1
|
Beckmann M, Schlüter J, Erdmann M, Kramer R, Cunningham S, Hackstein H, Zimmermann R, Heinzerling L. Interdependence of coagulation with immunotherapy and BRAF/MEK inhibitor therapy: results from a prospective study. Cancer Immunol Immunother 2024; 74:5. [PMID: 39487855 DOI: 10.1007/s00262-024-03850-y] [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: 04/14/2024] [Accepted: 09/27/2024] [Indexed: 11/04/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapies effectively treat a broadening spectrum of cancer entities but induce various immune-related side effects (irAEs). Recent reports suggest a correlation between ICI-induced systemic inflammation and thromboembolic events as well as an increased effectiveness by coadministration of anticoagulants. With cancer patients having a higher risk of thrombotic events per se, it is crucial to dissect and characterize the mechanisms that cause pro-coagulative effects induced by systemic tumor therapies and their potential interplay with anti-tumor response. A total of 31 patients with advanced skin cancer treated with either ICIs (n = 24) or BRAF/MEK inhibitors (n = 7) were longitudinally assessed for blood and coagulation parameters before as well as 7, 20 and 40 days after initiation of systemic tumor therapy. Changes were analyzed and compared between both groups. In addition, the influence of coagulation parameters on progression-free, recurrence-free and overall survival was investigated. The ICI cohort presented significantly increased factor VIII activity after one week of therapy (p 0.0225); while, protein S activity was reduced during the whole observation period. Additionally, von Willebrand factor activity and tissue factor concentrations increased under immunotherapy. Similar changes occurred under BRAF/MEK inhibitor therapy (BRAF/MEKi). Increased baseline levels of von Willebrand factor antigen and factor VIII:C before the start of ICI therapy correlated with a significantly higher risk of recurrence for patients receiving adjuvant immunotherapy. The findings suggest the induction of a pro-coagulant state under ICI and BRAF/MEKi and a role of coagulation parameters in the efficacy of ICI therapies.
Collapse
Affiliation(s)
- Malte Beckmann
- Department of Dermatology, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), University Hospital Erlangen, Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Julian Schlüter
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Michael Erdmann
- Department of Dermatology, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), University Hospital Erlangen, Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Rafaela Kramer
- Department of Dermatology, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), University Hospital Erlangen, Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Robert Zimmermann
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany
| | - Lucie Heinzerling
- Department of Dermatology, Comprehensive Cancer Center Erlangen-European Metropolitan Area of Nuremberg (CCC ER-EMN), University Hospital Erlangen, Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nuremberg (FAU), 91054, Erlangen, Germany.
- Department of Dermatology and Allergology, LMU University Hospital Munich, Ludwig-Maximilian University, Frauenlobstraße 9 - 11, 80337, Munich, Germany.
| |
Collapse
|
2
|
Gergely TG, Drobni ZD, Sayour NV, Ferdinandy P, Varga ZV. Molecular fingerprints of cardiovascular toxicities of immune checkpoint inhibitors. Basic Res Cardiol 2024:10.1007/s00395-024-01068-8. [PMID: 39023770 DOI: 10.1007/s00395-024-01068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by unleashing the power of the immune system against malignant cells. However, their use is associated with a spectrum of adverse effects, including cardiovascular complications, which can pose significant clinical challenges. Several mechanisms contribute to cardiovascular toxicity associated with ICIs. First, the dysregulation of immune checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) and its ligand (PD-L1), and molecular mimicry with cardiac autoantigens, leads to immune-related adverse events, including myocarditis and vasculitis. These events result from the aberrant activation of T cells against self-antigens within the myocardium or vascular endothelium. Second, the disruption of immune homeostasis by ICIs can lead to autoimmune-mediated inflammation of cardiac tissues, manifesting as cardiac dysfunction and heart failure, arrhythmias, or pericarditis. Furthermore, the upregulation of inflammatory cytokines, particularly tumor necrosis factor-alpha, interferon-γ, interleukin-1β, interleukin-6, and interleukin-17 contributes to cardiac and endothelial dysfunction, plaque destabilization, and thrombosis, exacerbating cardiovascular risk on the long term. Understanding the intricate mechanisms of cardiovascular side effects induced by ICIs is crucial for optimizing patient care and to ensure the safe and effective integration of immunotherapy into a broader range of cancer treatment protocols. The clinical implications of these mechanisms underscore the importance of vigilant monitoring and early detection of cardiovascular toxicity in patients receiving ICIs. Future use of these key pathological mediators as biomarkers may aid in prompt diagnosis of cardiotoxicity and will allow timely interventions.
Collapse
Affiliation(s)
- Tamás G Gergely
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia D Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Péter Ferdinandy
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
3
|
Sakurai T, Yamahara N, Yaguchi T, Baba Y, Nishida H. Immune checkpoint inhibitor-induced Trousseau syndrome: a case report. Acta Neurol Belg 2024; 124:1033-1036. [PMID: 37897651 DOI: 10.1007/s13760-023-02413-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023]
Affiliation(s)
- Takeo Sakurai
- Department of Neurology, Gifu Prefectural General Medical Center, 4-6-1 Noisshiki, Gifu, 500-8717, Japan.
| | - Naoki Yamahara
- Department of Neurology, Gifu Prefectural General Medical Center, 4-6-1 Noisshiki, Gifu, 500-8717, Japan
| | - Tomonori Yaguchi
- Department of Neurology, Gifu Prefectural General Medical Center, 4-6-1 Noisshiki, Gifu, 500-8717, Japan
| | - Yasutomo Baba
- Department of Respiratory Medicine, Gifu Prefectural General Medical Center, Gifu, 500-8717, Japan
| | - Hiroshi Nishida
- Department of Neurology, Gifu Prefectural General Medical Center, 4-6-1 Noisshiki, Gifu, 500-8717, Japan
| |
Collapse
|
4
|
Yang M, Cao H, Wang C, Yu C, Sun P. Incidence of thromboembolic events in non-small cell lung cancer patients treated with immune checkpoint inhibitors: A systematic review and meta-analysis. J Cancer Res Ther 2024; 20:509-521. [PMID: 38687920 DOI: 10.4103/jcrt.jcrt_1031_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 01/08/2024] [Indexed: 05/02/2024]
Abstract
ABSTRACT The incidence of thromboembolic events (TEs) in non-small cell lung cancer (NSCLC) patients treated with immune checkpoint inhibitors (ICIs) has rarely been reported. The MEDLINE, EMBASE, and the Cochrane Library databases were searched. The primary outcome was the incidence of TEs, and the secondary outcome was the relationship between TEs and overall survival (OS) following ICI therapy. A subgroup analysis of TE incidents was performed according to the TE type and combination regimens. The I2 statistic was used to determine the heterogeneity, and funnel plots and Egger's test were used to assess publication bias. A total of 16,602 patients with NSCLC in 63 experimental arms were included in the analysis. The rate of TEs ranged from 0.1% to 13.8%, and the pooled overall incidence of all-grade TEs was 3% (95% confidence interval [CI], 2%-4%). The pooled rate of high-grade TEs was 1% (95% CI, 1%-2%). The venous and arterial TE rates were 3% (95% CI, 2%-4%) and 1% (95% CI, 1%-2%), respectively. Patients who received immunotherapy + chemoradiotherapy had the highest incidence of TEs (7%). The TE pooled rate was higher in patients treated with combined ICIs than in those treated with mono ICIs (4% vs. 2%). The OS was lower in patients with TEs than in those without TEs (hazard ratio, 1.4; 95% CI, 1.02%-1.92%). The incidence of TEs in NSCLC patients treated with ICIs was reasonable. Nonetheless, clinicians must be aware of potential thrombotic complications and treat them promptly.
Collapse
Affiliation(s)
- Miaomiao Yang
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Hongxin Cao
- Department of Medical Oncology, Qilu Hospital of Shandong University, Jinan, Shandong, P.R. China
| | - Congcong Wang
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Caiyan Yu
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| | - Ping Sun
- Department of Oncology, Yantai Yuhuangding Hospital, Affiliated with Medical College of Qingdao University, Yantai, Shandong, P.R. China
| |
Collapse
|
5
|
Liu G, Chen T, Zhang X, Hu B, Shi H. Immune checkpoint inhibitor-associated cardiovascular toxicities: A review. Heliyon 2024; 10:e25747. [PMID: 38434280 PMCID: PMC10907684 DOI: 10.1016/j.heliyon.2024.e25747] [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: 07/19/2023] [Revised: 02/01/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionary effects on therapeutic strategies for multiple malignancies. Their efficacy depends on their ability to reactivate the host immune system to fight cancer cells. However, adverse reactions to ICIs are common and involve several organs, limiting their use in clinical practice. Although the incidence of cardiovascular toxicity is relatively low, it is associated with serious consequences and high mortality rates. The primary cardiovascular toxicities include myocarditis, pericarditis, Takotsubo syndrome, arrhythmia, vasculitis, acute coronary syndrome, and venous thromboembolism. Currently, the mechanism underlying ICI-associated cardiovascular toxicity remains unclear and underexplored. The diagnosis and monitoring of ICI-associated cardiovascular toxicities mainly include the following indicators: symptoms, signs, laboratory examination, electrocardiography, imaging, and pathology. Treatments are based on the grade of cardiovascular toxicity and mainly include drug withdrawal, corticosteroid therapy, immunosuppressants, and conventional cardiac treatment. This review focuses on the incidence, underlying mechanisms, clinical manifestations, diagnoses, and treatment strategies.
Collapse
Affiliation(s)
- Guihong Liu
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Chen
- Tao Chen Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xin Zhang
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Binbin Hu
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Huashan Shi
- Guihong Liu Department of Biotherapy, State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Tan Q, Gao R, Zhang X, Yang J, Xing P, Yang S, Wang D, Wang G, Wang S, Yao J, Zhang Z, Tang L, Yu X, Han X, Shi Y. Longitudinal plasma proteomic analysis identifies biomarkers and combinational targets for anti-PD1-resistant cancer patients. Cancer Immunol Immunother 2024; 73:47. [PMID: 38349411 PMCID: PMC10864508 DOI: 10.1007/s00262-024-03631-7] [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: 11/21/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024]
Abstract
The response rate of anti-PD1 therapy is limited, and the influence of anti-PD1 therapy on cancer patients is unclear. To address these challenges, we conducted a longitudinal analysis of plasma proteomic changes with anti-PD1 therapy in non-small cell lung cancer (NSCLC), alveolar soft part sarcoma (ASPS), and lymphoma patients. We included 339 plasma samples before and after anti-PD1 therapy from 193 patients with NSCLC, ASPS, or lymphoma. The plasma proteins were detected using data-independent acquisition-mass spectrometry and customable antibody microarrays. Differential proteomic characteristics in responders (R) and non-responders (NR) before and after anti-PD1 therapy were elucidated. A total of 1019 proteins were detected using our in-depth proteomics platform and distributed across 10-12 orders of abundance. By comparing the differential plasma proteome expression between R and NR groups, 50, 206, and 268 proteins were identified in NSCLC, ASPS, and lymphoma patients, respectively. Th17, IL-17, and JAK-STAT signal pathways were identified upregulated in NR group, while cellular senescence and transcriptional misregulation pathways were activated in R group. Longitudinal proteomics analysis revealed the IL-17 signaling pathway was downregulated after treatment. Consistently, many proteins were identified as potential combinatorial therapeutic targets (e.g., IL-17A and CD22). Five noninvasive biomarkers (FLT4, SFTPB, GNPTG, F5, and IL-17A) were further validated in an independent lymphoma cohort (n = 39), and another three noninvasive biomarkers (KIT, CCL3, and TNFSF1) were validated in NSCLC cohort (n = 76). Our results provide molecular insights into the anti-PD1 therapy in cancer patients and identify new therapeutic strategies for anti-PD1-resistant patients.
