1
|
Lescroart M, Kemp H, Imauven O, Raphalen JH, Bagate F, Schmidt J, Issa N, Decavele M, Moreau AS, Tamion F, Mourvillier B, Calvet L, Canet E, Lebert C, Pons S, Lacave G, Wallet F, Winiszewski H, Merdji H, De Chambrun MP, Argaud L, Kimmoun A, Dumas G, Zafrani L. Cardiogenic shock in patients with active onco-hematological malignancies: A multicenter retrospective study. J Crit Care 2025; 87:155028. [PMID: 39848115 DOI: 10.1016/j.jcrc.2025.155028] [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: 10/28/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/25/2025]
Abstract
PURPOSE Onco-hematological (OH) patients face significant cardiovascular risks due to malignancy and drug toxicity. Data are limited on the characteristics and outcomes of OH patients with cardiogenic shock (CS) in intensive care units (ICUs). METHODS This multicenter retrospective study included 214 OH patients with CS across 22 ICUs (2010-2021). The objectives were to (i) identify risk factors for 30-day mortality, (ii) describe early and long-term outcomes, and (iii) assess the prognostic impact of malignancy by comparing OH patients to a control group of CS patients. RESULTS The 30-day survival rate was 44.8 %. Multivariate analysis identified previous cardiomyopathy (OR = 1.61), acute kidney injury (OR = 1.62), lactate levels (OR = 1.08 per 1 mmol/L), pulmonary embolism (OR = 3.04), invasive mechanical ventilation (OR = 3.48), and epinephrine use (OR = 2.09) as factors associated with 30-day mortality. Among ICU survivors, 54 % were alive at 1 year with a median left ventricular ejection fraction of 52 %. OH malignancy was significantly associated with 30-day mortality (HR 2.54). CONCLUSION The prognosis for OH patients with CS in the ICU is poor, with epinephrine use associated with worse outcomes. Further research is needed to refine risk stratification and improve treatments for this population.
Collapse
Affiliation(s)
- Mickael Lescroart
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France; CHRU de Nancy, Médecine Intensive et Réanimation Brabois, Université de Lorraine, Vandœuvre-Lès-Nancy, France
| | - Hélène Kemp
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France
| | - Olivier Imauven
- Groupe hospitalier Diaconesses - Croix Saint-Simon, institut d'anesthésie de l'Est Parisien, Paris, France
| | - Jean Herlé Raphalen
- Intensive Care Unit, Necker University Hospital, Assistance Publique-Hôpitaux de Paris, 149 rue de Sèvres, 75015 Paris, France
| | - François Bagate
- Service de Médecine Intensive Réanimation, AP-HP, CHU Henri Mondor, DHU A-TVB, 51, avenue du Mal de Lattre de Tassigny, 94010 Créteil Cedex, France
| | - Julien Schmidt
- Unité de médecine intensive et réanimation, Assistance Publique-Hôpitaux de Paris, Avicenne Hospital, Groupe Hospitalier Paris Seine Saint-Denis, Bobigny, France
| | - Nahema Issa
- Réanimation médicale, groupe hospitalier Saint-André, 1, rue Jean-Burguet, 33075 Bordeaux cedex, France; Médecine interne et maladies infectieuses, groupe hospitalier Saint-André, 1, rue Jean-Burguet, 33075 Bordeaux cedex, France
| | - Maxens Decavele
- APHP, Groupe Hospitalier Universitaire APHP-Sorbonne Université, site Pitié-Salpêtrière, Service Médecine Intensive et Réanimation (Département R3S), 75013, Paris, France
| | - Anne-Sophie Moreau
- CHU de Lille, Hôpital Salengro, Service de Médecine Intensive Réanimation, rue Emile-Laine, 59037 Lille, France
| | - Fabienne Tamion
- Service de Réanimation Médicale, Normandie Univ, UNIROUEN, U1096, CHU de Rouen, F 76000 Rouen, France
| | - Bruno Mourvillier
- Centre Hospitalo-Universitaire de Reims (CHU), Hôpital Robert-Debré, Service de Réanimation médicale, Reims, France
| | - Laure Calvet
- Service de Reanimation Medicale, Hopital Gabriel Monpied, CHU, Clermont-Ferrand, France
| | - Emmanuel Canet
- Medecine Intensive Reanimation, University Hospital Center, Nantes, France
| | - Christine Lebert
- Service Medico-Chirurgical, Unité de soins Intensifs, Centre Hospitalier de La Roche-sur-Yon, France
| | - Stephanie Pons
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anaesthesiology and Critical Care, Pitié-Salpêtrière Hospital, Paris, France; Pulmonary and Critical Care Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Guillaume Lacave
- Service de Réanimation Médico-Chirurgicale, Centre Hospitalier de Versailles, Le Chesnay, France
| | - Florent Wallet
- Médecine Intensive et Réanimation, Hôpital Lyon Sud, Pierre-Bénite 69495, France
| | - Hadrien Winiszewski
- Médecine Intensive et Réanimation, Centre Hospitalier Universitaire de Besançon, Université de Franche-Comté, France
| | - Hamid Merdji
- Service de Médecine Intensive et Réanimation, Nouvel Hôpital Civil, Hôpitaux Universitaires de Strasbourg, INSERM UMR 1260, Regenerative NanoMedicine, FMTS, Strasbourg, France
| | - Marc Pineton De Chambrun
- Sorbonne Université, UPMC Univ Paris 06, INSERM UMRS_1166-iCAN, Institute of Cardiometabolism and Nutrition, 75651 Paris Cedex 13, France; Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Medical Intensive Care Unit, 75651 Paris Cedex 13, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Hôpital Edouard Herriot, Service de Médecine Intensive-Réanimation, 5, Place d'Arsonval, 69437 Lyon Cedex 03, France
| | - Antoine Kimmoun
- CHRU de Nancy, Médecine Intensive et Réanimation Brabois, Université de Lorraine, Vandœuvre-Lès-Nancy, France; INSERM U942, MASCOT, Paris, France
| | - Guillaume Dumas
- Medical Intensive Care Unit, Service de Médecine Intensive-Réanimation, CHU Grenoble-Alpes, Université Grenoble-Alpes, INSERM, U1042-HP2, Grenoble, France
| | - Lara Zafrani
- Hospital Saint-Louis et Université Paris Cité, Assistance Publique-Hôpitaux de Paris, France.
| |
Collapse
|
2
|
Rajak P. Immune checkpoint inhibitors: From friend to foe. Toxicol Rep 2025; 14:102033. [PMID: 40353246 PMCID: PMC12063143 DOI: 10.1016/j.toxrep.2025.102033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 04/18/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025] Open
Abstract
Immune checkpoints are crucial in regulating the activation of cell-mediated and humoral immune responses. However, cancer cells hijack this mechanism to evade the immune surveillance and anti-cancer response. Typically, receptors like PD-1 and CTLA4, expressed on immune cells, prevent the activation and differentiation of T cells. They also inhibit the development of autoimmune reactions. However, ligands such as PD-L1 for the receptor PD-1 are also expressed on the surface of cancer cells that help prevent the activation of anti-cancer immune responses by blocking the signalling pathways mediated by PD-1 and CTLA4. Immune checkpoint inhibitors (ICIs) have promising therapeutic efficacy for treating several cancers by activating T cells and their differentiation into effector cells against tumours. Nonetheless, hyperactivated immune cells usually contribute to detrimental issues, also known as immune-related adverse effects (IrAE). IrAEs have been observed in multiple organs, leading to neurological issues, colitis, endocrine dysfunction, renal issues, hepatitis, pneumonitis, and dermatitis. The interplay between hyperactivated T cells and Treg cells helps in orchestrating the development of autoimmunity. Moreover, the crosstalk between proinflammatory interleukins and the development of autoantibodies also mediates the multiorgan effects of ICIs in cancer patients. IrAEs are generally managed by terminating the ICI therapy, reducing the ICI dose, and by using corticosteroids to subvert inflammation. Therefore, the present review aims to delineate the impacts of ICIs on the development of autoimmune diseases and inflammatory outcomes in cancer patients. In addition, mechanistic insight involving immune cells, cytokines, and autoantibodies for ICI-mediated IrAEs will also be discussed with updated findings in this field.
