cardiology, Department of Internal Medicine, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chan Seok Park
- Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Chi Young Shim
- Division of Cardiology, Severance Cardiovascular Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Woo-Baek Chung
- Division of Cardiology, Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Il Suk Sohn
- Department of Cardiology, Kyung Hee University Hospital at Gangdong, Kyung Hee University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
44
|
Li T, Sun S, Li Y, Zhang Y, Wei L. Immunotherapy revolutionizing brain metastatic cancer treatment: personalized strategies for transformative outcomes. Front Immunol 2024; 15:1418580. [PMID: 39136027 PMCID: PMC11317269 DOI: 10.3389/fimmu.2024.1418580] [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: 04/16/2024] [Accepted: 07/10/2024] [Indexed: 08/15/2024] Open
Abstract
Brain metastatic cancer poses a significant clinical challenge, with limited treatment options and poor prognosis for patients. In recent years, immunotherapy has emerged as a promising strategy for addressing brain metastases, offering distinct advantages over conventional treatments. This review explores the evolving landscape of tumor immunotherapy in the context of brain metastatic cancer, focusing on the intricate interplay between the tumor microenvironment (TME) and immunotherapeutic approaches. By elucidating the complex interactions within the TME, including the role of immune cells, cytokines, and extracellular matrix components, this review highlights the potential of immunotherapy to reshape the treatment paradigm for brain metastases. Leveraging immune checkpoint inhibitors, cellular immunotherapies, and personalized treatment strategies, immunotherapy holds promise in overcoming the challenges posed by the blood-brain barrier and immunosuppressive microenvironment of brain metastases. Through a comprehensive analysis of current research findings and future directions, this review underscores the transformative impact of immunotherapy on the management of brain metastatic cancer, offering new insights and opportunities for personalized and precise therapeutic interventions.
Collapse
Affiliation(s)
- Ting Li
- Medical Oncology Department of Thoracic Cancer 1, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Shichen Sun
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yubing Li
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Yanyu Zhang
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| | - Linlin Wei
- Department of Radiotherapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning, China
| |
Collapse
|
45
|
Esen BH, Özbek L, Oğuz S, Selçukbiricik F. Characterizing immune checkpoint inhibitor-related cutaneous adverse reactions: A comprehensive analysis of FDA adverse event reporting system (FAERS) database. Heliyon 2024; 10:e33765. [PMID: 39071598 PMCID: PMC11283008 DOI: 10.1016/j.heliyon.2024.e33765] [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: 10/18/2023] [Revised: 06/06/2024] [Accepted: 06/26/2024] [Indexed: 07/30/2024] Open
Abstract
Background The increasing adoption of immune checkpoint inhibitors (ICIs) in clinical settings highlights their efficacy in treating diverse conditions, while also emphasizing the potential for common cutaneous adverse reactions to arise. The aim of this study is to investigate a multitude of impacting factors and determinants among patients presenting with ICI-associated cutaneous adverse reactions. Methods We conducted a comprehensive analysis of ICI-associated cutaneous adverse reactions using data from the FAERS. Our study spans from January 1, 2015, to March 31, 2023, focusing on ICIs, including anti-PD-1, anti-PD-L1, and anti-CTLA-4 agents. Findings Among the 334,293 reported irAR, 17,431 were identified as cutaneous adverse reactions (ARs). Predominant cutaneous ARs included rash (21.01 %), pruritus (11.22 %), and pemphigoid (3.90 %). Stevens-Johnson syndrome emerged as the most reported severe cutaneous adverse reaction (SCAR) (2.08 %). Anti-CTLA-4 agents exhibited higher cutaneous toxicity compared to anti-PD-1 and anti-PD-L1 agents. Anti-PD-1 agents demonstrated an elevated mortality rate. The combined use of ICIs with chemotherapy amplified the risk of SCAR and mortality. Targeted therapy was a risk factor for cutaneous ARs but was associated with reduced mortality. The median onset day for cutaneous toxicity was 21 days, while for SCAR, it was 23 days. Weight and age were identified as predictors of SCAR, cutaneous toxicity, and mortality. Skin cancer increased skin toxicity, while lung cancer heightened SCAR formation. The number of administered ICIs positively correlated with SCAR, skin toxicity, and mortality. Interpretation This study highlights the significance of early identification and effective management of cutaneous toxicities, along with personalized follow-up care, as essential strategies for minimizing risks and preventing treatment disruptions.