Collapse
Affiliation(s)
- Qiaoyun Tan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Ruyun Gao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xiaomei Zhang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Jianliang Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Puyuan Xing
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Sheng Yang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Dan Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Guibing Wang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Shasha Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Jiarui Yao
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Zhishang Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Le Tang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China
| | - Xiaobo Yu
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences-Beijing (PHOENIX Center), Beijing Institute of Lifeomics, Beijing, 102206, China
| | - Xiaohong Han
- Clinical Pharmacology Research Center, Peking Union Medical College Hospital, State Key Laboratory of Complex Severe and Rare Diseases, NMPA Key Laboratory for Clinical Research and Evaluation of Drug, Beijing Key Laboratory of Clinical PK & PD Investigation for Innovative Drugs, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100730, China.
| | - Yuankai Shi
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing Key Laboratory of Clinical Study On Anticancer Molecular Targeted Drugs, Beijing, 100021, China.
| |
Collapse
|
7
|
Freitas-Dias C, Gonçalves F, Martins F, Lemos I, Gonçalves LG, Serpa J. Interaction between NSCLC Cells, CD8 + T-Cells and Immune Checkpoint Inhibitors Potentiates Coagulation and Promotes Metabolic Remodeling-New Cues on CAT-VTE. Cells 2024; 13:305. [PMID: 38391918 PMCID: PMC10886748 DOI: 10.3390/cells13040305] [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: 01/03/2024] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/24/2024] Open
Abstract
BACKGROUND Cancer-associated thrombosis (CAT) and venous thromboembolism (VTE) are frequent cancer-related complications associated with high mortality; thus, this urges the identification of predictive markers. Immune checkpoint inhibitors (ICIs) used in cancer immunotherapy allow T-cell activation against cancer cells. Retrospective studies showed increased VTE following ICI administration in some patients. Non-small cell lung cancer (NSCLC) patients are at high risk of thrombosis and thus, the adoption of immunotherapy, as a first-line treatment, seems to be associated with coagulation-fibrinolysis derangement. METHODS We pharmacologically modulated NSCLC cell lines in co-culture with CD8+ T-cells (TCD8+) and myeloid-derived suppressor cells (MDSCs), isolated from healthy blood donors. The effects of ICIs Nivolumab and Ipilimumab on NSCLC cell death were assessed by annexin V and propidium iodide (PI) flow cytometry analysis. The potential procoagulant properties were analyzed by in vitro clotting assays and enzyme-linked immunosorbent assays (ELISAs). The metabolic remodeling induced by the ICIs was explored by 1H nuclear magnetic resonance (NMR) spectroscopy. RESULTS Flow cytometry analysis showed that TCD8+ and ICIs increase cell death in H292 and PC-9 cells but not in A549 cells. Conditioned media from NSCLC cells exposed to TCD8+ and ICI induced in vitro platelet aggregation. In A549, Podoplanin (PDPN) levels increased with Nivolumab. In H292, ICIs increased PDPN levels in the absence of TCD8+. In PC-9, Ipilimumab decreased PDPN levels, this effect being rescued by TCD8+. MDSCs did not interfere with the effect of TCD8+ in the production of TF or PDPN in any NSCLC cell lines. The exometabolome showed a metabolic remodeling in NSCLC cells upon exposure to TCD8+ and ICIs. CONCLUSIONS This study provides some insights into the interplay of immune cells, ICIs and cancer cells influencing the coagulation status. ICIs are important promoters of coagulation, benefiting from TCD8+ mediation. The exometabolome analysis highlighted the relevance of acetate, pyruvate, glycine, glutamine, valine, leucine and isoleucine as biomarkers. Further investigation is needed to validate this finding in a cohort of NSCLC patients.
Collapse
Affiliation(s)
- Catarina Freitas-Dias
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (C.F.-D.); (F.G.); (F.M.); (I.L.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
- Faculdade de Ciências, FCUL, Universidade de Lisboa, Campo Grande, 130, 1169-056 Lisboa, Portugal
| | - Filipe Gonçalves
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (C.F.-D.); (F.G.); (F.M.); (I.L.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Filipa Martins
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (C.F.-D.); (F.G.); (F.M.); (I.L.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Isabel Lemos
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (C.F.-D.); (F.G.); (F.M.); (I.L.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| | - Luís G. Gonçalves
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157 Oeiras, Portugal;
| | - Jacinta Serpa
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, NMS, FCM, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056 Lisboa, Portugal; (C.F.-D.); (F.G.); (F.M.); (I.L.)
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023 Lisboa, Portugal
| |
Collapse
|
8
|
Inoue T, Kumai T, Ohara K, Takahara M. Cerebral Infarction as a Rare Adverse Event of Immune Checkpoint Inhibitors in Patients With Head and Neck Squamous Cell Carcinoma: A Case Series. Cureus 2023; 15:e47406. [PMID: 38021593 PMCID: PMC10658212 DOI: 10.7759/cureus.47406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are a novel treatment option for treating head and neck squamous cell carcinoma (HNSCC). Among the immune-related adverse effects, cerebral infarction (CI) is a rare but fatal complication, and it has been reported in various cancers, except HNSCC. Herein, we describe three cases of patients diagnosed with HNSCC who experienced CI following ICI treatment. In addition, we conducted a comprehensive literature review on ICI-related thrombosis. Three patients with recurrent HNSCC were treated with nivolumab. Two patients had a history of CI, or heart disease, and were concurrently prescribed antithrombotic medications during nivolumab treatment. The number of nivolumab administrations varied from 1-25 before the onset of CI. All patients experienced worsening of neurological symptoms due to CI, irrespective of antithrombotic treatment, and they ultimately succumbed to the disease within 16-222 days following their initial ICI administration. ICIs may cause thromboembolisms, leading to CI. Based on our review of the literature, a history of thromboembolism or heart disease could be a risk factor for ICI-related thrombosis.
Collapse
Affiliation(s)
- Takahiro Inoue
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, JPN
| | - Takumi Kumai
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, JPN
| | - Kenzo Ohara
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, JPN
| | - Miki Takahara
- Department of Otolaryngology, Head and Neck Surgery, Asahikawa Medical University, Asahikawa, JPN
| |
Collapse
|
9
|
Li SY, Guo YL, Tian JW, Zhang HJ, Li RF, Gong P, Yu ZL. Anti-Tumor Strategies by Harnessing the Phagocytosis of Macrophages. Cancers (Basel) 2023; 15:2717. [PMID: 37345054 DOI: 10.3390/cancers15102717] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 05/07/2023] [Accepted: 05/09/2023] [Indexed: 06/23/2023] Open
Abstract
Macrophages are essential for the human body in both physiological and pathological conditions, engulfing undesirable substances and participating in several processes, such as organism growth, immune regulation, and maintenance of homeostasis. Macrophages play an important role in anti-bacterial and anti-tumoral responses. Aberrance in the phagocytosis of macrophages may lead to the development of several diseases, including tumors. Tumor cells can evade the phagocytosis of macrophages, and "educate" macrophages to become pro-tumoral, resulting in the reduced phagocytosis of macrophages. Hence, harnessing the phagocytosis of macrophages is an important approach to bolster the efficacy of anti-tumor treatment. In this review, we elucidated the underlying phagocytosis mechanisms, such as the equilibrium among phagocytic signals, receptors and their respective signaling pathways, macrophage activation, as well as mitochondrial fission. We also reviewed the recent progress in the area of application strategies on the basis of the phagocytosis mechanism, including strategies targeting the phagocytic signals, antibody-dependent cellular phagocytosis (ADCP), and macrophage activators. We also covered recent studies of Chimeric Antigen Receptor Macrophage (CAR-M)-based anti-tumor therapy. Furthermore, we summarized the shortcomings and future applications of each strategy and look into their prospects with the hope of providing future research directions for developing the application of macrophage phagocytosis-promoting therapy.
Collapse
Affiliation(s)
- Si-Yuan Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Yong-Lin Guo
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Jia-Wen Tian
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - He-Jing Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Rui-Fang Li
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Ping Gong
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Anesthesiology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| | - Zi-Li Yu
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) & Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
10
|
Stadler JC, Keller L, Mess C, Bauer AT, Koett J, Geidel G, Heidrich I, Vidal-Y-Sy S, Andreas A, Stramaglia C, Sementsov M, Haberstroh W, Deitert B, Hoehne IL, Reschke R, Haalck T, Pantel K, Gebhardt C, Schneider SW. Prognostic value of von Willebrand factor levels in patients with metastatic melanoma treated by immune checkpoint inhibitors. J Immunother Cancer 2023; 11:jitc-2022-006456. [PMID: 37258039 DOI: 10.1136/jitc-2022-006456] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/28/2023] [Indexed: 06/02/2023] Open
Abstract
BACKGROUND An increased incidence of thrombotic complications associated with an increased mortality rate has been observed under immune checkpoint inhibition (ICI). Recent investigations on the coagulation pathways have highlighted the direct role of key coagulatory proteins and platelets in cancer initiation, angiogenesis and progression. The aim of this study was to evaluate the prognostic value of von Willebrand factor (vWF) and its regulatory enzyme a disintegrin and metalloproteinase with a thrombospondin type 1 motif, member 13 (ADAMTS13), D-dimers and platelets in a cohort of patients with metastatic melanoma receiving ICI. METHODS In a prospective cohort of 83 patients with metastatic melanoma, we measured the systemic levels of vWF-antigen (vWF:Ag), ADAMTS13 activity, D-dimers and platelets, before the beginning of the treatment (baseline), and 6, 12 and 24 weeks after. In parallel, we collected standard biological parameters used in clinical routine to monitor melanoma response (lactate deshydrogenase (LDH), S100). The impact of neutrophil-to-lymphocyte ratio (NLR) and C-reactive protein (CRP) on overall survival (OS) in patients receiving ICI was assessed. Univariable and multivariable Cox proportional models were then used to investigate any potential association of these parameters to clinical progression (progression-free survival (PFS) and OS). Baseline values and variations over therapy course were compared between primary responders and resistant patients. RESULTS Patients with melanoma present with dysregulated levels of vWF:Ag, ADAMTS13 activity, D-dimers, LDH, S100 and CRP at the beginning of treatment. With a median clinical follow-up of 26 months, vWF:Ag interrogated as a continuous variable was significantly associated with PFS in univariate and multivariate analysis (HR=1.04; p=0.007). Lower values of vWF:Ag at baseline were observed in the primary responders group (median: 29.4 µg/mL vs 32.9 µg/mL; p=0.048) when compared with primary resistant patients. As for OS, we found an association with D-dimers and ADAMTS13 activity in univariate analysis and vWF:Ag in univariate and multivariate analysis including v-raf murine sarcoma viral oncogene homolog B1 (BRAF) mutation and Eastern Cooperative Oncology Group (ECOG) performance status. Follow-up over the course of treatment depicts different evolution profiles for vWF:Ag between the primary response and resistance groups. CONCLUSIONS In this prospective cohort, coagulatory parameters such as ADAMTS13 activity and D-dimers are associated with OS but baseline vWF:Ag levels appeared as the only parameter associated with response and OS to ICI. This highlights a potential role of vWF as a biomarker to monitor ICI response of patients with malignant melanoma.