Collapse
Affiliation(s)
- Prem Rajak
- Toxicology Research Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol, West Bengal, India
| |
Collapse
|
3
|
Sayer M, Hamano H, Nagasaka M, Lee BJ, Doh J, Patel PM, Zamami Y, Ozaki AF. Time dependent predictors of cardiac inflammatory adverse events in cancer patients receiving immune checkpoint inhibitors. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:40. [PMID: 40296103 PMCID: PMC12036232 DOI: 10.1186/s40959-025-00331-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 03/24/2025] [Indexed: 04/30/2025]
Abstract
BACKGROUND Cardio-inflammatory immune related adverse events (irAEs) while receiving immune checkpoint inhibitor (ICI) therapy are particularly consequential due to their associations with poorer treatment outcomes. Evaluation of predictive factors of these serious irAEs with a time dependent approach allows better understanding of patients most at risk. OBJECTIVE To identify different elements of patient data that are significant predictors of early and late-onset or delayed cardio-inflammatory irAEs through various predictive modeling strategies. METHODS A cohort of patients receiving ICI therapy from January 1, 2010 to May 1, 2022 was identified from TriNetX meeting inclusion/exclusion criteria. Patient data collected included occurrence of early and later cardio-inflammatory irAEs, patient survival time, patient demographic information, ICI therapies, comorbidities, and medication histories. Predictive and statistical modeling approaches identified unique risk factors for early and later developing cardio-inflammatory irAEs. RESULTS A cohort of 66,068 patients on ICI therapy were identified in the TriNetX platform; 193 (0.30%) experienced early cardio-inflammatory irAEs and 175 (0.26%) experienced later cardio-inflammatory irAEs. Significant predictors for early irAEs included: anti-PD-1 therapy at index, combination ICI therapy at index, and history of peripheral vascular disease. Significant predictors for later irAEs included: a history of myocarditis and/or pericarditis, cerebrovascular disease, and history of non-steroidal anti-inflammatory medication use. CONCLUSIONS Cardio-inflammatory irAEs can be divided into clinically meaningful categories of early and late based on time since initiation of ICI therapy. Considering distinct risk factors for early-onset and late-onset events may allow for more effective patient monitoring and risk assessment.
Collapse
Affiliation(s)
- Michael Sayer
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA
| | - Hirofumi Hamano
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Misako Nagasaka
- Division of Hematology and Oncology, University of California, Irvine, CA, USA
| | - Benjamin J Lee
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Jean Doh
- Department of Pharmacy, University of California Irvine Health, Orange, CA, USA
| | - Pranav M Patel
- Division of Cardiology, Department of Medicine, University of California, Irvine, CA, USA
| | - Yoshito Zamami
- Department of Pharmacy, Okayama University Hospital, Okayama, Japan
| | - Aya F Ozaki
- School of Pharmacy & Pharmaceutical Sciences, University of California, 802 W Peltason Dr, Room 106A, Irvine, CA, 92617, USA.
| |
Collapse
|
4
|
Patel S, Dave K, Garcia MJ, Gongora CA, Travin MI, Zhang L. Multimodal Imaging of Immune Checkpoint Inhibitor Myocarditis. J Clin Med 2025; 14:2850. [PMID: 40283680 PMCID: PMC12028134 DOI: 10.3390/jcm14082850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically changed the landscape of cancer treatment and are increasingly used either as monotherapy or in combination with other ICIs, chemotherapy, and molecularly targeted agents. ICI myocarditis is a rare but potentially fatal irAE associated with the use of ICI characterized by T-cell mediated cardiomyocyte death. Diagnosing ICI myocarditis can be intricate as its atypical presentations. Multimodal imaging plays a crucial role in the diagnosis and risk stratification of ICI myocarditis. Current management strategies for ICI myocarditis include corticosteroids and immunosuppressants. Multidisciplinary collaboration is vital in these cases-combining oncology expertise with cardiology insights.
Collapse
Affiliation(s)
- Shreyans Patel
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Kartikeya Dave
- Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Mario J. Garcia
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Carlos A. Gongora
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| | - Mark I. Travin
- Division of Nuclear Medicine, Department of Radiology, Montefiore Medical Center and the Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA;
| | - Lili Zhang
- Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (M.J.G.); (C.A.G.); (L.Z.)
| |
Collapse
|
5
|
De Perna ML, Rigamonti E, Zannoni R, Espeli V, Moschovitis G. Immune Checkpoint Inhibitors and Cardiovascular Adverse Events. ESC Heart Fail 2025. [PMID: 40205958 DOI: 10.1002/ehf2.15281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/11/2025] Open
Abstract
In the last years, we assisted to a tremendous increase in therapeutic options for the management of cancers, with immunotherapy at the forefront of this innovation. Immune checkpoint inhibitors (ICIs) have been developed to enhance the activity of the immune system against cancer cells (1) and the number of approvals for ICIs has rapidly increased. ICIs have also been associated with disinhibited cytotoxic T cells that damage healthy tissue in multiple organs, causing immune-related adverse events (AEs). Cardiovascular AEs (CVAe) are increasingly reported: myocarditis, Takotsubo syndrome, pericarditis and pericardial effusion, worsening of atherosclerosis, acute coronary syndromes, non-inflammatory heart failure, and ischaemic stroke. They are classified into five grades, based on presenting symptoms, level of cardiac biomarkers, and imaging. Even though myocarditis occurs more frequently than previously thought, clinically relevant myocarditis is a rare irAE compared to other irAE (0.5-1.2%). The clinical manifestations range from mild symptoms such as to chest pain, heart failure, and cardiogenic shock. The prognosis is severe, with mortality rates ranging from 25% to 50%. It is frequently associated with the concomitant use of combination of checkpoint inhibitors. The treatment strategies are tripartite: (i) holding ICI to prevent further toxicity, (ii) immunosuppression to alleviate inflammatory changes, and (iii) supportive therapy to address cardiac complications. Glucocorticoids represent the first-line treatment. In hemodynamically unstable patients, treatment with high-dose steroids should be initiated (intravenous methylprednisolone 1000 or 1250 mg oral methylprednisolone during 4 days). ICI-associated pericarditis can be accompanied by no/mild pericardial effusion up to cardiac tamponade. The treatment is made of nonsteroidal anti-inflammatory drugs and colchicine, corticosteroids if needed, and pericardiocentesis for the large effusions. ICIs could be continued for Grade 1 pericarditis, while temporary suspension of ICI is warranted for more severe cases. There is significant potential for accelerated atherosclerosis with ICIs as a long-term effect, but atherosclerosis-related CVAEs are not frequent, especially during treatment; increasing evidence associates ICIs with progression of atherosclerosis and increased atherosclerotic cardiovascular disease. ICIs can lead to arrhythmias: atrial fibrillation, supraventricular and ventricular tachycardias. Non-inflammatory heart failure syndrome have been observed in ICI-treated patients. Immune checkpoint inhibitors seem to be involved in the development of right ventricular dysfunction and pulmonary arterial hypertension. It is of the outmost importance to improve the collaboration among the different medical figures, such as cardiologists, oncologists, endocrinologists, and immunologists, both in clinical practice and in basic science research, to better recognize these adverse events, to understand their pathophysiological mechanisms, and to improve the overall survival and quality of life of the affected patients.