Collapse
Affiliation(s)
| | - Laşin Özbek
- Koç University, School of Medicine, İstanbul, Turkey
| | - Sinem Oğuz
- Koç University, School of Medicine, İstanbul, Turkey
| | - Fatih Selçukbiricik
- Koç University, School of Medicine, İstanbul, Turkey
- Koç University Hospital, Department of Medical Oncology, İstanbul, Turkey
| |
Collapse
|
46
|
Hwang JK, Marston DJ, Wrapp D, Li D, Tuyishime M, Brackenridge S, Rhodes B, Quastel M, Kapingidza AB, Gater J, Harner A, Wang Y, Rountree W, Ferrari G, Borrow P, McMichael AJ, Gillespie GM, Haynes BF, Azoitei ML. A high affinity monoclonal antibody against HLA-E-VL9 enhances natural killer cell anti-tumor killing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602401. [PMID: 39026709 PMCID: PMC11257447 DOI: 10.1101/2024.07.08.602401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Natural killer (NK) cells kill target cells following triggering via germline-encoded receptors interacting with target cell-expressed ligands (direct killing), or via antibody-dependent cellular cytotoxicity (ADCC) mediated by FcγRIIIa. NK cytotoxicity is modulated by signaling through activating or inhibitory receptors. A major checkpoint is mediated by the NK inhibitory receptor NKG2A/CD94 and its target cell ligand, HLA-E, which is complexed with HLA signal sequence-derived peptides termed VL9 (HLA-E-VL9). We have previously reported the isolation of a murine HLA-E-VL9-specific IgM antibody 3H4 and the generation of a higher affinity IgG version (3H4v3). Here we have used phage display library selection to generate a high affinity version of 3H4v3, called 3H4v31, with an ∼700 fold increase in binding affinity. We show using an HLA-E-VL9+ K562 tumor model that, in vitro, the addition of 3H4v31 to target cells increased direct killing of targets by CD16-negative NK cell line NK-92 and also mediated ADCC by NK-92 cells transfected with CD16. Moreover, ADCC by primary NK cells was also enhanced in vitro by 3H4v31. 3H4v31 was also able to bind and enhance target cell lysis of endogenously expressed HLA-E-VL9 on human cervical cancer and human pancreatic cancer cell lines. In vivo, 3H4v31 slowed the growth rate of HLA-E-VL9+ K562 tumors implanted into NOD/SCID/IL2rγ null mice compared to isotype control when injected with NK-92 cells intratumorally. Together, these data demonstrate that mAb 3H4v31 can enhance NK cell killing of HLA-E-VL9-expressing tumor cells in vitro by both direct killing activity and by ADCC. Moreover, mAb 3H4v31 can enhance NK cell control of tumor growth in vivo. We thus identify HLA-E-VL9 monoclonal antibodies as a promising novel anti-tumor immunotherapy. One Sentence Summary A high affinity monoclonal antibody against HLA-E-VL9 enhances natural killer cell anti-tumor killing by checkpoint inhibition and antibody dependent cellular cytotoxicity.
Collapse
|
47
|
Pasello G, Pavan A, De Nuzzo M, Frega S, Ferro A, Dal Maso A, Bonanno L, Guarneri V, Girardi F. Immune-related adverse events in patients treated with immunotherapy for locally advanced or metastatic NSCLC in real-world settings: a systematic review and meta-analysis. Front Oncol 2024; 14:1415470. [PMID: 39045561 PMCID: PMC11263096 DOI: 10.3389/fonc.2024.1415470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Randomized clinical trials (RCTs) represent the mainstay for the approval of new treatments. However, stringent inclusion criteria often cause them to depart from the daily clinical practice. Real-world (RW) evidence have a complementing role, filling the gap between the efficacy of a treatment and its effectiveness. Immune checkpoint inhibitors (ICIs) have changed the treatment scenario for non-small cell lung cancer (NSCLC); immune-related adverse events (irAEs) could become life-threatening events, when not timely managed. We performed a systematic review and meta-analysis on the RW impact of irAEs through the years. Methods The systematic review focused on irAEs occurred in locally advanced or metastatic NSCLC patients, treated with ICIs in a RW setting. We queried two electronic databases (Embase and Medline) from 1996 to August 2022. We then conducted a meta-analysis dividing the results in two cohorts (2015-2018 and 2019-2021). We described the prevalence of patients with irAEs of any or severe grade (G). Estimates were expressed as proportions up to the second decimal point (effect size, ES). IrAEs of interest were those involving the skin, the liver, the endocrine system or the gastro-intestinal system. Results Overall, 21 RW studies on 5,439 patients were included in the quantitative and qualitative synthesis. The prevalence of G≥3 irAEs was slightly lower in the 2015-2018 subgroup, while the prevalence of irAEs of any grade was similar for both periods. Overall, we observed a higher ES for gastrointestinal, hepatic and lung irAEs, while a lower ES was reported for skin or endocrine irAEs. Endocrine irAEs were reported in 10 out of 21 studies, with a slight increase in the most recent studies, while cutaneous toxicities were mostly reported in two studies lead within the first time-period. Pulmonary, gastrointestinal, and hepatic toxicities, showed a more heterogeneous distribution of ES over time. Discussion Our findings showed that the frequency of irAEs remained stable across the two calendar periods examined in our meta-analysis. This finding suggests that RW data might not be able to identify a potential learning curve in detection and management of irAEs.