Collapse
Affiliation(s)
- Julia-Christina Stadler
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Laura Keller
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Centre de Recherche en Cancerologie de Toulouse, Toulouse, France
| | - Christian Mess
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Alexander T Bauer
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Koett
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Glenn Geidel
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Isabel Heidrich
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Sabine Vidal-Y-Sy
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antje Andreas
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carlotta Stramaglia
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mark Sementsov
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Wiebcke Haberstroh
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Benjamin Deitert
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Inka Lilott Hoehne
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Reschke
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Haalck
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Klaus Pantel
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christoffer Gebhardt
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan W Schneider
- Department of Dermatology and Venereology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Fleur Hiege Center for Skin Cancer Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
11
|
Özdemir BC, Espinosa da Silva C, Arangalage D, Monney P, Guler SA, Huynh-Do U, Stirnimann G, Possamai L, Trepp R, Hoepner R, Salmen A, Gerard CL, Hruz P, Christ L, Rothschild SI. Multidisciplinary recommendations for essential baseline functional and laboratory tests to facilitate early diagnosis and management of immune-related adverse events among cancer patients. Cancer Immunol Immunother 2023:10.1007/s00262-023-03436-0. [PMID: 37017694 DOI: 10.1007/s00262-023-03436-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/22/2023] [Indexed: 04/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have fundamentally changed the treatment landscape of various cancers. While ICI treatments result in improved survival, quality of life and are cost-effective, the majority of patients experience at least one immune-related adverse event (irAE). Many of these side effects cause little discomfort or are asymptomatic; however, irAEs can affect any organ and are potentially life-threatening. Consequently, early diagnosis and appropriate treatment of irAEs are critical for optimizing long-term outcomes and quality of life in affected patients. Some irAEs are diagnosed according to typical symptoms, others by abnormal findings from diagnostic tests. While there are various guidelines addressing the management of irAEs, recommendations for the early recognition of irAEs as well as the optimal extent and frequency of laboratory tests are mostly lacking. In clinical practice, blood sampling is usually performed before each ICI administration (i.e., every 2-3 weeks), often for several months, representing a burden for patients as well as health care systems. In this report, we propose essential laboratory and functional tests to improve the early detection and management of irAEs and in cancer patients treated with ICIs. These multidisciplinary expert recommendations regarding essential laboratory and functional tests can be used to identify possible irAEs at an early time point, initiate appropriate interventions to improve patient outcomes, and reduce the burden of blood sampling during ICI treatment.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Cristina Espinosa da Silva
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
- Division of Epidemiology and Biostatistics, School of Public Health, San Diego State University, San Diego, USA
| | - Dimitri Arangalage
- Department of Cardiology, INSERM U1148, Bichat Hospital, University of Paris, Paris, France
| | - Pierre Monney
- Department of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sabina A Guler
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Guido Stirnimann
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lucia Possamai
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Roman Trepp
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism (UDEM), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Camille L Gerard
- Department of Oncology, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
- The Francis Crick Institute, London, UK
| | - Petr Hruz
- Department of Gastroenterology, University Hospital Basel, Basel, Switzerland
| | - Lisa Christ
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department Internal Medicine, Center for Oncology and Hematology, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
12
|
Cheng Y, Ouyang W, Jie C, Zhang J, Yu J, Xie C. A case of late and lethal Trousseau's syndrome induced by pembrolizumab in lung adenocarcinoma. Immunotherapy 2023; 15:71-75. [PMID: 36628567 DOI: 10.2217/imt-2022-0018] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Trousseau's syndrome is a relatively rare reported event in immunotherapy-related clinical trials, mostly occurring in the early period of immune checkpoint inhibitor (ICI) therapy. Here, we report an unusual case of late and lethal Trousseau's syndrome during pembrolizumab maintenance therapy in a lung adenocarcinoma harboring tumor protein p53 (TP53) mutation. The patient has experienced severe coagulation abnormalities manifesting as cerebral infarction, partial infarction of both kidneys and spleen after 23 cycles of pembrolizumab use and was resistant to anticoagulants. The late occurrence of coagulation abnormalities in this case reveals a possible correlation between TP53 mutations and Trousseau's syndrome when patients are treated with ICIs.
Collapse
Affiliation(s)
- Yajie Cheng
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Wen Ouyang
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Chen Jie
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Junhong Zhang
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Jing Yu
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| | - Conghua Xie
- Department of Radiation & Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, People's Republic of China.,Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China.,Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, 430071, China
| |
Collapse
|
13
|
Schlüter J, Cunningham S, Zimmermann R, Achenbach S, Kramer R, Erdmann M, Beckmann M, Heinzerling L, Hackstein H. Characterization of the impact of immune checkpoint inhibitors on platelet activation and aggregation. Immunobiology 2023; 228:152311. [PMID: 36495598 DOI: 10.1016/j.imbio.2022.152311] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/18/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Immune checkpoint inhibitors (ICIs) are effective oncological drugs which block cellular check-point receptors typically targeted by tumor immune evasion strategies. Despite their benefits, clinicians have reported treatment-associated thromboembolism during ICI therapy in recent years. Though several theories on this ICI-associated pathogenesis exist, the direct effects of ICIs on platelets remains unknown. We therefore investigated the potential direct and indirect effect of PD-1, PD-L1 and CTLA-4-targeting ICIs on platelet functionality in multifaceted in vitro experiments. Interestingly, we could not observe a clear effect of ICI on platelet aggregation and primary hemostasis in whole blood and platelet concentrate-based assays. Furthermore, the presence of ICIs in toll-like receptor stimulation had no significant impact on platelet surface marker expression. In a second approach, we investigated the indirect immunological impact of ICIs on platelet activation by exposing platelets to supernatants from ICI- and Staphylococcal enterotoxin B-exposed PBMCs. Whereas ICIs affected IL-2 levels in supernatants, we could not detect clear differences in the secretion of pro-thrombogenic factors and platelet responses. The obtained data suggest that the direct influence of ICIs on platelet activation or the influence of altered T cell function on platelet activation cannot be considered a major factor in the development of thrombotic events.
Collapse
Affiliation(s)
- Julian Schlüter
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Sarah Cunningham
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany.
| | - Robert Zimmermann
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Susanne Achenbach
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| | - Rafaela Kramer
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Michael Erdmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Malte Beckmann
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany
| | - Lucie Heinzerling
- Department of Gynecology and Obstetrics, University Hospital Erlangen, Erlangen 91054, Germany; Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich 80539, Germany
| | - Holger Hackstein
- Department of Transfusion Medicine and Hemostaseology, University Hospital Erlangen, Erlangen 91054, Germany
| |
Collapse
|
14
|
Li H, Li H, Tang L, Niu H, He L, Luo Q. Associations Between Immune-Related Venous Thromboembolism and Efficacy of Immune Checkpoint Inhibitors: A Systematic Review and Meta-Analysis. Clin Appl Thromb Hemost 2023; 29:10760296231206799. [PMID: 37844585 PMCID: PMC10586005 DOI: 10.1177/10760296231206799] [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: 08/17/2023] [Revised: 09/10/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023] Open
Abstract
This study aims to summarize the available data and determine if the presence of venous thromboembolism (VTE) immune-related adverse event (irAE) in patients with immune checkpoint inhibitor (ICI) therapy is associated with improved treatment efficacy and clinical outcomes, which in turn was used to help optimize patient selection for anticoagulation therapy and inform rational treatment strategies for overcoming the mechanisms of ICI resistance. PubMed, Embase, Web of Science, and Cochrane Library were searched up to March 18, 2023, for studies assessing the relationship between VTE irAE development during ICI therapy and cancer outcomes. Seven primary articles with a total of 4437 patients were included in the overall survival (OS) meta-analysis. Patients with VTE had a significant increase in overall mortality compared to patients without VTE in adjusted hazard ratios (HRs 1.36, 95% confidence interval [CI] 1.06-1.75, P = .02). In the studies where immortal time bias (ITB) was accounted for, patients with VTE irAE also had poor OS than those without. HR and the corresponding 95% CI values in the non-ITB group were 2.53 (1.75-3.66, P < .00001) with low heterogeneity (P = .17, I2 = 48%) and 1.21 (1.06-1.37, P = .004) in the ITB group with no heterogeneity (P = .95, I2 = 0%), respectively. Despite the heterogeneity identified, the evidence does suggest that VTE irAE occurrence could be served as a prognostic indicator, with higher frequencies of occurrence associated with poorer OS. However, the fundamental role of this association with clinical consequences should be further investigated in large cohorts and clinical trials.
Collapse
Affiliation(s)
- Huimin Li
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Hong Li
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Le Tang
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Haiwen Niu
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Lili He
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| | - Qin Luo
- Department of Respiratory and Neurology, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|
15
|
Kostoff RN, Briggs MB, Kanduc D, Dewanjee S, Kandimalla R, Shoenfeld Y, Porter AL, Tsatsakis A. Modifiable contributing factors to COVID-19: A comprehensive review. Food Chem Toxicol 2023; 171:113511. [PMID: 36450305 PMCID: PMC9701571 DOI: 10.1016/j.fct.2022.113511] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/29/2022]
Abstract
The devastating complications of coronavirus disease 2019 (COVID-19) result from an individual's dysfunctional immune response following the initial severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Multiple toxic stressors and behaviors contribute to underlying immune system dysfunction. SARS-CoV-2 exploits the dysfunctional immune system to trigger a chain of events ultimately leading to COVID-19. The current study identifies eighty immune system dysfunction-enabling toxic stressors and behaviors (hereafter called modifiable contributing factors (CFs)) that also link directly to COVID-19. Each CF is assigned to one of the five categories in the CF taxonomy shown in Section 3.3.: Lifestyle (e.g., diet, substance abuse); Iatrogenic (e.g., drugs, surgery); Biotoxins (e.g., micro-organisms, mycotoxins); Occupational/Environmental (e.g., heavy metals, pesticides); Psychosocial/Socioeconomic (e.g., chronic stress, lower education). The current study shows how each modifiable factor contributes to decreased immune system capability, increased inflammation and coagulation, and increased neural damage and neurodegeneration. It is unclear how real progress can be made in combatting COVID-19 and other similar diseases caused by viral variants without addressing and eliminating these modifiable CFs.
Collapse
Affiliation(s)
- Ronald Neil Kostoff
- Independent Consultant, Gainesville, VA, 20155, USA,Corresponding author. Independent Consultant, 13500 Tallyrand Way, Gainesville, VA, 20155, USA
| | | | - Darja Kanduc
- Dept. of Biosciences, Biotechnologies, and Biopharmaceutics, University of Bari, Via Orabona 4, Bari, 70125, Italy
| | - Saikat Dewanjee
- Advanced Pharmacognosy Research Laboratory, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, 700032, India
| | - Ramesh Kandimalla
- Applied Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad, 500007, Telangana, India
| | - Yehuda Shoenfeld
- Zabludowicz Center for Autoimmune Diseases, Sheba Medical Center, Tel-Hashomer, 5265601, Israel
| | - Alan L. Porter
- School of Public Policy, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Aristidis Tsatsakis
- Department of Forensic Sciences and Toxicology, Faculty of Medicine, University of Crete, 71003, Heraklion, Greece
| |
Collapse
|
16
|
Zhao L, Zheng H, Zhu L, Jiang P. Trousseau's syndrome in a non-small cell lung cancer patient aggravated by pembrolizumab. J Oncol Pharm Pract 2023; 29:233-238. [PMID: 35593090 DOI: 10.1177/10781552221102311] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
INTRODUCTION We present a case of Trousseau's syndrome in a non-small cell lung cancer patient recurrently aggravated by pembrolizumab. The adverse events related to immune checkpoint inhibitors (ICIs) on thrombogenesis remain unclear. CASE REPORT A 48-year-old woman was diagnosed with right lung adenocarcinoma (cT1bN3M1a, IVA) and with programmed cell death-1 positive. Brain magnetic resonance imaging (MRI) showed multiple asymptomatic bilateral cerebral infarctions as Trousseau's syndrome. After the patient was administered pembrolizumab, bilateral cerebral infarctions were aggravated. MANAGEMENT AND OUTCOME Although the patient was given prophylactic anticoagulant therapy respectively before two doses of pembrolizumab, Trousseau's syndrome still aggravated recurrently. DISCUSSION Trousseau's syndrome is rarely reported following the administration of ICIs. It is possible that pembrolizumab may trigger disorders of the coagulation-fibrinolysis system in cancer patients.