Collapse
Affiliation(s)
- Maria Luisa De Perna
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Elia Rigamonti
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Raffaele Zannoni
- Service de Médicine Interne générale, Département de médecine interne, Hopitaux Universitaires de Genève (HUG), rue Gabrielle-Perret-Gentil 4, Genève, Switzerland
| | - Vittoria Espeli
- Department of Oncology, Ente Ospedaliero Cantonale, Istituto Oncologico della Svizzera Italiana, Lugano, Switzerland
| | - Giorgio Moschovitis
- Istituto Cardiocentro Ticino, Department of Cardiology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| |
Collapse
|
6
|
Hachem AM, Desai A, Beinart N, Ostos-Mendoza KC, Rodriguez ASL, de Leon Derby RD, Ebrahimi S, Palaskas NL. Updates in Diagnosis and Treatment of Immune Checkpoint Inhibitor Myocarditis. Curr Cardiol Rep 2025; 27:78. [PMID: 40178703 DOI: 10.1007/s11886-025-02232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW To provide an update on the literature regarding diagnosis and management of immune checkpoint inhibitor myocarditis. RECENT FINDINGS The diagnosis of immune checkpoint inhibitor myocarditis has evolved to include more reliance on performing endomyocardial biopsy to clarify the diagnosis in selected cases. Additionally, there is recognition of a spectrum of disease both clinically and on endomyocardial biopsy suggesting that there is a range of severity from mild to fulminant. The treatment of immune checkpoint inhibitor myocarditis is shifting towards increased use of additional immunosuppressive medications as steroid sparing agents. There are increased studies including two randomized controlled trials evaluating abatacept in the treatment of immune checkpoint inhibitor myocarditis. This review summarizes the latest literature regarding diagnosis and management of immune checkpoint inhibitor myocarditis and provides our experience and approach to this rare but potentially fatal condition.
Collapse
Affiliation(s)
| | - Aditya Desai
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lakeside School, Seattle, WA, USA
| | - Noah Beinart
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keila C Ostos-Mendoza
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Ana Sofia Lopez Rodriguez
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Regina Diaz de Leon Derby
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Sara Ebrahimi
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicolas L Palaskas
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1451, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Gray R, Manisty C, Cheng R, Dastidar A, Mamas M, Ghosh A. Immune checkpoint inhibitors: Unravelling atherosclerotic cardiovascular risk. Atherosclerosis 2025; 403:119147. [PMID: 40037087 DOI: 10.1016/j.atherosclerosis.2025.119147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Affiliation(s)
| | | | - Richard Cheng
- Division of Cardiology, University of Washington, USA
| | - Amardeep Dastidar
- Bristol Heart Institute, NIHR Biomedical Research Centre Cardiovascular & North Bristol NHS Trust, Bristol, UK
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, UK
| | | |
Collapse
|
8
|
Jensen G, Wang X, Kuempel J, Palaskas N, Chen Z, Yu W, Chen Y, Mohammad H, Luo W, Chang J. Immune checkpoint inhibitor-associated myocarditis: a historical and comprehensive review. Am J Physiol Heart Circ Physiol 2025; 328:H734-H751. [PMID: 39925096 DOI: 10.1152/ajpheart.00687.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025]
Abstract
The most fatal side effect associated with revolutionary immune checkpoint inhibitor (ICI) cancer therapies is myocarditis, a rare and devastating complication with a mortality rate approaching 40%. This review comprehensively examines the limited knowledge surrounding this recently recognized condition, emphasizing the absence of evidence-based therapeutic strategies, diagnostic modalities, and reliable biomarkers that hinder effective management. It explores advancements in preclinical models that are uncovering disease mechanisms and enabling the identification of therapeutic targets. These efforts have informed the design of early clinical trials aimed at reducing mortality. With the growing prevalence of ICI therapies in oncology, addressing critical gaps, such as long-term outcomes and risk stratification, has become increasingly urgent. By synthesizing current evidence, this work seeks to enhance understanding and guide the development of strategies to improve patient outcomes and ensure the continued safe use of ICIs in cancer care.
Collapse
Affiliation(s)
- Garrett Jensen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Xinjie Wang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jacob Kuempel
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Nicolas Palaskas
- Department of Cardiology, MD Anderson Cancer Center, Houston, Texas, United States
| | - Zhishi Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Wei Yu
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Yanping Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Haseeb Mohammad
- Texas A&M University College of Medicine, Houston, Texas, United States
| | - Weijia Luo
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jiang Chang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| |
Collapse
|
9
|
Al-Mondhiry J, Barac A. Immune Checkpoint Inhibitor-Related Myocarditis, Myopathy, and More: Unraveling the Tangled Science of Immune-Related Adverse Events. JACC CardioOncol 2025; 7:265-267. [PMID: 40246384 PMCID: PMC12046833 DOI: 10.1016/j.jaccao.2025.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Affiliation(s)
- Jafar Al-Mondhiry
- Inova Schar Cancer Institute, Fairfax, Virginia, USA; University of Virginia School of Medicine, Charlottesville, Virginia, USA.
| | - Ana Barac
- Inova Schar Cancer Institute, Fairfax, Virginia, USA; Inova Schar Heart and Vascular, Fairfax, Virginia, USA; Georgetown University School of Medicine, Washington, DC, USA. https://twitter.com/AnaBaracCardio
| |
Collapse
|
10
|
Chen YC, Dolladille C, Rao A, Palaskas NL, Deswal A, Lehmann L, Cautela J, Courand PY, Hayek S, Zhu H, Cheng RK, Alexandre J, Baldassarre LA, Roubille F, Laufer-Perl M, Asnani A, Ederhy S, Tamura Y, Francis S, Gaughan EM, Johnson DB, Flint DL, Rainer PP, Bailly G, Ewer SM, Aras MA, Arangalage D, Cariou E, Florido R, Peretto G, Itzhaki Ben Zadok O, Akhter N, Narezkina A, Levenson JE, Liu Y, Crusz SM, Issa N, Piriou N, Leong D, Sandhu S, Turker I, Moliner P, Obeid M, Heinzerling L, Chang WT, Stewart A, Venkatesh V, Du Z, Yadavalli A, Kim D, Chandra A, Zhang KW, Power JR, Moslehi J, Salem JE, Zaha VG. Immune Checkpoint Inhibitor Myocarditis and Left Ventricular Systolic Dysfunction. JACC CardioOncol 2025; 7:234-248. [PMID: 40246381 PMCID: PMC12046861 DOI: 10.1016/j.jaccao.2025.01.020] [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: 11/27/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 04/19/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have transformed cancer treatment, but ICI myocarditis (ICI-M) remains a potentially fatal complication. The clinical implications and predictors of left ventricular ejection fraction (LVEF) <50% in ICI-M are not well understood. OBJECTIVES The aim of this study was to identify factors associated with LVEF <50% vs ≥50% at the time of hospitalization for ICI-M. A secondary objective was to evaluate the relationship between LVEF and 30-day all-cause mortality. METHODS The International ICI-Myocarditis Registry, a retrospective, international, multicenter database, included 757 patients hospitalized with ICI-M. Patients were stratified by LVEF as reduced LVEF (<50%) or preserved LVEF (≥50%) on admission. Cox proportional hazards models were used to assess the associations between LVEF and clinical events, and multivariable logistic regression was conducted to examine factors linked to LVEF. RESULTS Of 757 patients, 707 had documented LVEFs on admission: 244 (35%) with LVEF <50% and 463 (65%) with LVEF ≥50%. Compared with patients with LVEF ≥50%, those with LVEF <50% were younger (<70 years), had a body mass index of <25 kg/m2, and were more likely to have received chest radiation (24.2% vs 13.5%; P < 0.001). Multivariable analysis identified predictors of LVEF <50%, including exposure to v-raf murine sarcoma viral oncogene homolog B1/mitogen-activated protein kinase inhibitors, pre-existing heart failure, dyspnea at presentation, and at least 40 days from ICI initiation to ICI-M onset. Conversely, myositis symptoms were associated with LVEF ≥50%. LVEF <50% was marginally associated with 30-day all-cause mortality (unadjusted log-rank P = 0.062; adjusted for age, cancer types, and ICI therapy, HR: 1.50; 95% CI: 1.02-2.20). CONCLUSIONS Dyspnea, time from ICI initiation, a history of heart failure, and prior cardiotoxic therapy may be predictors of an initial LVEF <50% in patients with ICI-M.