Collapse
Affiliation(s)
- Giulia Pasello
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - Alberto Pavan
- Medical Oncology Department, Azienda ULSS 3 Serenissima, Dell’Angelo General Hospital, Mestre and SS Giovanni e Paolo General Hospital, Venice, Italy
| | - Mattia De Nuzzo
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - Stefano Frega
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - Alessandra Ferro
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | | | - Laura Bonanno
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - Valentina Guarneri
- Department of Surgery, Oncology and Gastroenterology, University of Padova, Padua, Italy
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| | - Fabio Girardi
- Oncologia 2, Istituto Oncologico Veneto (IOV) IRCCS, Padua, Italy
| |
Collapse
|
48
|
Tan S, Ding X, Pan D, Xu Y, Wang C, Yan J, Chen C, Wang L, Wang X, Yang M, Xu Y. Synthesis and Characterization of a Novel PET Tracer for Noninvasive Evaluation of FGL1 Status in Tumors. Mol Pharm 2024; 21:3425-3433. [PMID: 38836286 DOI: 10.1021/acs.molpharmaceut.4c00137] [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] [Indexed: 06/06/2024]
Abstract
Fibrinogen-like protein 1 (FGL1) is a potential novel immune checkpoint target for malignant tumor diagnosis and therapy. Accurate detection of FGL1 levels in tumors via noninvasive PET imaging might be beneficial for managing the disease. To achieve this, multiple FGL1-targeting peptides (FGLP) were designed, and a promising candidate, 68Ga-NOTA-FGLP2, was identified through a high-throughput screening approach using microPET imaging of 68Ga-labeled peptides. Subsequent in vitro cell experiments showed that uptake values of 68Ga-NOTA-FGLP2 in FGL1 positive Huh7 tumor cells were significantly higher than those in FGL1 negative U87 MG tumor cells. Further microPET imaging showed that the Huh7 xenografts were clearly visualized with a favorable contrast. ROI analysis showed that the uptake values of the tracer in Huh7 xenografts were 2.63 ± 0.07% ID/g at 30 min p.i.. After treatment with an excess of unlabeled FGLP2, the tumor uptake significantly decreased to 0.54 ± 0.05% ID/g at 30 min p.i.. Moreover, the uptake in U87 MG xenografts was 0.44 ± 0.06% ID/g at the same time point. The tracer was excreted mainly through the renal system. 18F-FDG PET imaging was also performed in mice bearing Huh7 and U87 MG xenografts, respectively. However, there was no significant difference in the uptake between the tumors with different FGL1 expressions. Preclinical data indicated that 68Ga-NOTA-FGLP2 might be a suitable radiotracer for in vivo noninvasive visualization of tumors with abundant expression of FGL1. Further investigation of 68Ga-NOTA-FGLP2 for tumor diagnosis and therapy is undergoing.