Collapse
Affiliation(s)
- Lihong Zhao
- Department of Respiratory and Critical Care Medicine, 66571Tianjin First Central Hospital, Tianjin, China
| | - Hong Zheng
- Department of Respiratory and Critical Care Medicine, 66571Tianjin First Central Hospital, Tianjin, China
| | - Liqin Zhu
- Department of Pharmacy, 66571Tianjin First Central Hospital, Tianjin, China
| | - Ping Jiang
- Department of Respiratory and Critical Care Medicine, 66571Tianjin First Central Hospital, Tianjin, China
| |
Collapse
|
17
|
Imamura K, Tomita Y, Sato R, Ikeda T, Iyama S, Jodai T, Takahashi M, Takaki A, Akaike K, Hamada S, Sakata S, Saruwatari K, Saeki S, Ikeda K, Suzuki M, Sakagami T. Clinical Implications and Molecular Characterization of Drebrin-Positive, Tumor-Infiltrating Exhausted T Cells in Lung Cancer. Int J Mol Sci 2022; 23:ijms232213723. [PMID: 36430217 PMCID: PMC9694580 DOI: 10.3390/ijms232213723] [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: 10/19/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022] Open
Abstract
T cells express an actin-binding protein, drebrin, which is recruited to the contact site between the T cells and antigen-presenting cells during the formation of immunological synapses. However, little is known about the clinical implications of drebrin-expressing, tumor-infiltrating lymphocytes (TILs). To address this issue, we evaluated 34 surgical specimens of pathological stage I-IIIA squamous cell lung cancer. The immune context of primary tumors was investigated using fluorescent multiplex immunohistochemistry. The high-speed scanning of whole-slide images was performed, and the tissue localization of TILs in the tumor cell nest and surrounding stroma was automatically profiled and quantified. Drebrin-expressing T cells were characterized using drebrin+ T cells induced in vitro and publicly available single-cell RNA sequence (scRNA-seq) database. Survival analysis using the propensity scores revealed that a high infiltration of drebrin+ TILs within the tumor cell nest was independently associated with short relapse-free survival and overall survival. Drebrin+ T cells induced in vitro co-expressed multiple exhaustion-associated molecules. The scRNA-seq analyses confirmed that the exhausted tumor-infiltrating CD8+ T cells specifically expressed drebrin. Our study suggests that drebrin-expressing T cells present an exhausted phenotype and that tumor-infiltrating drebrin+ T cells affect clinical outcomes in patients with resectable squamous cell lung cancer.
Collapse
Affiliation(s)
- Kosuke Imamura
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Yusuke Tomita
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
- Correspondence: ; Tel.: +81-96-373-5012; Fax: +81-96-373-5328
| | - Ryo Sato
- Laboratory of Stem Cell and Neuro-Vascular Biology, Cell and Developmental Biology Center, National Heart, Lung, and Blood Institute, National Institutes of Health, 10 Center Drive, Bethesda, MD 20892, USA
| | - Tokunori Ikeda
- Laboratory of Clinical Pharmacology and Therapeutics, Faculty of Pharmaceutical Sciences, Sojo University, 4-22-1, Ikeda Nishi-ku, Kumamoto-shi 860-0082, Kumamoto, Japan
| | - Shinji Iyama
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Takayuki Jodai
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Misako Takahashi
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Akira Takaki
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Kimitaka Akaike
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Shohei Hamada
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Shinya Sakata
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Koichi Saruwatari
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Sho Saeki
- Department of Respiratory Medicine, Kumamoto University Hospital, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Koei Ikeda
- Department of Thoracic and Breast Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Makoto Suzuki
- Department of Thoracic and Breast Surgery, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| | - Takuro Sakagami
- Department of Respiratory Medicine, Faculty of Life Sciences, Kumamoto University, Honjo 1-1-1, Chuo-ku, Kumamoto-shi 860-8556, Kumamoto, Japan
| |
Collapse
|
18
|
Langouo Fontsa M, Aiello MM, Migliori E, Scartozzi M, Lambertini M, Willard-Gallo K, Solinas C. Thromboembolism and Immune Checkpoint Blockade in Cancer Patients: An Old Foe for New Research. Target Oncol 2022; 17:497-505. [DOI: 10.1007/s11523-022-00908-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 12/19/2022]
|
19
|
Early diagnosis and bevacizumab combined with chemotherapy improved pulmonary tumor thrombotic microangiopathy with lung adenocarcinoma: A case report. CURRENT PROBLEMS IN CANCER: CASE REPORTS 2022. [DOI: 10.1016/j.cpccr.2022.100173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
20
|
Cánovas MS, Garay DF, Moran LO, Pérez JR, Rubio CMG, de Mena ML, Portero BO, Castro JB, Lage Y, Lavin DC, Blanco ABR, de Soignie AMMF, Perejón JZB, Colomo LJ, Boluda NB, Moreno JB, Verduguez TQ, Garrido CR, Huertas RM, Puig CFI, Martín AJM. Immune checkpoint inhibitors-associated thrombosis in patients with lung cancer and melanoma: a study of the Spanish society of medical oncology (SEOM) thrombosis and cancer group. Clin Transl Oncol 2022; 24:2010-2020. [PMID: 35668339 PMCID: PMC9418291 DOI: 10.1007/s12094-022-02860-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 05/12/2022] [Indexed: 12/03/2022]
Abstract
Purpose Immune Checkpoint Inhibitors (ICI) can be associated with thrombotic events, both venous and arterial (VTE/AT). However, there is a paucity of information regarding patients in routine clinical practice. Methods/patients Retrospective, multicenter study promoted by the Thrombosis and Cancer Section of the Spanish Society of Medical Oncology (SEOM). Patients with melanoma and lung cancer who initiated ICI between 01/01/2015 and 31/12/2019 were recruited. Minimum follow-up was 6 months (unless it was not possible because of death). The primary objective was to calculate the incidence of ICI-associated VTE/AT and the secondary objectives included to analyze its impact on survival and to identify predictor variables for VTE/AT. Results 665 patients with lung cancer were enrolled. The incidence of VTE/AT during follow-up was 8.4%. Median overall survival (OS) was lower in the VTE/AT group (12 months 95% CI 4.84–19.16 vs. 19 months 95% CI 16.11–21.9; p = 0.0049). Neutrophil/lymphocyte ratio (NLR) and anemia upon initiation of IT, as well as a history of thrombosis between cancer diagnosis and the start of ICI, were predictive variables for developing of VTE/AT (p < 0.05). 291 patients with melanoma were enrolled. There was a 5.8% incidence rate of VTE/AT during follow-up. Median OS was lower in the VTE/AT group (10 months 95% CI 0.0–20.27 vs. 29 months 95% CI 19.58–36.42; p = 0.034). NLR and lactate dehydrogenase (LDH) at the beginning of ICI were predictor variables for VTE/AT (p < 0.05). Conclusions ICI increases the risk of VTE/AT in patients with lung cancer and melanoma, which impact OS.
Collapse
Affiliation(s)
- Manuel Sánchez Cánovas
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain. .,Hematology and Medical Oncology Department, Hospital Universitario José María Morales Meseguer, Murcia, Spain.
| | - David Fernández Garay
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Complejo Hospitalario de Jaén, Jaén, Spain
| | - Laura Ortega Moran
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jaime Rubio Pérez
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Fundación Jiménez Díaz, Madrid, Spain
| | - Carlos Miguel Guirao Rubio
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario de Elche, Elche, Spain
| | - Miriam Lobo de Mena
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario de Valencia, Valencia, Spain
| | - Berta Obispo Portero
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Infanta Leonor, Madrid, Spain
| | - Jesús Brenes Castro
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Instituto Catalán de Oncología, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Yolanda Lage
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Diego Cacho Lavin
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Universitario Marqués de Valdecilla, Santander, Spain
| | - Ana Belen Rupérez Blanco
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Complejo Hospitalario de Toledo, Toledo, Spain
| | - Ana Manuela Martín Fernández de Soignie
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Universitario de Fuenlabrada, Fuenlabrada, Spain
| | - Jonatan Zacarías Benoit Perejón
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Complejo Hospitalario de Jaén, Jaén, Spain
| | - Laura Jiménez Colomo
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Instituto Catalán de Oncología, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Noel Blaya Boluda
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Hematology and Medical Oncology Department, Hospital Universitario José María Morales Meseguer, Murcia, Spain
| | - Javier Bosque Moreno
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario de Elche, Elche, Spain
| | - Teresa Quintanar Verduguez
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario de Elche, Elche, Spain
| | - Carmen Rosa Garrido
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Unidad de Investigación, Hospital Universitario de Jaén, FIBAO, Jaén, Spain
| | - Roberto Martín Huertas
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Carme Font I Puig
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Andrés Jesús Muñoz Martín
- Spanish Society of Medical Oncology (SEOM) Thrombosis and Cancer Group, Madrid, Spain.,Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| |
Collapse
|
21
|
Mechanisms underlying immune-related adverse events during checkpoint immunotherapy. Clin Sci (Lond) 2022; 136:771-785. [PMID: 35621125 DOI: 10.1042/cs20210042] [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: 01/18/2022] [Revised: 05/01/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022]
Abstract
Immune checkpoint (IC) proteins are some of the most important factors that tumor cells hijack to escape immune surveillance, and inhibiting ICs to enhance or relieve antitumor immunity has been proven efficient in tumor treatment. Immune checkpoint blockade (ICB) agents such as antibodies blocking programmed death (PD) 1, PD-1 ligand (PD-L) 1, and cytotoxic T lymphocyte-associated antigen (CTLA)-4 have been approved by the U.S. Food and Drug Administration (FDA) to treat several types of cancers. Although ICB agents have shown outstanding clinical success, and their application has continued to expand to additional tumor types in the past decade, immune-related adverse events (irAEs) have been observed in a wide range of patients who receive ICB treatment. Numerous studies have focused on the clinical manifestations and pathology of ICB-related irAEs, but the detailed mechanisms underlying irAEs remain largely unknown. Owing to the wide expression of IC molecules on distinct immune cell subpopulations and the fact that ICB agents generally affect IC-expressing cells, the influences of ICB agents on immune cells in irAEs need to be determined. Here, we discuss the expression and functions of IC proteins on distinct immune cells and the potential mechanism(s) related to ICB-targeted immune cell subsets in irAEs.
Collapse
|
22
|
Cantrell R, Palumbo JS. Hemostasis and tumor immunity. Res Pract Thromb Haemost 2022; 6:e12728. [PMID: 35647476 PMCID: PMC9130907 DOI: 10.1002/rth2.12728] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/19/2022] [Accepted: 05/01/2022] [Indexed: 12/13/2022] Open
Abstract
Significant data have accumulated demonstrating a reciprocal relationship between cancer and the hemostatic system whereby cancer promotes life‐threatening hemostatic system dysregulation (e.g., thromboembolism, consumptive coagulopathy), and hemostatic system components directly contribute to cancer pathogenesis. The mechanistic underpinnings of this relationship continue to be defined, but it is becoming increasingly clear that many of these mechanisms involve crosstalk between the hemostatic and immune systems. This is perhaps not surprising given that there is ample evidence for bidirectional crosstalk between the hemostatic and immune systems at multiple levels that likely evolved to coordinate the response to injury, host defense, and tissue repair. Much of the data linking hemostasis and immunity in cancer biology focus on innate immune system components. However, the advent of adaptive immunity‐based cancer therapies such as immune checkpoint inhibitors has revealed that the relationship of hemostasis and immunity in cancer extends to the adaptive immune system. Adaptive immunity‐based cancer therapies appear to be associated with an increased risk of thromboembolic complications, and hemostatic system components appear to regulate adaptive immune functions through diverse mechanisms to affect tumor progression. In this review, the evidence for crosstalk between hemostatic and adaptive immune system components is discussed, and the implications of this relationship in the context of cancer therapy are reviewed. A better understanding of these relationships will likely lead to strategies to make existing adaptive immune based therapies safer by decreasing thromboembolic risk and may also lead to novel targets to improve adaptive immune‐based cancer treatments.