Collapse
Affiliation(s)
- Yen-Chou Chen
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA; Division of Cardiology and Cardiovascular Research Center, Taipei Medical University Hospital, Taipei, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei, Taiwan.
| | - Charles Dolladille
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, Caen, France; Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Department of Pharmacology, Caen, France; Department of Pharmacology, Sorbonne University, INSERM, CIC-1901, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Anjali Rao
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Nicolas L Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Lorenz Lehmann
- Department of Cardiology, Angiology, and Pneumology, University Hospital of Heidelberg, Heidelberg, Germany; German Centre for Cardiovascular Research, partner site Heidelberg/Mannheim, Mannheim, Germany; German Cancer Research Center, Heidelberg, Germany
| | - Jennifer Cautela
- Department of Cardiology, University Mediterranean Centre of CardioOncology, Nord Hospital, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, INSERM, INRAE, C2VN, Marseille, France
| | - Pierre-Yves Courand
- Fédération de Cardiologie, Hôpital de la Croix-Rousse et Hôpital Lyon Sud, Hospices Civils de Lyon, Lyon, France; Université de Lyon, Université Claude Bernard, Lyon, France
| | - Salim Hayek
- Department of Cardiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Han Zhu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California; Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California, USA
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, Washington, USA
| | - Joachim Alexandre
- Normandie Université, UNICAEN, INSERM U1086 ANTICIPE, Caen, France; Caen-Normandy University Hospital, PICARO Cardio-Oncology Program, Department of Pharmacology, Caen, France
| | | | - François Roubille
- Department of Cardiology, INI-CRT, CHU de Montpellier, PhyMedExp, Université de Montpellier, Inserm, CNRS, Montpellier, France
| | - Michal Laufer-Perl
- Department of Cardiology, Tel Aviv Sourasky Medical Center, affiliated to the Tel Aviv University Faculty of Medicine, Tel Aviv-Yafo, Israel
| | - Aarti Asnani
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Stephane Ederhy
- Cardiology Department, Hospital Saint Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Yuichi Tamura
- Cardiovascular Center, International University of Health and Welfare, Mita Hospital, Tokyo, Japan
| | - Sanjeev Francis
- Cardiovascular Service Line, Maine Medical Center, Portland, Maine, USA
| | - Elizabeth M Gaughan
- Division of Hematology and Oncology, University of Virginia, Charlottesville, Virginia, USA
| | - Douglas B Johnson
- Division of Hematology and Oncology, Vanderbilt University, Nashville, Tennessee, USA
| | - Danette L Flint
- Heart and Vascular Center, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz Austria; BioTechMed Graz, Graz, Austria; St. Johann in Tirol General Hospital, St. Johann in Tirol, Austria
| | - Guillaume Bailly
- Assistance Publique-Hôpitaux de Paris Hôpital Lariboisière, Paris, France
| | - Steven M Ewer
- Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, USA
| | - Mandar A Aras
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA
| | - Dimitri Arangalage
- Department of Cardiology, Bichat Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Université de Paris, UMRS1148, INSERM, Paris, France; Université de Paris, Paris, France
| | - Eve Cariou
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
| | - Roberta Florido
- Division of Cardiology, Johns Hopkins University, Baltimore, Maryland, USA
| | - Giovanni Peretto
- Disease Unit for Myocarditis and Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Nausheen Akhter
- Division of Cardiology, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Anna Narezkina
- Division of Cardiovascular Medicine, University of California-San Diego, San Diego, California, USA
| | - Joshua E Levenson
- Heart and Vascular Institute, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Yan Liu
- Division of Cardiology, Department of Internal Medicine, Dell Medical School, University of Texas at Austin, Austin, Texas, USA
| | - Shanthini M Crusz
- Barts Health NHS Trust, University College London Hospital, London, United Kingdom
| | - Nahema Issa
- Bordeaux University Hospital, Bordeaux, France
| | - Nicolas Piriou
- Nantes Univesrité, CHU Nantes, Centre de Reference Cardiomyopathies, l'Institut du Thorax, Nantes, France
| | - Darryl Leong
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Shahneen Sandhu
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Isik Turker
- Department of Cardiology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Pedro Moliner
- Bellvitge University Hospital, Catalan Institute of Oncology, Cardiology Department, Bio-Heart Cardiovascular Diseases Research Group, Bellvitge Biomedical Research Institute, CIBER CV, l'Hospitalet de Llobregat, Barcelona, Spain
| | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois, University of Lausanne, LCIT Center, Immunology and Allergy Service, Lausanne, Switzerland
| | - Lucie Heinzerling
- Department of Dermatology and Allergy, University Hospital, LMU Munich, Germany
| | | | - Andrew Stewart
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Vishnu Venkatesh
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Zoe Du
- University of Texas at Dallas, Dallas, Texas, USA
| | | | - Dohyeong Kim
- University of Texas at Dallas, Dallas, Texas, USA
| | - Alvin Chandra
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Kathleen W Zhang
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - John R Power
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA; Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California
| | - Javid Moslehi
- Division of Cardiology, University of California-San Francisco, San Francisco, California, USA
| | - Joe-Elie Salem
- Department of Pharmacology, Sorbonne University, INSERM, CIC-1901, Hôpital Pitié-Salpétrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Vlad G Zaha
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
11
|
Camilli M, Maggio L, Tinti L, Torre I, Viscovo M, Viscovo M, Tamburrini G, Lombardo A, Cardinale DM, Minotti G, Rocca B. Cardio-oncology: Emerging Concepts in Cardiovascular Sequelae of Cancer Therapies, Translational Research and Reverse Cardio-oncology. Eur Cardiol 2025; 20:e05. [PMID: 40170756 PMCID: PMC11959581 DOI: 10.15420/ecr.2024.49] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/11/2024] [Indexed: 04/03/2025] Open
Abstract
Cardio-oncology was established with the aim of defining primary and secondary prevention approaches through surveillance and the use of tools to stratify and diminish the cardiovascular risk to cancer patients. This branch of medicine also contributes to establishing a new field in translational medicine for cardiovascular disease by focusing on the interplay between cancer and heart disease. In this first article in the new cardio-oncology section of the journal, we explore the main concepts of emerging anti-cancer therapies and their plausible cardiotoxic effects and we will describe advances and gaps in knowledge, highlighting how cardio-oncology is contributing to translational cardiology. We will speculate on the complex interplay between cancer and heart failure and discuss an emerging concept known as reverse cardio-oncology. We also present the perspective that cardio-oncology represents a promising platform area of research, allowing the discovery of novel pathways involved in cardiovascular disease through the identification of toxicities induced by targeted cancer therapies.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Luca Maggio
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Ilaria Torre
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Marcello Viscovo
- Department of Laboratory and Hematology Sciences, Fondazione Policlinico Universitario A Gemelli IRCCSRome, Italy
- Department of Radiology and Hematology Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Giulia Tamburrini
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Università Cattolica del Sacro CuoreRome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCSRome, Italy
| | | | - Giorgio Minotti
- Unit of Drug Sciences, Fondazione Policlinico Universitario Campus Bio-MedicoRome, Italy
| | - Bianca Rocca
- Department of Medicine and Surgery, Libera Università MediterraneaBari, Italy
- Department of Safety and Bioethics, Università Cattolica del Sacro CuoreRome, Italy
| |
Collapse
|
12
|
Travers S, Alexandre J, Baldassarre LA, Salem JE, Mirabel M. Diagnosis of cancer therapy-related cardiovascular toxicities: A multimodality integrative approach and future developments. Arch Cardiovasc Dis 2025; 118:185-198. [PMID: 39947997 DOI: 10.1016/j.acvd.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 03/14/2025]
Abstract
Diagnosing cancer therapy-related cardiovascular toxicities may be a challenge. The interplay between cancer and cardiovascular diseases, beyond shared cardiovascular and cancer risk factors, and the increasingly convoluted cancer therapy schemes have complicated cardio-oncology. Biomarkers used in cardio-oncology include serum, imaging and rhythm modalities to ensure proper diagnosis and prognostic stratification of cardiovascular toxicities. For now, troponin and natriuretic peptides, multimodal cardiovascular imaging (led by transthoracic echocardiography combined with cardiac magnetic resonance or computed tomography angiography) and electrocardiography (12-lead or Holter monitor) are cornerstones in cardio-oncology. However, the imputability of cancer therapies is sometimes difficult to assess, and more refined biomarkers are currently being studied to increase diagnostic accuracy. Advances reside partly in pathophysiology-based serum biomarkers, improved cardiovascular imaging through new technical developments and remote monitoring for rhythm disorders. A multiparametric omics approach, enhanced by deep-learning techniques, should open a new era for biomarkers in cardio-oncology in the years to come.