Collapse
Affiliation(s)
- Siyi Tan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang Ding
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yue Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ce Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Junjie Yan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Chongyang Chen
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Lizhen Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Xinyu Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Min Yang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| | - Yuping Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, China
| |
Collapse
|
49
|
Wang Y, Xiong C, Yu W, Zhou M, Shugg TA, Hsu FC, Eadon MT, Su J, Song Q. PCCA variant rs16957301 is a novel AKI risk genotype-specific for patients who receive ICI treatment: Real-world evidence from All of Us cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.20.24309197. [PMID: 38946978 PMCID: PMC11213073 DOI: 10.1101/2024.06.20.24309197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Immune checkpoint inhibitors (ICIs) enhance the immune system's ability to target and destroy cancer cells by blocking inhibitory pathways. Despite their efficacy, these treatments can trigger immune-related adverse events (irAEs), such as acute kidney injury (ICI-AKI), complicating patient management. The genetic predispositions to ICI-AKI are not well understood, necessitating comprehensive genomic studies to identify risk factors and improve therapeutic strategies. Objective To identify genetic predispositions for ICI-AKI using large-scale real-world data. Methods A systematic literature search led to 14 candidate variants related to irAEs. We performed a candidate variant association study with these 14 variants using the All of Us cohort (AoU, v7, cutoff date: 7/1/2022). A cohort for cancer patients receiving ICI and a general cohort were established to evaluate ICI-AKI risk. Logistic regression, adjusted for sex, was used to evaluate the impact of each candidate genotype, separately for self-reported and ancestry-estimated race. Kaplan-Meier survival analysis assessed the genetic effects on AKI-free survival. Results The ICI cohort (n=414) showed a one-year AKI incidence rate of 23.2%, significantly higher than the general cohort (6.5%, n=213,282). The rs16957301 variant (chr13:100324308, T>C) in the PCCA gene was a significant risk genotype for ICI-AKI among self-reported Caucasians (Beta=0.93, Bonferroni-corrected P-value=0.047) and ancestry estimated Caucasians (Beta = 0.94, Bonferroni-corrected P-value=0.044). Self-reported Caucasians with the rs16957301 risk genotypes (TC/CC) developed AKI significantly earlier (3.6 months) compared to the reference genotype (TT, 7.0 months, log-rank P=0.04). Consistent results were found in ancestry-estimated Caucasians. This variant did not present significant AKI risks in the general cohort (Beta: -0.008-0.035, FDR: 0.75-0.99). Conclusion Real-world evidence from the All of Us cohort suggests that, in Caucasians, PCCA variant rs16957301 is a novel AKI risk genotype specific to ICI treatment. Additional studies are warranted to validate rs16957301 as risk marker for AKI in Caucasian patients treated with ICIs and to assess its risk in other ancestral populations.
Collapse
|
50
|
Ke B, Jin P, Wang XJ, Liu N, Liang H, Zhang RP. Oncogenic and immunological role of EDIL3 in human tumours: From pan-cancer analysis to validation in gastric cancer. Heliyon 2024; 10:e32291. [PMID: 38882287 PMCID: PMC11180325 DOI: 10.1016/j.heliyon.2024.e32291] [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: 01/02/2024] [Revised: 05/30/2024] [Accepted: 05/31/2024] [Indexed: 06/18/2024] Open
Abstract
Background Epidermal growth factor-like repeats and discoidin I-like domains 3 (EDIL3) is a secreted extracellular matrix protein implicated in diverse physiological and pathological processes including embryonic development, angiogenesis, and anti-inflammatory responses. Recent reports have indicated that EDIL3 play critical roles in carcinogenesis and progression of many cancers. Herein, we performed a pan-cancer investigation to study the potential functions of EDIL3 in various cancers and experimentally validate its function in gastric cancer (GC). Methods We analysed EDIL3 expression profiles in different tumours using The Cancer Genome Atlas database. The Kaplan-Meier Plotter was used to investigate the prognostic value of EDIL3, while receiver operating characteristic curve was performed to analyze its diagnostic efficacy. Several bioinformatics tools were used to study the association between EDIL3 and promoter methylation, gene enrichment analysis, immune infiltration, immune-related genes, and drug sensitivity. Molecular biology experiments were conducted to validate the tumorigenic effects of EDIL3. Results EDIL3 is variably expressed in different cancers and is closely associated with clinical outcomes. An inverse correlation between EDIL3 and DNA methylation has been observed in 13 cancers. Enrichment analysis indicated that EDIL3 is correlated with many cellular pathways such as extracellular matrix receptor interactions and focal adhesion. EDIL3 was tightly associated with immune infiltration and immune checkpoints. EDIL3 knockdown can promote GC calls apoptosis while preventing proliferation, migration, and invasion in vitro. Conclusion EDIL3 is a promising prognostic, diagnostic, and immunological biomarker in various cancers, which could be applied as a new target for cancer therapy.
Collapse
Affiliation(s)
- Bin Ke
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Peng Jin
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Xue-Jun Wang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Ning Liu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Han Liang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| | - Ru-Peng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, 300060, Tianjin, China
| |
Collapse
|