Collapse
Affiliation(s)
- Rachel Cantrell
- Cancer and Blood Diseases Institute Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine Cincinnati Ohio USA
| | - Joseph S. Palumbo
- Cancer and Blood Diseases Institute Cincinnati Children's Hospital Medical Center and the University of Cincinnati College of Medicine Cincinnati Ohio USA
| |
Collapse
|
23
|
McCrae KR, Swaidani S, Diaz-Montero CM, Khorana AA. Old is new again: emergence of thromboembolic complications in cancer patients on immunotherapy. Thromb Res 2022; 213:S51-S57. [PMID: 36061419 PMCID: PMC9435305 DOI: 10.1016/j.thromres.2022.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cancer immunotherapy has emerged as one of the most important new treatments for cancer in many years, moving rapidly to front-line therapy for several cancers. Cancer immunotherapy is based on treatment with immune checkpoint inhibitors (ICI), which are monoclonal antibodies directed toward immunoregulatory proteins including PD-1, PD-L1 and CTLA-4. ICI inhibit interactions between these proteins and their ligands, disabling physiologic immune regulatory networks and enhancing anti-tumor immunity. However, since the immune response cannot be directed specifically to the tumor, ICI are associated with immune-related adverse events (irAEs) resulting from immune-mediated attack of normal tissues. We and others have reported a high incidence of thrombosis in patients treated with ICI, which may approach 20%. Given the rapidly increasing use of ICIs, it is clear that ICI-Associated Thrombosis (IAT) is a major emerging clinical problem. However, there is a remarkable knowledge gap concerning mechanisms of IAT. IAT may be a composite irAE resulting from activation of blood and vascular cells, leading to thromboinflammation. Cancer itself is an inflammatory disorder, and inducing further inflammation through ICI administration may stimulate procoagulant activity by multiple cell types. Moreover, some blood and vascular cells express ICI target proteins. Here, we review the results of several studies describing the clinical manifestations of IAT, as well as our recent studies demonstrating that elevated levels of myeloid derived suppressor cells and inflammatory cytokines may serve as biomarkers of IAT. It is hoped that the concepts reviewed here may stimulate further research into this important clinical problem.
Collapse
Affiliation(s)
- Keith R McCrae
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Shadi Swaidani
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - C Marcela Diaz-Montero
- Center for Immunotherapy & Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Alok A Khorana
- Department of Hematology & Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| |
Collapse
|
24
|
Singh AK, Malviya R. Coagulation and inflammation in cancer: Limitations and prospects for treatment. Biochim Biophys Acta Rev Cancer 2022; 1877:188727. [PMID: 35378243 DOI: 10.1016/j.bbcan.2022.188727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/26/2022] [Accepted: 03/29/2022] [Indexed: 02/08/2023]
Abstract
The development of so-called immune checkpoint inhibitors (ICIs), which target specific molecular processes of tumour growth, has had a transformative effect on cancer treatment. Widespread use of antibody-based medicines to inhibit tumour cell immune evasion by modulating T cell responses is becoming more common. Despite this, response rates are still low, and secondary resistance is an issue that arises often. In addition, a wide range of serious adverse effects is triggered by enhancing the immunological response. As a result of an increased mortality rate, a higher prevalence of thrombotic complications is connected with an increased incidence of immunological reactions, complement activation, and skin toxicity. This suggests that the tumour microenvironment's interaction between coagulation and inflammation is important at every stage of the tumour's life cycle. The coagulation system's function in tumour formation is the topic of this review. By better understanding the molecular mechanisms in which tumour cells circulate, plasmatic coagulation and immune system cells are engaged, new therapy options for cancer sufferers may be discovered.
Collapse
Affiliation(s)
- Arun Kumar Singh
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India
| | - Rishabha Malviya
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, Uttar Pradesh, India.
| |
Collapse
|
25
|
Goel A, Khorana A, Kartika T, Gowda S, Tao DL, Thawani R, Shatzel JJ. Assessing the risk of thromboembolism in cancer patients receiving immunotherapy. Eur J Haematol 2022; 108:271-277. [PMID: 34905252 PMCID: PMC9009190 DOI: 10.1111/ejh.13734] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 11/28/2022]
Abstract
Malignancy has long been implicated with hypercoagulability, leading to an increased rate of both venous and arterial thromboembolic events (VTE and ATE). Immunotherapy has established itself as a cornerstone of modern cancer therapy by promoting antitumor immune responses, though there have been some suggestions that immune-related adverse events could include increased rates of VTE and ATE. In this review, we examine the available evidence regarding the use of immune checkpoint inhibitors (ICIs) and thrombosis. First, we describe the potential mechanisms by which ICIs might lead to thrombophilia given the overlap between the immune system, coagulation cascade, and platelet adhesion and activation. In addition, while there are some preclinical data evaluating immunotherapy-associated ATEs in animal models, there is a paucity of evidence exploring potential mechanism of VTEs in ICIs. Second, we review the incidence of ATE and VTE in patients receiving ICIs in the published literature. Finally, we discuss current limitations in understanding, areas of conflicting evidence, and approaches to further investigation.
Collapse
Affiliation(s)
- Abhishek Goel
- University of California San Diego, San Diego, California, USA
| | | | - Thomas Kartika
- Division of Hematology and Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Sonia Gowda
- Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Derrick L. Tao
- Providence Portland Medical Center, Earle A. Chiles Research Institute, Portland, Oregon, USA
| | - Rajat Thawani
- Division of Hematology and Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
| | - Joseph J. Shatzel
- Division of Hematology and Oncology, Department of Medicine, Oregon Health & Science University, Portland, Oregon, USA
- Department of Biomedical Engineering, Oregon Health & Science University, Portland, Oregon, USA
| |
Collapse
|
26
|
Ronen D, Bsoul A, Lotem M, Abedat S, Yarkoni M, Amir O, Asleh R. Exploring the Mechanisms Underlying the Cardiotoxic Effects of Immune Checkpoint Inhibitor Therapies. Vaccines (Basel) 2022; 10:vaccines10040540. [PMID: 35455289 PMCID: PMC9031363 DOI: 10.3390/vaccines10040540] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Adaptive immune response modulation has taken a central position in cancer therapy in recent decades. Treatment with immune checkpoint inhibitors (ICIs) is now indicated in many cancer types with exceptional results. The two major inhibitory pathways involved are cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death protein 1 (PD-1). Unfortunately, immune activation is not tumor-specific, and as a result, most patients will experience some form of adverse reaction. Most immune-related adverse events (IRAEs) involve the skin and gastrointestinal (GI) tract; however, any organ can be involved. Cardiotoxicity ranges from arrhythmias to life-threatening myocarditis with very high mortality rates. To date, most treatments of ICI cardiotoxicity include immune suppression, which is also not cardiac-specific and may result in hampering of tumor clearance. Understanding the mechanisms behind immune activation in the heart is crucial for the development of specific treatments. Histological data and other models have shown mainly CD4 and CD8 infiltration during ICI-induced cardiotoxicity. Inhibition of CTLA4 seems to result in the proliferation of more diverse T0cell populations, some of which with autoantigen recognition. Inhibition of PD-1 interaction with PD ligand 1/2 (PD-L1/PD-L2) results in release from inhibition of exhausted self-recognizing T cells. However, CTLA4, PD-1, and their ligands are expressed on a wide range of cells, indicating a much more intricate mechanism. This is further complicated by the identification of multiple co-stimulatory and co-inhibitory signals, as well as the association of myocarditis with antibody-driven myasthenia gravis and myositis IRAEs. In this review, we focus on the recent advances in unraveling the complexity of the mechanisms driving ICI cardiotoxicity and discuss novel therapeutic strategies for directly targeting specific underlying mechanisms to reduce IRAEs and improve outcomes.
Collapse
Affiliation(s)
- Daniel Ronen
- Department of Internal Medicine D, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Aseel Bsoul
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Michal Lotem
- Department of Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Merav Yarkoni
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
- Correspondence: ; Tel.: +972-2-6776564; Fax: +972-2-6411028
| |
Collapse
|
27
|
Shijubou N, Sawai T, Hatakeyama T, Munakata S, Yamazoe M. Alveolar Hemorrhage Caused by the Combination of Immune Checkpoint Inhibitors (ICIs) and Angiogenesis Inhibitors: The Underlying Long-Term Vascular Endothelial Growth Factor (VEGF) Inhibition. Cureus 2022; 14:e23272. [PMID: 35449623 PMCID: PMC9012575 DOI: 10.7759/cureus.23272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 11/16/2022] Open
Abstract
The combination of immune checkpoint inhibitors (ICIs) and other anticancer agents is the standard of care for various cancers. Bevacizumab, an anti-angiogenesis inhibitor, causes serious adverse events such as pulmonary hemorrhage (PH). Here, we present a case of drug-induced diffuse alveolar hemorrhage (DAH), an adverse event, in a patient with hepatocellular carcinoma who was treated with a combination of ICIs and anti-angiogenesis inhibitors after long-term use of lenvatinib, which inhibits vascular endothelial growth factor (VEGF). An 85-year-old man with hepatocellular carcinoma initially received lenvatinib, a multi-kinase inhibitor, but the drug was later switched to bevacizumab-atezolizumab combination therapy owing to disease progression. After five cycles, he developed dyspnea and diffuse ground-glass opacities, which improved with discontinuation of the combination therapy and initiation of steroid pulse therapy. Our case findings indicate that both ICIs and anti-angiogenesis inhibitors cause drug-induced DAH, and their combination may increase the severity of DAH. Moreover, long-term VEGF inhibition may induce the development of DAH. Clinicians need to be aware that long-term VEGF inhibition may be associated with DAH and should consider the risk management of such adverse events while using this combination therapy.
Collapse
|
28
|
Batinac T, Hlača N, Simetić L, Valković F, Peternel S, Prpić-Massari L. Stevens-Johnson Syndrome and Severe Anaemia: A Case of Toxicity Induced by Vemurafenib plus Cobimetinib following Pembrolizumab for Metastatic Melanoma. Acta Derm Venereol 2022; 102:adv00650. [PMID: 35146529 PMCID: PMC9558335 DOI: 10.2340/actadv.v102.1433] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
| | | | | | | | - Sandra Peternel
- Department of Dermatovenereology, Clinical Hospital Center Rijeka, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, HR-51000, Rijeka, Croatia.
| | | |
Collapse
|
29
|
Bauer AT, Gorzelanny C, Gebhardt C, Pantel K, Schneider SW. Interplay between coagulation and inflammation in cancer: Limitations and therapeutic opportunities. Cancer Treat Rev 2022; 102:102322. [DOI: 10.1016/j.ctrv.2021.102322] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 11/24/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022]
|
30
|
Hashimoto M, Nakayama T, Fujimoto S, Inoguchi S, Nishimoto M, Kikuchi T, Adomi S, Banno E, De Velasco MA, Saito Y, Shimizu N, Mori Y, Minami T, Fujita K, Nozawa M, Nose K, Yoshimura K, Uemura H. Disseminated intravascular coagulation induced by pazopanib following combination therapy of nivolumab plus ipilimumab in a patient with metastatic renal cell carcinoma. Anticancer Drugs 2022; 33:e818-e821. [PMID: 34486537 DOI: 10.1097/cad.0000000000001230] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Recently, combination therapy including immune checkpoint inhibition (ICI) has proven to be effective as first-line therapy for patients with metastatic renal cell carcinoma. Although the first-line combination therapies with ICI have shown clinical benefit, a number of patients require second-line treatment. We report a 60-year-old man with metastatic renal cell carcinoma who was treated with pazopanib soon after nivolumab plus ipilimumab combination therapy. He experienced Grade 3 disseminated intravascular coagulation (DIC). We suspect that this was caused by an interaction between pazopanib and nivolumab even though ICI therapy was discontinued. He was treated with thrombomodulin and platelet transfusion and recovered from DIC. Treatment with pazopanib was subsequently restarted. No evidence of DIC was observed thereafter. This severe adverse reaction may have been induced by an interaction between activated proinflammatory immune cells and cytokines from an exacerbated inflammatory state and pazopanib. This report highlights the need to perform careful monitoring of patients who receive molecular targeted therapy after ICI-based immunotherapy.