Collapse
Affiliation(s)
- Simon Travers
- INSERM UMR-S 1180, Université Paris-Saclay, 91400 Orsay, France; Laboratoire de Biochimie, DMU BioPhyGen, Hôpital Européen Georges Pompidou, AP-HP, 75015 Paris, France.
| | - Joachim Alexandre
- INSERM U1086 ANTICIPE, Biology-Research Building, UNICAEN, Normandie University Group, 14000 Caen, France; Department of Pharmacology, Biology-Research Building, PICARO Cardio-Oncology Programme, Caen-Normandy University Hospital, 14000 Caen, France.
| | - Lauren A Baldassarre
- Cardiovascular Medicine, Yale School of Medicine, 06510 New Haven CT, United States of America.
| | - Joe Elie Salem
- CIC-1901, Department of Pharmacology, Hôpital Pitié-Salpêtrière, AP-HP, Sorbonne Université, INSERM, 75013 Paris, France.
| | - Mariana Mirabel
- Cardiology Department, Institut Mutualiste Montsouris, 75014 Paris, France.
| |
Collapse
|
13
|
Lal JC, Fang MZ, Hussain M, Abraham A, Tonegawa-Kuji R, Hou Y, Chung MK, Collier P, Cheng F. Discovery of plasma proteins and metabolites associated with left ventricular cardiac dysfunction in pan-cancer patients. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:17. [PMID: 39948601 PMCID: PMC11823021 DOI: 10.1186/s40959-025-00309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/23/2025] [Indexed: 02/16/2025]
Abstract
BACKGROUND Cancer-therapy related cardiac dysfunction (CTRCD) remains a significant cause of morbidity and mortality in cancer survivors. In this study, we aimed to identify differential plasma proteins and metabolites associated with left ventricular dysfunction (LVD) in cancer patients. METHODS We analyzed data from 50 patients referred to the Cleveland Clinic Cardio-Oncology Center for echocardiograph assessment, integrating electronic health records, proteomic, and metabolomic profiles. LVD was defined as an ejection fraction ≤ 55% based on echocardiographic evaluation. Classification-based machine learning models were used to predict LVD using plasma metabolites and proteins as input features. RESULTS We identified 13 plasma proteins (P < 0.05) and 14 plasma metabolites (P < 0.05) associated with LVD. Key proteins included markers of inflammation (ST2, TNFRSF14, OPN, and AXL) and chemotaxis (RARRES2, MMP-2, MEPE, and OPN). Notably, sex-specific associations were observed, such as uridine (P = 0.003) in males. Furthermore, metabolomic features significantly associated with LVD included 1-Methyl-4-imidazoleacetic acid (P = 0.015), COL1A1 (P = 0.009), and MMP-2 (P = 0.016), and pointing to metabolic shifts and heightened inflammation in patients with LVD. CONCLUSION Our findings suggest that circulating metabolites may non-invasively detect clinical and molecular differences in patients with LVD, providing insights into underlying disease pathways and potential therapeutic targets.
Collapse
Affiliation(s)
- Jessica C Lal
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Gastrointestinal Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Michelle Z Fang
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Muzna Hussain
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Abel Abraham
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Reina Tonegawa-Kuji
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mina K Chung
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
- Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Patrick Collier
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Department of Cardiovascular Medicine, Heart, Vascular & Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA.
- Section of Cardiovascular Imaging, Robert and Suzanne Tomsich Department Of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, The Cleveland Clinic Foundation, Cleveland, OH, USA.
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Cleveland Clinic Genome Center, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
14
|
Todo M, Gatate Y, Nakano S, Kaneko G, Hagiwara M, Takahashi T, Umezawa Y, Ueda G, Ishikawa S, Makino Y, Oyama M, Shirotake S. Early detection of myocarditis caused by immune checkpoint inhibitor therapy with nivolumab and ipilimumab for advanced recurrent renal cell carcinoma. Cancer Immunol Immunother 2025; 74:97. [PMID: 39904795 PMCID: PMC11794898 DOI: 10.1007/s00262-025-03945-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Combination therapy with nivolumab and ipilimumab for advanced renal cell carcinoma (RCC) may cause immune-related myocarditis; however, its incidence in this cancer type and regimen remains unknown. At our institution, we measure biomarkers, such as high-sensitivity Troponin (hsTn), and perform electrocardiograms (ECGs) and echocardiography before and every month after the initiation of this therapy, and the findings obtained and patients' symptoms are continuously monitored by physicians and pharmacists. A retrospective survey was conducted on physiological and biochemical test findings and immune-related adverse events in patients with advanced RCC who received combination therapy with nivolumab and ipilimumab between October 1, 2018 and December 31, 2023. Patients suspected of having myocarditis consulted with cardiologists. Myocarditis due to this therapy was detected in 5 of the 86 patients (5.8%) assessed using the European Society of Cardiology 2022 guidelines. There were no fatal symptoms or death due to myocarditis. The median time to the onset of myocarditis was 25 days (21-86 days). The early detection of myocarditis caused by this therapy requires the monitoring of changes by periodically measuring hsTn and other cardiac markers and performing ECGs and echocardiography from the early stages of administration through to the end of treatment. In addition to checking symptoms, if these abnormalities are detected and myocarditis is suspected, prompt collaboration with cardiologists is recommended. Our management strategy of care by a onco-cardiology team may contribute to the early diagnosis and treatment of myocarditis.
Collapse
Affiliation(s)
- Maki Todo
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan.
| | - Yodo Gatate
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Shintaro Nakano
- Department of Cardiology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Go Kaneko
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Masayuki Hagiwara
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Takayuki Takahashi
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Yuta Umezawa
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Genji Ueda
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Shiho Ishikawa
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Yoshinori Makino
- Department of Pharmacy, Saitama Medical University Saitama International Medical Center, 1397-1, Yamane, Hidaka, Saitama, 350-1298, Japan
| | - Masafumi Oyama
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| | - Suguru Shirotake
- Department of Uro-Oncology, Saitama Medical University Saitama International Medical Center, Saitama, Japan
| |
Collapse
|
15
|
Efentakis P, Choustoulaki A, Kwiatkowski G, Varela A, Kostopoulos IV, Tsekenis G, Ntanasis-Stathopoulos I, Georgoulis A, Vorgias CE, Gakiopoulou H, Briasoulis A, Davos CH, Kostomitsopoulos N, Tsitsilonis O, Dimopoulos MA, Terpos E, Chłopicki S, Gavriatopoulou M, Andreadou I. Early microvascular coronary endothelial dysfunction precedes pembrolizumab-induced cardiotoxicity. Preventive role of high dose of atorvastatin. Basic Res Cardiol 2025; 120:263-286. [PMID: 38520533 PMCID: PMC11790778 DOI: 10.1007/s00395-024-01046-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024]
Abstract
Immune checkpoint inhibitors (ICIs) exhibit remarkable antitumor activity and immune-related cardiotoxicity of unknown pathomechanism. The aim of the study was to investigate the ICI class-dependent cardiotoxicity in vitro and pembrolizumab's (Pem's) cardiotoxicity in vivo, seeking for translational prevention means. Cytotoxicity was investigated in primary cardiomyocytes and splenocytes, incubated with ipilimumab, Pem and avelumab. Pem's cross-reactivity was assessed by circular dichroism (CD) on biotechnologically produced human and murine PD-1 and in silico. C57BL6/J male mice received IgG4 or Pem for 2 and 5 weeks. Echocardiography, histology, and molecular analyses were performed. Coronary blood flow velocity mapping and cardiac magnetic resonance imaging were conducted at 2 weeks. Human EA.hy926 endothelial cells were incubated with Pem-conditioned media from human mononuclear cells, in presence and absence of statins and viability and molecular signaling were assessed. Atorvastatin (20 mg/kg, daily) was administered in vivo, as prophylaxis. Only Pem exerted immune-related cytotoxicity in vitro. Pem's cross-reactivity with the murine PD-1 was confirmed by CD and docking. In vivo, Pem initiated coronary endothelial and diastolic dysfunction at 2 weeks and systolic dysfunction at 5 weeks. At 2 weeks, Pem induced ICAM-1 and iNOS expression and intracardiac leukocyte infiltration. At 5 weeks, Pem exacerbated endothelial activation and triggered cardiac inflammation. Pem led to immune-related cytotoxicity in EA.hy926 cells, which was prevented by atorvastatin. Atorvastatin mitigated functional deficits, by inhibiting endothelial dysfunction in vivo. We established for the first time an in vivo model of Pem-induced cardiotoxicity. Coronary endothelial dysfunction precedes Pem-induced cardiotoxicity, whereas atorvastatin emerges as a novel prophylactic therapy.