Collapse
Affiliation(s)
- Mamoru Hashimoto
- Department of Urology, Kindai University Faculty of Medicine, Osaka, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Zou XL, Chen WY, Zhang GY, Ke H, Yang QH, Li XB. Risk Factors, Incidence, and Prognosis of Thromboembolism in Cancer Patients Treated With Immune Checkpoint Inhibitors. Front Pharmacol 2021; 12:747075. [PMID: 34819857 PMCID: PMC8606686 DOI: 10.3389/fphar.2021.747075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 10/15/2021] [Indexed: 11/16/2022] Open
Abstract
In recent years, immune checkpoint inhibitors (ICIs) have become the standard treatment option for tumors. With the widespread application of ICIs, immune-related adverse events (irAEs) have gradually attracted the attention of researchers. Owing to the characteristics of ICIs, irAEs can affect each organ of the human body. Thromboembolism is uncommon in cancer patients receiving ICIs, but it may affect their survival. Most thromboembolic events do not cause serious effects after early prediction and treatment, but life-threatening toxic reactions are also observed. This condition should not be ignored because of vague and atypical symptoms, which make early diagnosis more challenging. This article focuses on the high-risk factors, underlying mechanisms, incidence, and prognosis of thromboembolism in patients using ICIs and briefly describes the intervention and treatment measures. This information would allow patients to effectively manage the side effects of thromboembolism during Immune checkpoint inhibitors treatment, ensuring the efficacy of ICIs and reducing mortality.
Collapse
Affiliation(s)
- Xue-Lin Zou
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| | - Wei-Yong Chen
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| | - Guang-Yan Zhang
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| | - Hua Ke
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| | - Qiu-Hong Yang
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| | - Xiao-Bo Li
- Department of Respiratory Medicine, Chengdu Seventh People's Hospital, Chengdu, China
| |
Collapse
|
32
|
Zhang F, Qiu B, Ji Y, Guo W, Li N, Xue Q, Gao S, He J. Comparison of surgical difficulty in patients with resectable non-small cell lung cancer under different neoadjuvant treatment modes: a retrospective cohort study. J Thorac Dis 2021; 13:5604-5616. [PMID: 34795911 PMCID: PMC8575808 DOI: 10.21037/jtd-21-1007] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/10/2021] [Indexed: 12/29/2022]
Abstract
Background Previous studies have reported on the efficacy and safety of neoadjuvant use of a programmed cell death 1 (PD-1) antibody, sintilimab, in patients with non-small cell lung cancer (NSCLC). This study aimed to further evaluate the difficulty of this surgery and the postoperative complication rates in patients with NSCLC receiving neoadjuvant sintilimab. Methods Patients who received neoadjuvant sintilimab (200 mg) in the Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital from March 2018 to March 2019 were enrolled in the neoadjuvant immunotherapy group (NI group). Another two cohorts who did not receive sintilimab were retrospectively selected by propensity score matching (PSM) at a ratio of 1:1 in the upfront surgery (M-US) and neoadjuvant chemotherapy (M-NC) groups. The postoperative complication rate, postoperative days (PODs), and other detailed objective indicators were compared by t-test or χ2 test. Results Thirty-seven patients were enrolled in each group. Postoperative complications were greater in the NI group (37.8%) than in the M-US (10.8%; P=0.013) or in the M-NC group (16.2%; P=0.036). The number of PODs (7) was greater in the NI group than in the M-US group (P=0.005). The total number of dissected lymph nodes was lower in the NI group than in the M-US group (P<0.001) or in the M-NC group (P<0.001). Lymph node dissection (LND) in the NI group was more difficult than in the M-US group (P=0.015), but intrathoracic adhesion, tumor invasion, and whole procedure difficulty were similar. Conclusions The administration of neoadjuvant sintilimab increased complications but did not increase the difficulty of surgery. Fewer lymph nodes were dissected in the NI group.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Qiu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Ji
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
33
|
Sütcüoğlu O, İlhan A, Tacar SY, Güven DC, Uçar G, Karadurmuş N, Yıldız F, Eraslan E, Uncu D, Tural D, Üner A, Günel N, Özdemir N, Kılıçkap S, Öksüzoğlu ÖB, Özet A, Yazıcı O. Thirty-day mortality rates after immunotherapy initiation. Immunotherapy 2021; 13:1419-1426. [PMID: 34676791 DOI: 10.2217/imt-2021-0082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Background: The aim of this study was to determine the cause of death in patients who died within 30 days after the first dose of immunotherapy. Methods: The data of 1432 patients treated with immunotherapy in six tertiary referral hospitals were retrospectively analyzed. Results: It was determined that 34 (2%) of the patients died within 30 days after the first dose of immunotherapy. Death occurred in all patients who received palliative therapy, and most patients (88%) received immunotherapy as second- or subsequent-line of therapy. The most common cause of death was disease progression and thromboembolic events. Conclusion: Preliminary results of the current study might give some clues to define the patient population in whom the fatal side effects of immunotherapy might be encountered.
Collapse
Affiliation(s)
- Osman Sütcüoğlu
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| | - Ayşegül İlhan
- Dr. Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Seher Yıldız Tacar
- Dr. Sadi Konuk Training & Research Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Deniz Can Güven
- Hacettepe University Cancer Institute, Department of Medical Oncology, Ankara, Turkey
| | - Gökhan Uçar
- Health of Science Ankara City Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Nuri Karadurmuş
- Health Sciences University Gulhane Training & Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Fatih Yıldız
- Dr. Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Emrah Eraslan
- Dr. Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Doğan Uncu
- Health of Science Ankara City Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Deniz Tural
- Dr. Sadi Konuk Training & Research Hospital, Department of Medical Oncology, Istanbul, Turkey
| | - Aytuğ Üner
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| | - Nazan Günel
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| | - Nuriye Özdemir
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| | - Saadettin Kılıçkap
- Hacettepe University Cancer Institute, Department of Medical Oncology, Ankara, Turkey
| | - Ömür Berna Öksüzoğlu
- Dr. Abdurrahman Yurtaslan Ankara Oncology Training & Research Hospital, Department of Medical Oncology, Ankara, Turkey
| | - Ahmet Özet
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| | - Ozan Yazıcı
- Gazi University, Department of Medical Oncology, Ankara, Turkey
| |
Collapse
|
34
|
A brief report on incidence, radiographic feature and prognostic significance of brain MRI changes after anti-PD-1/PD-L1 therapy in advanced non-small cell lung cancer. Cancer Immunol Immunother 2021; 71:1275-1280. [PMID: 34613418 DOI: 10.1007/s00262-021-03070-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Accepted: 09/26/2021] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Neurologic immune-related adverse events (nirAEs) are uncommon but potentially lethal complications of immune checkpoint inhibitor (ICI) treatment. However, the incidence, radiographic features and prognostic significance of brain magnetic resonance imaging (MRI) changes after ICI treatment remain largely unknown. METHODS Consecutive patients with advanced non-small cell lung cancer (NSCLC) at three participating institutions receiving anti-PD-1/PD-L1 therapy from June 2017 to September 2020 were screened, and those who received brain MRI within 6 weeks before ICI initiation and at least one follow-up brain MRI after ICI treatment were included. Serial brain MRI images were independently reviewed by two experienced radiologists. RESULTS With a median follow-up of 13.2 months, 27 (20.0%) of the 135 enrolled patients developed certain kind of brain MRI aberration. The 1-, 2- and 3-year cumulative incidence of brain MRI aberration was 17.1%, 36.3% and 52.2%, respectively. Brain MRI aberration indicative of stroke, mimicking typical white matter lesions and presenting as T2-hyperintensity suggestive of CNS vasculitis or encephalitis, was documented in 11, 9 and 4 patients, respectively. Patients with brain MRI aberration had higher clinical benefit rate (p = 0.030), longer progression-free survival (p = 0.015) and a tendency of improved overall survival (p = 0.054). CONCLUSIONS Brain MRI aberrations developed after ICI treatment are not uncommon, and their manifestations vary a lot. Patients developing brain MRI aberrations tended to have better prognosis, which needed to be further investigated.
Collapse
|
35
|
Life-threatening polymyositis with spontaneous hematoma induced by nivolumab in a patient with previously resected melanoma. Melanoma Res 2021; 31:85-87. [PMID: 33196530 DOI: 10.1097/cmr.0000000000000706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Single-agent anti-PD1 antibodies are usually very well tolerated, but serious toxicity can still occur. Despite the PD-1 pathway seems to be relevant in the pathogenesis of immune-related myositis, anti-PD1-related myositis is generally a rare side effect of the treatment and usually not serious. However, its frequency is likely to increase as the use of immune checkpoint blockades. We present here a case of life-threatening polymyositis with associated spontaneous muscular hematoma in a patient treated with single-agent nivolumab in the adjuvant setting. Spontaneous hematoma is an extremely rare complication with unclear etiology of idiopathic myositis. Very few cases have been reported in the literature and their outcome has been often fatal. To our knowledge, this is the first case of autoimmune myositis and spontaneous heamatoma associated with the administration of single-agent checkpoint blockade. Anti-PD1 antibodies have changed the treatment landscape for a number of cancer entities in the past few years. When given as single agent they are usually very well tolerated, but serious rare toxicity can still occur. We present here a case of polymyositis with associated spontaneous muscular hematoma in a patient treated with single agent nivolumab.
Collapse
|
36
|
Checkpoint inhibitors and thrombosis: what's up? Blood 2021; 137:1569-1570. [PMID: 33764429 DOI: 10.1182/blood.2020009480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
37
|
Yan AR, Samarawickrema I, Naunton M, Peterson GM, Yip D, De Rosa S, Mortazavi R. Risk Factors and Prediction Models for Venous Thromboembolism in Ambulatory Patients with Lung Cancer. Healthcare (Basel) 2021; 9:778. [PMID: 34205695 PMCID: PMC8233898 DOI: 10.3390/healthcare9060778] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 12/21/2022] Open
Abstract
Venous thromboembolism (VTE) is a significant cause of mortality in patients with lung cancer. Despite the availability of a wide range of anticoagulants to help prevent thrombosis, thromboprophylaxis in ambulatory patients is a challenge due to its associated risk of haemorrhage. As a result, anticoagulation is only recommended in patients with a relatively high risk of VTE. Efforts have been made to develop predictive models for VTE risk assessment in cancer patients, but the availability of a reliable predictive model for ambulate patients with lung cancer is unclear. We have analysed the latest information on this topic, with a focus on the lung cancer-related risk factors for VTE, and risk prediction models developed and validated in this group of patients. The existing risk models, such as the Khorana score, the PROTECHT score and the CONKO score, have shown poor performance in external validations, failing to identify many high-risk individuals. Some of the newly developed and updated models may be promising, but their further validation is needed.
Collapse
Affiliation(s)
- Ann-Rong Yan
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra 2617, Australia; (A.-R.Y.); (M.N.); (G.M.P.); (D.Y.)
| | - Indira Samarawickrema
- School of Nursing, Midwifery and Public Health, Faculty of Health, University of Canberra, Canberra 2617, Australia;
| | - Mark Naunton
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra 2617, Australia; (A.-R.Y.); (M.N.); (G.M.P.); (D.Y.)
| | - Gregory M. Peterson
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra 2617, Australia; (A.-R.Y.); (M.N.); (G.M.P.); (D.Y.)
- College of Health and Medicine, University of Tasmania, Hobart 7005, Australia
| | - Desmond Yip
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra 2617, Australia; (A.-R.Y.); (M.N.); (G.M.P.); (D.Y.)
- Department of Medical Oncology, The Canberra Hospital, Garran 2605, Australia
- ANU Medical School, Australian National University, Canberra 0200, Australia
| | - Salvatore De Rosa
- Department of Medical and Surgical Science, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Reza Mortazavi
- School of Health Sciences, Faculty of Health, University of Canberra, Canberra 2617, Australia; (A.-R.Y.); (M.N.); (G.M.P.); (D.Y.)
- Prehab Activity Cancer Exercise Survivorship Research Group, Faculty of Health, University of Canberra, Canberra 2617, Australia
| |
Collapse
|
38
|
Ho AK, Ho AMH, Cooksley T, Nguyen G, Erb J, Mizubuti GB. Immune-Related Adverse Events Associated With Immune Checkpoint Inhibitor Therapy. Anesth Analg 2021; 132:374-383. [PMID: 33009134 DOI: 10.1213/ane.0000000000005029] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
As part of immune surveillance, killer T lymphocytes search for cancer cells and destroy them. Some cancer cells, however, develop escape mechanisms to evade detection and destruction. One of these mechanisms is the expression of cell surface proteins which allow the cancer cell to bind to proteins on T cells called checkpoints to switch off and effectively evade T-cell-mediated destruction. Immune checkpoint inhibitors (ICIs) are antibodies that block the binding of cancer cell proteins to T-cell checkpoints, preventing the T-cell response from being turned off by cancer cells and enabling killer T cells to attack. In other words, ICIs restore innate antitumor immunity, as opposed to traditional chemotherapies that directly kill cancer cells. Given their relatively excellent risk-benefit ratio when compared to other forms of cancer treatment modalities, ICIs are now becoming ubiquitous and have revolutionized the treatment of many types of cancer. Indeed, the prognosis of some patients is so much improved that the threshold for admission for intensive care should be adjusted accordingly. Nevertheless, by modulating immune checkpoint activity, ICIs can disrupt the intricate homeostasis between inhibition and stimulation of immune response, leading to decreased immune self-tolerance and, ultimately, autoimmune complications. These immune-related adverse events (IRAEs) may virtually affect all body systems. Multiple IRAEs are common and may range from mild to life-threatening. Management requires a multidisciplinary approach and consists mainly of immunosuppression, cessation or postponement of ICI treatment, and supportive therapy, which may require surgical intervention and/or intensive care. We herein review the current literature surrounding IRAEs of interest to anesthesiologists and intensivists. With proper care, fatality (0.3%-1.3%) is rare.
Collapse
Affiliation(s)
- Adrienne K Ho
- From the Department of Clinical Oncology, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Anthony M-H Ho
- Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Tim Cooksley
- Department of Acute Medicine and Critical Care, Manchester University NHS Foundation Trust, Manchester, United Kingdom
| | - Giang Nguyen
- Department of Radiology, Queen's University, Kingston, Ontario, Canada
| | - Jason Erb
- Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| | - Glenio B Mizubuti
- Department of Anesthesiology and Perioperative Medicine, Queen's University, Kingston, Ontario, Canada
| |
Collapse
|
39
|
Sussman TA, Li H, Hobbs B, Funchain P, McCrae KR, Khorana AA. Incidence of thromboembolism in patients with melanoma on immune checkpoint inhibitor therapy and its adverse association with survival. J Immunother Cancer 2021; 9:jitc-2020-001719. [PMID: 33436486 PMCID: PMC7805375 DOI: 10.1136/jitc-2020-001719] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2020] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Thromboembolism (TE) in cancer significantly contributes to morbidity and mortality. Little is known about the incidence of arterial TE (ATE) and venous TE (VTE) in patients with melanoma on immune checkpoint inhibitor (ICI) therapy. METHODS We conducted a retrospective cohort study of patients with melanoma receiving ICI from July 2015 through December 2017 at the Cleveland Clinic. TE, including VTE events of deep venous thrombosis, pulmonary embolism, visceral vein thrombosis, and ATE events of myocardial infarction, stroke, peripheral arterial embolism, or transient ischemic attack after ICI initiation were identified. Overall survival (OS) from ICI initiation was estimated by Kaplan-Meier and Cox hazard models; associations between TE, ICI regimen, and clinical risk factors were evaluated using log-rank test. RESULTS The study population comprised 228 patients with median age of 65 years (23-91 years), 67% male, and median follow-up of 27.3 months. Pembrolizumab was most commonly used (38.7%), followed by combination of ipilimumab plus nivolumab (29.4%), ipilimumab (20%), and nivolumab (12.3%). Most had stage IV disease (81.1%) and 11% had brain metastases (BM) at treatment initiation. Fifty-one TE events occurred in 47 patients (20.6%), including 37 (16.2%) VTE and 14 (6.1%) ATE. Cumulative incidence of TE after ICI initiation was 9.3% (95% CI: 6.0% to 13.6%) at 6 months, and 16.0% (95% CI: 11.6% to 21.2%) at 12 months. The 6-month and 12-month VTE cumulative incidence rates were higher with combination ICI than single agent (16.7% vs 5.0% and 21.3% vs 9.5%, respectively; p=0.02). Risk factors significantly associated with VTE in multivariate analysis included combination ICI (HR 2.70; 95% CI: 1.28 to 5.70; p=0.009), Khorana Score ≥1 (HR 2.24; 95% CI: 1.06 to 4.74; p=0.03), history of coronary artery disease (HR 2.71; 95% CI: 1.16 to 6.29); p=0.02), and anticoagulation at treatment start (HR 4.14; 95% CI: 1.60 to 10.7; p=0.003). Of patients without BM, OS was worse in patients with TE compared with those without (2-year OS 50.8% vs 71.3%; HR 2.27; 95% CI: 1.36 to 3.79; p=0.002), when adjusted for age and stage. CONCLUSIONS ICI is associated with a high incidence of TE in patients with melanoma, with higher rates with combination therapy; TE is associated with substantial worsening of survival. Further studies are needed to identify pathophysiology, biomarkers, and preventive approaches.
Collapse
Affiliation(s)
- Tamara A Sussman
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Hong Li
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Brian Hobbs
- Department of Population Health, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Pauline Funchain
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Keith R McCrae
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| | - Alok A Khorana
- Department of Hematology and Oncology, Taussig Cancer Institute, Cleveland Clinic Foundation, Cleveland, Ohio, USA
| |
Collapse
|
40
|
Chen J, Alduais Y, Chen B. Therapeutic and Systemic Adverse Events of Immune Checkpoint Inhibitors Targeting the PD-1/PD-L1 axis for Clinical Management of NSCLC. Cell Transplant 2021; 30:9636897211041587. [PMID: 34606729 PMCID: PMC8493325 DOI: 10.1177/09636897211041587] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/14/2022] Open
Abstract
Non-small-cell lung cancer takes up the majority of lung carcinoma-caused deaths. It is reported that targeting PD-1/PD-L1, a well-known immune evasion checkpoint, can eradicate tumor. Checkpoint inhibitors, such as monoclonal antibodies, are actively employed in cancer treatment. Thus, this review aimed to assess the therapeutic and toxic effects of PD-1/PD-L1 inhibitors in treatment of NSCLC. So far, 6 monoclonal antibodies blocking PD-1/PD-L1 interaction are identified and used in clinical trials and randomized controlled trials for NSCLC therapy. These antibody-based therapies for NSCLC were collected by using search engine PubMed, and articles about the assessment of adverse events were collected by using Google search. Route of administration and dosage are critical parameters for efficient immunotherapy. Although antibodies can improve overall survival and are expected to be target-specific, they can cause systemic adverse effects in the host. Targeting certain biomarkers can limit the toxicity of adverse effects of the antibody-mediated therapy. Clinical experts with knowledge of adverse effects (AEs) of checkpoint inhibitors can help manage and reduce mortalities associated with antibody-based therapy of NSCLC.
Collapse
Affiliation(s)
- Jing Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Yaser Alduais
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| | - Baoan Chen
- Department of Hematology and Oncology, Zhongda Hospital, School of Medicine, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing, 210009, Jiangsu, China
| |
Collapse
|
41
|
Nakano Y, Koide N, Koyama Y, Nitta K, Koizumi T. Lung adenocarcinoma initially presenting as Trousseau's syndrome treated successfully with pembrolizumab: A case report. Thorac Cancer 2020; 12:557-559. [PMID: 33350050 PMCID: PMC7882382 DOI: 10.1111/1759-7714.13794] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 12/05/2020] [Accepted: 12/07/2020] [Indexed: 11/28/2022] Open
Abstract
A 60-year-old woman was urgently admitted to our hospital because of vertigo and left hemiplegia. Laboratory examination showed thrombocytopenia, high levels of D-dimer and carcinoembryonic antigen. Brain magnetic resonance imaging (MRI) revealed multiple bilateral cerebral infarctions. Chest computed tomography (CT) showed an irregularly shaped tumor in the upper lobe of the left lung and mediastinal node swelling. The histopathological findings revealed adenocarcinoma negative for anaplastic lymphoma kinase fusion gene, sensitive epidermal growth factor receptor mutations. A diagnosis of lung adenocarcinoma initially presenting as arterial thromboembolism was made, and she was treated with direct oral anticoagulant (DOAC). Subsequently, pembrolizumab therapy was initiated because tumor cells were positive for programmed cell death protein 1 (PD-L1;60%), and resulted in reduction of the tumor with normalization of the platelet count and d-dimer. The treatment has been continued for over one year without any recurrence of the disease or thromboembolism.
Collapse
Affiliation(s)
- Yuta Nakano
- Department of Surgery, Nagano Prefectural Kiso Hospital, Nagano, Japan
| | - Naohiko Koide
- Department of Surgery, Nagano Prefectural Kiso Hospital, Nagano, Japan
| | - Yoshinori Koyama
- Department of Surgery, Nagano Prefectural Kiso Hospital, Nagano, Japan
| | - Kazuhito Nitta
- Department of Neurology, Nagano Prefectural Kiso Hospital, Nagano, Japan
| | - Tomonobu Koizumi
- Department of Comprehensive Cancer Therapy, Shinshu University School of Medicine, Matsumoto, Japan
| |
Collapse
|
42
|
Wu Z, Man S, Sun R, Li Z, Wu Y, Zuo D. Recent advances and challenges of immune checkpoint inhibitors in immunotherapy of non-small cell lung cancer. Int Immunopharmacol 2020; 85:106613. [DOI: 10.1016/j.intimp.2020.106613] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
|
43
|
Shannon VR, Subudhi SK, Huo L, Faiz SA. Diffuse alveolar hemorrhage with nivolumab monotherapy. Respir Med Case Rep 2020; 30:101131. [PMID: 32577370 PMCID: PMC7303994 DOI: 10.1016/j.rmcr.2020.101131] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 02/06/2023] Open
Abstract
We describe a 60 year old man who developed respiratory insufficiency after treatment with 2 rounds of nivolumab monotherapy. Imaging revealed subtle ground glass infiltrates which progressed to diffuse opacities and consolidation. The patient was treated with high dose corticosteroids, empiric antimicrobial therapy and infliximab. Bronchoscopy with lavage revealed negative cultures and progressive bloody aliquots of fluid consistent with diffuse alveolar hemorrhage. The patient succumbed to respiratory failure. An autopsy study confirmed extensive alveolar hemorrhage. Our reports highlights clinical and diagnostic findings with immunotherapy-induced pneumonitis.
Collapse
Affiliation(s)
- Vickie R Shannon
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Sumit K Subudhi
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Lei Huo
- Department of Anatomical Pathology, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - Saadia A Faiz
- Department of Pulmonary Medicine, The University of Texas MD Anderson Cancer Center, Houston, USA
| |
Collapse
|
44
|
Alberti A, Mancin M, Cortinovis D, Bidoli P, Sala L. Disseminated intravascular coagulation in advanced lung adenocarcinoma during first-line pembrolizumab. Immunotherapy 2020; 12:629-633. [DOI: 10.2217/imt-2020-0018] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Aim: Every year 1.6 million people worldwide die from lung cancer, making it one of the most frequent and deadly tumors. Pembrolizumab is a humanized monoclonal antibody against PD-1 that has antitumor activity in advanced non-small-cell lung cancer, with increased activity in tumors that express programmed death ligand 1. Methods & results: We report the first case of pembrolizumab-related disseminated intravascular coagulation (DIC). After excluding other causes of DIC, a diagnosis of pembrolizumab-related DIC was performed and the patient was treated with corticosteroid therapy until signs resolution. Conclusion: Disorders of coagulation–fibrinolysis system related to immunotherapy are rare, but often clinically serious and life threatening, so it is necessary to pay close attention to the various symptoms and signs during immunotherapy.