Collapse
MESH Headings
- Animals
- Atorvastatin/administration & dosage
- Atorvastatin/pharmacology
- Mice, Inbred C57BL
- Humans
- Male
- Cardiotoxicity/prevention & control
- Antibodies, Monoclonal, Humanized/toxicity
- Mice
- Immune Checkpoint Inhibitors/toxicity
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Coronary Vessels/drug effects
- Coronary Vessels/physiopathology
- Coronary Vessels/metabolism
- Disease Models, Animal
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Antineoplastic Agents, Immunological/toxicity
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Angeliki Choustoulaki
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Aimilia Varela
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis V Kostopoulos
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - George Tsekenis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Constantinos E Vorgias
- Department of Biochemistry & Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Harikleia Gakiopoulou
- Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ourania Tsitsilonis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
- Medical College, Jagiellonian University, Krakow, Poland
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece.
| |
Collapse
|
16
|
Makwana B, Malode A, Khadke S, Patel V, Shah R, Patel M, Parikh A, Dani SS, Ganatra S. Cardiac Complications of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy. Cardiol Clin 2025; 43:151-167. [PMID: 39551556 DOI: 10.1016/j.ccl.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy have revolutionized cancer treatment but can cause life-threatening cardiovascular toxicities through immune-related adverse events. Myocarditis is the most common and potentially fatal toxicity with immune checkpoint inhibitors. T-cell therapies can potentially lead to cytokine release syndrome. Diagnosis of ICI-myocarditis requires a multimodal approach, including biomarkers, imaging, and endomyocardial biopsy, while CRS is characterized by a clinical syndrome resembling distributive shock. Management involves discontinuing the offending therapy, immunosuppression with corticosteroids for ICI-myocarditis, and interleukin-6 antagonists for CRS. Collaboration between oncologists and cardiologists is crucial for early recognition and prompt treatment.
Collapse
Affiliation(s)
- Bhargav Makwana
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aishwarya Malode
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sumanth Khadke
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Vahin Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Rushin Shah
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Manav Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aneri Parikh
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sourbha S Dani
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sarju Ganatra
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA; Department of Medicine (Research), Cardio-Oncology Program, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA.
| |
Collapse
|
17
|
Khattab M, Baig M, El Zarif T, Barac A, Ferencik M, Henry ML, Lopez-Mattei J, Redheuil A, Salem JE, Scherrer-Crosbie M, Yang EH, Baldassarre LA. How to Use Imaging: Complex Cases of Atherosclerosis, Myocardial Inflammation, and Cardiomyopathy in Cardio-Oncology. Circ Cardiovasc Imaging 2025; 18:e015981. [PMID: 39772610 DOI: 10.1161/circimaging.124.015981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025]
Abstract
It is well understood that cancer therapies including chemotherapy, tyrosine kinase inhibitors, immune checkpoint inhibitors, and radiation can increase the risk of cardiovascular disease in patients with cancer. This can manifest as a multitude of pathologies including left ventricular dysfunction, myocarditis, cardiomyopathy, accelerated atherosclerosis, and coronary vasospasm. Multimodal cardiac imaging plays a critical role in diagnosing such pathologies by relying on noninvasive tools including echocardiograms, cardiac magnetic resonance imaging, positron emission tomography, single-photon emission computed tomography, and coronary computed tomography angiography. These methods have unique considerations and in recent years have made significant progress in their diagnostic capabilities in this patient population. As the field of cardio-oncology continues to expand rapidly, guidance on the management of such toxicities and the development of imaging technologies is crucial. In this review, we present 2 complex cases of atherosclerosis and myocarditis in patients with cancer, highlighting our rationale for management and discussing the nuances of various cardiac imaging modalities.
Collapse
Affiliation(s)
- Mohamad Khattab
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (M.K., M.B., T.E.Z., L.A.B.)
| | - Mariam Baig
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (M.K., M.B., T.E.Z., L.A.B.)
| | - Talal El Zarif
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (M.K., M.B., T.E.Z., L.A.B.)
| | - Ana Barac
- Inova Heart Institute, Fairfax, VA (A.B.)
| | - Maros Ferencik
- Knight Cardiovascular Institute, Oregon Health Sciences University, Portland (M.F.)
| | - Mariana L Henry
- Geisel School of Medicine at Dartmouth, Hanover, NH (M.L.H.)
| | | | - Alban Redheuil
- Department of Pharmacology, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, INSERM, CIC-1901, UNICO-GRECO Cardio-oncology Program, France (A.R., J.-E.S.)
| | - Joe-Elie Salem
- Department of Pharmacology, Assistance Publique-Hôpitaux de Paris, Sorbonne Université, INSERM, CIC-1901, UNICO-GRECO Cardio-oncology Program, France (A.R., J.-E.S.)
| | | | - Eric H Yang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles (E.H.Y.)
| | - Lauren A Baldassarre
- Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT (M.K., M.B., T.E.Z., L.A.B.)
| |
Collapse
|
18
|
Romann SW, Giannitsis E, Frey N, Lehmann LH. Troponin Elevation in Asymptomatic Cancer Patients: Unveiling Connections and Clinical Implications. Curr Heart Fail Rep 2024; 21:505-514. [PMID: 39254897 PMCID: PMC11511716 DOI: 10.1007/s11897-024-00681-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/22/2024] [Indexed: 09/11/2024]
Abstract
PURPOSE OF THE REVIEW Elevated troponin levels are well established e.g., for the diagnosis of suspected acute coronary syndrome in symptomatic patients. In contrast, troponin elevations in asymptomatic cancer patients emerge as a complex phenomenon, challenging traditional perceptions of its association solely with cardiac events. RECENT FINDINGS Recent data support the predictive value of cardiac biomarker for all-cause mortality and cardiotoxicity in cancer patients. This review gives an overview about the current literature about cardiac troponins in prediction and identification of high-risk cancer patients. The overview is focusing on diagnostic challenges, biomarker significance, and gaps of knowledge. Latest publications highlight the relevance of cardiac troponin in risk analysis before cancer treatment as well as a potential diagnostic gatekeeper for further cardiological diagnostics and therapy.
Collapse
Affiliation(s)
- Sebastian W Romann
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Norbert Frey
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany
| | - Lorenz H Lehmann
- Department of Internal Medicine III: Cardiology, Angiology & Pulmonology, Cardio-Oncology Unit, Heidelberg University Hospital, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, 69120, Heidelberg, Germany.