Collapse
Affiliation(s)
- Andrea Alberti
- Department of Oncology, San Gerardo Hospital, Asst-Monza, via Pergolesi 33 Monza, Italy
| | - Maddalena Mancin
- Department of Oncology, San Gerardo Hospital, Asst-Monza, via Pergolesi 33 Monza, Italy
| | - Diego Cortinovis
- Department of Oncology, San Gerardo Hospital, Asst-Monza, via Pergolesi 33 Monza, Italy
| | - Paolo Bidoli
- Department of Oncology, San Gerardo Hospital, Asst-Monza, via Pergolesi 33 Monza, Italy
| | - Luca Sala
- Department of Oncology, San Gerardo Hospital, Asst-Monza, via Pergolesi 33 Monza, Italy
| |
Collapse
|
45
|
Saleh R, Toor SM, Taha RZ, Al-Ali D, Sasidharan Nair V, Elkord E. DNA methylation in the promoters of PD-L1, MMP9, ARG1, galectin-9, TIM-3, VISTA and TGF-β genes in HLA-DR - myeloid cells, compared with HLA-DR + antigen-presenting cells. Epigenetics 2020; 15:1275-1288. [PMID: 32419601 PMCID: PMC7678924 DOI: 10.1080/15592294.2020.1767373] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Myeloid cells, including antigen-presenting cells (APCs) and myeloid-derived suppressor cells (MDSCs) play opposing roles to orchestrate innate and adaptive immune responses during physiological and pathological conditions. We investigated the role of DNA methylation in regulating the transcription of inhibitory/suppressive molecules in myeloid suppressive cells (identified as CD33+HLA-DR-) in comparison to APCs. We selected a number of immune checkpoints (ICs), IC ligands, and immunosuppressive molecules that have been implicated in MDSC function, including PD-L1, TIM-3, VISTA, galectin-9, TGF-β, ARG1 and MMP9. We examined their mRNA expression levels, and investigated whether DNA methylation regulates their transcription in sorted myeloid cell subpopulations. We found that mRNA levels of PD-L1, TIM-3, TGF-β, ARG1 and MMP9 in CD33+HLA-DR- cells were higher than APCs. However, VISTA and galectin-9 mRNA levels were relatively similar in both myeloid subpopulations. CpG islands in the promoter regions of TGF-β1, TIM-3 and ARG1 were highly unmethylated in CD33+HLA-DR-cells, compared with APCs, suggesting that DNA methylation is one of the key mechanisms, which regulate their expression. However, we did not find differences in the methylation status of PD-L1 and MMP9 between CD33+HLA-DR- and APCs, suggesting that their transcription could be regulated via other genetic and epigenetic mechanisms. The promoter methylation status of VISTA was relatively similar in both myeloid subpopulations. This study provides novel insights into the epigenetic mechanisms, which control the expression of inhibitory/suppressive molecules in circulating CD33+HLA-DR- cells in a steady-state condition, possibly to maintain immune tolerance and haemostasis.
Collapse
Affiliation(s)
- Reem Saleh
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Salman M Toor
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Rowaida Z Taha
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | | | - Varun Sasidharan Nair
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| | - Eyad Elkord
- Cancer Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF) , Doha, Qatar
| |
Collapse
|
46
|
Joseph JJ, Rajan A, Gulley JL, Ito S, Kessler CM. Acquired Coagulopathy With Immune Checkpoint Inhibitors: An Underrecognized Association Between Inflammation and Coagulation. JTO Clin Res Rep 2020; 1:100049. [PMID: 34589945 PMCID: PMC8474288 DOI: 10.1016/j.jtocrr.2020.100049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/22/2020] [Accepted: 04/24/2020] [Indexed: 01/14/2023] Open
Abstract
Introduction Immune-related adverse events affecting virtually every organ system have been described in individuals receiving immune checkpoint inhibitors. The spectrum of hematologic adverse effects is diverse and includes autoimmune cytopenias, hemolysis, or inhibition of coagulation factors. The interplay of inflammation and the coagulation cascade is complex, and immune checkpoint inhibitors can induce coagulopathy by disrupting the intricate link between these pathways. Methods We report acquired coagulopathy in two patients treated with the programmed death-ligand 1 antibodies, atezolizumab and avelumab, respectively. Clinical findings and results of extensive laboratory workup are reported. We hypothesize that cytokine release is a potential pathologic mechanism responsible for acquired coagulopathy. Results Symptoms included fever, fatigue, and disorientation in one patient and fever, myalgias, and skin rash in the other. Laboratory features included an abnormal coagulation profile; low fibrinogen levels; and elevated D-dimer, ferritin, and triglycerides. Treatment consisted of intravenous glucocorticoids in both cases and the use of fresh frozen plasma, cryoprecipitate, and clotting factor support in one patient. Conclusions Recognition of acquired coagulopathy as a complication of immunotherapy and its aggressive management are crucial to reduce morbidity and mortality associated with this condition.
Collapse
Affiliation(s)
- Jacinth J Joseph
- Cellular Therapy Branch, National Heart, Lung, and Blood Institutes, Bethesda, Maryland.,Department of Hematology/Oncology, Medstar Washington Hospital Center, Georgetown University, Washington, District of Columbia
| | - Arun Rajan
- Thoracic and Gastrointestinal Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - James L Gulley
- Genitourinary Malignancies Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Sawa Ito
- Hematology and Oncology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland
| | - Craig M Kessler
- Division of Hematology/Oncology, Lombardi Comprehensive Cancer Center, MedStar Georgetown University Hospital, Washington, District of Columbia
| |
Collapse
|
47
|
Kuriyama H, Fukushima S, Nakahara S, Kanemaru H, Miyashita A, Aoi J, Tomita Y, Kawasaki T, Nosaka K, Ihn H. Serious disseminated intravascular coagulation associated with combination therapy of nivolumab and ipilimumab in advanced melanoma. J Dermatol 2020; 47:e235-e237. [PMID: 32275077 DOI: 10.1111/1346-8138.15336] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Affiliation(s)
- Haruka Kuriyama
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Fukushima
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Satoshi Nakahara
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hisashi Kanemaru
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Azusa Miyashita
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Jun Aoi
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yusuke Tomita
- Department of, Respiratory Medicine, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Kawasaki
- Department of, Gastroenterology and Hepatology, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Kisato Nosaka
- Department of, Hematology, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Hironobu Ihn
- Departments of, Department of, Dermatology and Plastic Surgery, Graduate Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
48
|
Ren Q, Zhu P, Zhang H, Ye T, Liu D, Gong Z, Xia X. Identification and validation of stromal-tumor microenvironment-based subtypes tightly associated with PD-1/PD-L1 immunotherapy and outcomes in patients with gastric cancer. Cancer Cell Int 2020; 20:92. [PMID: 32226313 PMCID: PMC7092673 DOI: 10.1186/s12935-020-01173-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Accepted: 03/13/2020] [Indexed: 12/13/2022] Open
Abstract
Background Immunotherapies targeting programmed cell death 1 (PD-1) and programmed death-ligand 1 (PD-L1) have been approved for gastric cancer (GC) patients. However, a large proportion of patients with T-cell-inflamed tumor microenvironment do not respond to the PD-1/PD-L1 blockade. The stromal component of the tumor microenvironment has been associated with immunotherapy. This study aims to explore the clinical significance of the non-immune cells in the tumor microenvironment and their potential as biomarkers for immunotherapy. Methods A total of 383 patients with GC from the Cancer Genome Atlas (TCGA) cohort, 300 patients with GC from the GSE62254 cohort in Gene Expression Omnibus (GEO) were included in the study. A stromal score was generated using the ESTIMATE algorithm, and the likelihood of response to PD-1/PD-L1 immunotherapy of GC patients was predicted using the TIDE algorithm. The prognostic value of the stromal score from GC cases was evaluated by the Kaplan–Meier method and Cox regression analysis. Gene set enrichment analysis (GSEA) was also conducted. Results The stromal score showed significant differences in different molecular subtypes and T stages. Multivariate analyses further confirmed that the stromal score was an independent indicator of overall survival (OS) in the two cohorts. The low stromal score group showed higher tumor mutation burden (TMB) and micro-satellite instability (MSI), and was more sensitive to immune checkpoint inhibitor according to the TIDE algorithm. Activation of the transforming growth factor and epithelial–mesenchymal transition were observed in the high stromal score subtype, which is associated with T-cell suppression, and may be responsible for resistance to PD-1/PD-L1 therapy. BPIFB2 was confirmed as a hub gene relevant to immunotherapy. Conclusion The stromal score was associated with cancer progression and molecular subtypes, and may serve as a novel biomarker for predicting the prognosis and response to immunotherapy in patients with GC.
Collapse
Affiliation(s)
- Qianqian Ren
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Peng Zhu
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, 430022 China
| | - Hui Zhang
- Department of Internal Medicine, Wuhan Hankou Hospital, Wuhan, 430011 China
| | - Tianhe Ye
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Dehan Liu
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| | - Zhao Gong
- Department of Hepatobiliary Surgery, Wuhan No.1 Hospital, Wuhan, 430022 China
| | - Xiangwen Xia
- 1Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, 430022 China.,Hubei Province Key Laboratory of Molecular Imaging, Wuhan, 430022 China
| |
Collapse
|
49
|
Rubio K, Castillo-Negrete R, Barreto G. Non-coding RNAs and nuclear architecture during epithelial-mesenchymal transition in lung cancer and idiopathic pulmonary fibrosis. Cell Signal 2020; 70:109593. [PMID: 32135188 DOI: 10.1016/j.cellsig.2020.109593] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/28/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022]
Abstract
Lung cancer (LC) is the leading cause of cancer-related deaths worldwide. On the other hand, idiopathic pulmonary fibrosis (IPF) is the most common interstitial lung disease showing a prevalence of 20 new cases per 100,000 persons per year. Despite differences in cellular origin and pathological phenotypes, LC and IPF are lung diseases that share common features, including hyperproliferation of specific cell types in the lung, involvement of epithelial-mesenchymal transition (EMT) and enhanced activity of signaling pathways, such as tissue growth factor (TGFB), epidermal growth factor (EGF), fibroblast growth factor (FGF), wingless secreted glycoprotein (WNT) signaling, among others. EMT is a process during which epithelial cells lose their cell polarity and cell-cell adhesion, and acquire migratory and invasive properties to become mesenchymal cells. EMT involves numerous morphological hallmarks of hyperproliferative diseases, like cell plasticity, resistance to apoptosis, dedifferentiation and proliferation, thereby playing a central role during organ fibrosis and cancer progression. EMT was considered as an "all-or-none" process. In contrast to these outdated dichotomist interpretations, recent reports suggest that EMT occurs gradually involving different epithelial cell intermediate states with mesenchyme-like characteristics. These cell intermediate states of EMT differ from each other in their cell plasticity, invasiveness and metastatic potential, which in turn are induced by signals from their microenvironment. EMT is regulated by several transcription factors (TFs), which are members of prominent families of master regulators of transcription. In addition, there is increasing evidence for the important contribution of noncoding RNAs (ncRNAs) to EMT. In our review we highlight articles dissecting the function of different ncRNAs subtypes and nuclear architecture in cell intermediate states of EMT, as well as their involvement in LC and IPF.
Collapse
Affiliation(s)
- Karla Rubio
- Brain and Lung Epigenetics (BLUE), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris-Est Créteil (UPEC), 94010 Créteil, France; Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Parkstraße 1, 61231 Bad Nauheim, Germany
| | - Rafael Castillo-Negrete
- Brain and Lung Epigenetics (BLUE), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris-Est Créteil (UPEC), 94010 Créteil, France; Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Parkstraße 1, 61231 Bad Nauheim, Germany
| | - Guillermo Barreto
- Brain and Lung Epigenetics (BLUE), Glycobiology, Cell Growth and Tissue Repair Research Unit (Gly-CRRET), Université Paris-Est Créteil (UPEC), 94010 Créteil, France; Lung Cancer Epigenetic, Max-Planck-Institute for Heart and Lung Research, Parkstraße 1, 61231 Bad Nauheim, Germany; Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russian Federation; Universities of Giessen and Marburg Lung Center (UGMLC), The German Center of Lung Research (Deutsches Zentrum für Lungenforschung, DZL), Germany.
| |
Collapse
|
50
|
Kon E, Benhar I. Immune checkpoint inhibitor combinations: Current efforts and important aspects for success. Drug Resist Updat 2019; 45:13-29. [DOI: 10.1016/j.drup.2019.07.004] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022]
|