- German Cancer Research Center (DKFZ), 69120, Heidelberg, Germany.
| |
Collapse
|
19
|
Tamura Y, Tamura Y. Evaluating Troponin-Based Monitoring in Patients Undergoing Immune Checkpoint Inhibitor Therapy. JACC. ADVANCES 2024; 3:101374. [PMID: 39583871 PMCID: PMC11584775 DOI: 10.1016/j.jacadv.2024.101374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Affiliation(s)
- Yudai Tamura
- Pulmonary Hypertension Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
- Cardiology, Kahoku Central Hospital, Ishikawa, Japan
| | - Yuichi Tamura
- Pulmonary Hypertension Center, International University of Health and Welfare Mita Hospital, Tokyo, Japan
- Department of Cardiology, International University of Health and Welfare School of Medicine, Narita, Japan
| |
Collapse
|
20
|
van den Berg PF, Bracun V, Noordman M, van der Meer P, Shi C, Oosting SF, Aboumsallem JP, de Wit S, Meijers WC, Jalving M, van Kruchten M, de Boer RA. Elevations of Cardiac Troponin in Patients Receiving Immune Checkpoint Inhibitors: Data From a Prospective Study. JACC. ADVANCES 2024; 3:101375. [PMID: 39583866 PMCID: PMC11584941 DOI: 10.1016/j.jacadv.2024.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 11/26/2024]
Abstract
Background Immune checkpoint inhibitors (ICIs) are increasingly used in the treatment of cancer. However, immune-related adverse events are prevalent in patients receiving ICI therapy. A serious immune-related adverse event is ICI-myocarditis, which is complex to diagnose given that the significance of early symptoms and biomarker trajectories, such as high-sensitivity troponin T (hs-TnT) are unclear. Objectives The purpose of the study was to evaluate kinetics of hs-TnT in cancer patients receiving ICI and to identify patients at risk of developing ICI-myocarditis. Methods This prospective, observational, single-center study included 164 patients receiving ICI therapy. Patients' history, demographics, and clinical characteristics, as well as survival statistics, were collected from electronic patient records and used to analyze associations between elevated hs-TnT (≥14 ng/L) and a significant rise in hs-TnT (100% rise from baseline, with an absolute value ≥2x upper limit of normal (ie, ≥28 ng/L) with ICI-myocarditis. Results We included 164 patients with a mean follow-up time of 1.60 ± 0.91 years. Melanoma was the most common type of cancer in the patient population, and most patients received treatment with programmed cell death protein 1 (PD-1). Twenty-six patients (16%) exhibited significant hs-TnT elevations, while 8 patients (5%) developed ICI-myocarditis. In 18 of 26 (69%) patients, ICI-myocarditis could not be diagnosed with certainty, while 10 of 26 (38%) patients had no other signs of symptoms of cardiac damage. All 8 myocarditis cases were preceded by significantly higher hs-TnT elevations than asymptomatic patients. Despite a high ICI-myocarditis incidence in our study population, cardiac mortality remained low (4%). Conclusions Significant hs-TnT elevations occur more often than previously reported, are often asymptomatic, and do not always lead to myocarditis diagnosis.
Collapse
Affiliation(s)
- Pieter F. van den Berg
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel Noordman
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Canxia Shi
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Sjoukje F. Oosting
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Sanne de Wit
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Wouter C. Meijers
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel van Kruchten
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudolf A. de Boer
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| |
Collapse
|
21
|
Oyakawa T, Muraoka N, Iida K, Fujita A, Yokoyama K, Ishikawa H, Murakami H. Relevance of surveillance manual for the early detection of immune checkpoint inhibitor-induced myocarditis: A case series. Asia Pac J Oncol Nurs 2024; 11:100598. [PMID: 39582548 PMCID: PMC11582373 DOI: 10.1016/j.apjon.2024.100598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 09/18/2024] [Indexed: 11/26/2024] Open
Abstract
Objective The European Cardio-Oncology Guidelines recommend regular electrocardiography and troponin testing during immune checkpoint inhibitors (ICIs) treatment, but their efficacy for monitoring ICI treatment remains unclear. This study aimed to evaluate the effectiveness of a surveillance protocol for early detection of ICI-induced myocarditis. Methods Between May 2014 and May 2024, patients who began treatment with ICIs at our hospital and developed ICI-induced myocarditis were included in this study. We created a straightforward management flowchart for myocarditis. The protocol was based on monitoring troponin T levels. We confirmed the efficacy of our surveillance protocol using a case series of ICI-induced myocarditis. Results During the observation period, 3481 patients were newly started on ICIs at our hospital. Five patients were previously diagnosed with myocarditis, and five patients were diagnosed with myocarditis after the implementation of the surveillance protocol. The manual enabled the early detection of myocarditis, and the mortality rate for myocarditis at our hospital improved from 60% to 0%. The incidence of conduction system disorders significantly reduced from 100% to 0% (P < 0.01). After the surveillance protocol was initiated, there were no cases of myocarditis requiring immunosuppressive drugs beyond steroids. Conclusions This study confirmed the relevance of a troponin-based surveillance protocol for the early detection of ICI-induced myocarditis. The implementation of the surveillance protocol reduced mortality from myocarditis and significantly reduced serious complications of conduction system disorders. Although this study is a small case series of patients who developed myocarditis, we confirm the effectiveness of surveillance for myocarditis.
Collapse
Affiliation(s)
- Takuya Oyakawa
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Nao Muraoka
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kei Iida
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
- Mishimatoukai Hospital, Shizuoka, Japan
| | - Ayano Fujita
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | | | - Haruyasu Murakami
- Division of Advanced Medical Development, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
22
|
Guan Z, Yao T, Liu G, Liu J, Guo L, Li Z, Ma J. Peripheral biomarkers to assess risk, severity, and prognosis of immune checkpoint inhibitor-associated myocarditis: a retrospective clinical study. Front Cardiovasc Med 2024; 11:1465743. [PMID: 39512372 PMCID: PMC11540693 DOI: 10.3389/fcvm.2024.1465743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Background Immune checkpoint inhibitor-associated myocarditis (ICI myocarditis) is an infrequent but potentially fatal immune-related adverse event. This study aimed to identify valuable indicators for risk prediction and evaluation of disease severity and outcomes. Methods A total of 79 patients with severe or mild ICI myocarditis and 158 controls without post-ICI immune-related adverse events were enrolled in this retrospective study. The clinical application value of a series of simple biomarkers were tested. Results Higher levels of the systemic immune-inflammation index (SII), neutrophil-to-eosinophil ratio (NER), aspartate transferase-to-albumin ratio (AAR), and lactic dehydrogenase-to-albumin ratio (LAR) at myocarditis onset were associated with severe disease conditions. In the receiver operating characteristic analysis, biomarkers areas under the curve (AUC) close to or greater than 0.8 were LAR (AUC: 0.810) and AAR (AUC: 0.806). Patients with higher SII, AAR, and LAR also exhibited poorer overall survival. The SII, NER, AAR, and LAR before the last ICI treatment increased relative to baseline in patients with ICI myocarditis, whereas no significant changes in the tested biomarkers were observed in the control group. For SII, AAR, and LAR, high ratios of the biomarker levels before the last ICI to baseline was associated with the incidence of myocarditis. Conclusions Surveillance of these economical biomarkers during ICI therapy might contribute to the risk prediction of ICI myocarditis, as well as the assessment of disease severity and prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jingtao Ma
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
23
|
Gradone AL, Ma VT, Vasbinder A, Fecher LA, Yentz S, Hayek SS, Lao CD. Increased myositis and possible myocarditis in melanoma patients treated with immune checkpoint inhibitors in the COVID-19 era. Cancer Immunol Immunother 2024; 73:259. [PMID: 39369180 PMCID: PMC11456101 DOI: 10.1007/s00262-024-03803-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 08/08/2024] [Indexed: 10/07/2024]
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI)-mediated myocarditis results in significant morbidity and mortality. At our institution, we noted an increased incidence of ICI-mediated myocarditis cases, leading to further investigation in our database of advanced melanoma patients treated with ICI therapy. METHODS A single-center, retrospective cohort analysis of patients with advanced melanoma identified cases of ICI-mediated myocarditis and myositis. RESULTS 366 patients with advanced melanoma received a dose of ICI from September 2014 to October 2019. Of these patients, there were 0 cases of ICI-mediated myocarditis (0%, 95% CI 0%-1.0%) and 2 cases of ICI-mediated myositis (0.55%, 95% CI 0.07%-1.96%). From November 2019 to December 2021, an additional 246 patients with advanced melanoma were identified. Of these patients, 10 (4.1%, 95% CI 1.97%-7.35%) developed ICI-mediated myocarditis and 10 developed ICI-mediated myositis. CONCLUSION Our study suggests an increase in prevalence of ICI-mediated muscle damage including myositis and myocarditis in the COVID-19 era. Differentiation of these patients and further risk stratification may allow for development of guidelines for nuanced management of this serious complication.
Collapse
Affiliation(s)
- Allison L Gradone
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Vincent T Ma
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Internal Medicine, University of Wisconsin, Madison, WI, USA
- Department of Dermatology, University of Wisconsin, Madison, WI, USA
| | - Alexi Vasbinder
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Leslie A Fecher
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Sarah Yentz
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Salim S Hayek
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Christopher D Lao
- Division of Hematology and Oncology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
24
|
Barac A, Wadlow RC, Deeken JF, deFilippi C. Cardiac Troponin I and T in ICI Myocarditis Screening, Diagnosis, and Prognosis. JACC CardioOncol 2024; 6:804-807. [PMID: 39479330 PMCID: PMC11520212 DOI: 10.1016/j.jaccao.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024] Open
Affiliation(s)
- Ana Barac
- Inova Schar Heart and Vascular, Fairfax, Virginia, USA
- Inova Schar Cancer, Fairfax, Virginia, USA
| | | | | | | |
Collapse
|
25
|
Ruiz-Esteves KN, Shank KR, Deutsch AJ, Gunturi A, Chamorro-Pareja N, Colling CA, Zubiri L, Perlman K, Ouyang T, Villani AC, Florez JC, Gusev A, Reynolds KL, Miller KK, Udler MS, Sise ME, Rengarajan M. Identification of Immune Checkpoint Inhibitor-Induced Diabetes. JAMA Oncol 2024; 10:1409-1416. [PMID: 39207773 PMCID: PMC11362970 DOI: 10.1001/jamaoncol.2024.3104] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/15/2024] [Indexed: 09/04/2024]
Abstract
Importance Immune checkpoint inhibitors (ICIs) have revolutionized cancer care; however, accompanying immune-related adverse events (irAEs) confer substantial morbidity and occasional mortality. Life-threatening irAEs may require permanent cessation of ICI, even in patients with positive tumor response. Therefore, it is imperative to comprehensively define the spectrum of irAEs to aid individualized decision-making around the initiation of ICI therapy. Objective To define incidence, risk factors, and clinical spectrum of an irreversible and life-threatening irAE: ICI-induced diabetes. Design, Setting, and Participants This cohort study, conducted at an academic integrated health care system examined 14 328 adult patients treated with ICIs, including 64 patients who developed ICI-induced diabetes, from July 2010 to January 2022. The data were analyzed from 2022 to 2023. Cases of ICI-induced diabetes were manually confirmed; detailed clinical phenotyping was performed at diagnosis and 1-year follow-up. For 862 patients, genotyping data were available, and polygenic risk for type 1 diabetes was determined. Main Outcomes and Measures For ICI-induced diabetes cases and controls, demographic characteristics, comorbidities, tumor category, and ICI category were compared. Among ICI-induced diabetes cases, markers of glycemic physiology were examined at diagnosis and 1-year follow-up. For patients with available genotyping, a published type 1 diabetes polygenic score (T1D GRS2) was calculated. Results Of 14 328 participants, 6571 (45.9%) were women, and the median (range) age was 66 (8-106) years. The prevalence of ICI-induced diabetes among ICI-treated patients was 0.45% (64 of 14 328), with an incidence of 124.8 per 100 000 person-years. Preexisting type 2 diabetes (odds ratio [OR], 5.91; 95% CI, 3.34-10.45) and treatment with combination ICI (OR, 2.57; 95% CI, 1.44-4.59) were significant clinical risk factors of ICI-induced diabetes. T1D GRS2 was associated with ICI-induced diabetes risk, with an OR of 4.4 (95% CI, 1.8-10.5) for patients in the top decile of T1D GRS2, demonstrating a genetic association between spontaneous autoimmunity and irAEs. Patients with ICI-induced diabetes were in 3 distinct phenotypic categories based on autoantibodies and residual pancreatic function, with varying severity of initial presentation. Conclusions and Relevance The results of this analysis of 14 328 ICI-treated patients followed up from ICI initiation determined the incidence, risk factors and clinical spectrum of ICI-induced diabetes. Widespread implementation of this approach across organ-specific irAEs may enhance diagnosis and management of these conditions, and this becomes especially pertinent as ICI treatment rapidly expands to treat a wide spectrum of cancers and is used at earlier stages of treatment.
Collapse
Affiliation(s)
- Karina N. Ruiz-Esteves
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Kaitlyn R. Shank
- Department of Medicine, Massachusetts General Hospital and Department of Medicine, Brigham and Women’s Hospital, Boston
| | - Aaron J. Deutsch
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alekhya Gunturi
- Department of Medicine, Massachusetts General Hospital and Boston University School of Medicine, Boston
| | - Natalia Chamorro-Pareja
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Caitlin A. Colling
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | - Leyre Zubiri
- Department of Medicine, Massachusetts General Hospital, Boston, MA and Harvard Medical School, Boston
| | | | - Tianqi Ouyang
- Department of Medicine, Massachusetts General Hospital, Boston
| | - Alexandra-Chloé Villani
- Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard University, Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Jose C. Florez
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Alexander Gusev
- Division of Population Sciences, Dana-Farber Cancer Institute and Harvard Medical School, Broad Institute, Cambridge, Massachusetts
- Division of Genetics, Brigham and Women’s Hospital and Harvard Medical School, Boston
| | - Kerry L. Reynolds
- Department of Medicine, Mass General Cancer Center, Massachusetts General Hospital, Boston
| | - Karen K. Miller
- Neuroendocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston
| | - Miriam S. Udler
- Department of Medicine and Center for Genomic Medicine, Massachusetts General Hospital, Programs in Metabolism and Medical and Population Genetics, Broad Institute, Cambridge
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Meghan E. Sise
- Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston
| | - Michelle Rengarajan
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Broad Institute of Massachusetts Institute of Technology and Harvard University, Boston
| |
Collapse
|
26
|
Lee KK, Bain T, Flapan A. Cardiogenic shock secondary to immune checkpoint inhibitor associated myocarditis. J R Coll Physicians Edinb 2024; 54:225-230. [PMID: 39175216 DOI: 10.1177/14782715241276900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Immune checkpoint inhibitors have transformed the treatment for multiple cancers and are increasingly used in recent years, but they can cause potentially life-threatening cardiac toxicity. We report a case of a 64-year-old gentleman who presented to the Emergency Department with symptoms of fatigue and breathlessness whilst receiving treatment with an immune checkpoint inhibitor, pembrolizumab, for cholangiocarcinoma. He was found to be in cardiogenic shock with an abnormal electrocardiogram and elevated cardiac troponin at presentation. Echocardiogram demonstrated severely impaired right and left ventricular function. Computed tomography pulmonary angiography and invasive coronary angiography excluded pulmonary embolism and acute myocardial infarction, respectively, and he was diagnosed with immune checkpoint inhibitor associated myocarditis. He was treated with high-dose methylprednisolone and a dobutamine infusion. Within days, his troponin and C-reactive protein levels decreased, and his left ventricular function improved. He was established on heart failure therapies and discharged from hospital 12 days later.
Collapse
Affiliation(s)
- Kuan Ken Lee
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Tanith Bain
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| | - Andrew Flapan
- Edinburgh Heart Centre, Royal Infirmary of Edinburgh, Edinburgh, UK
| |
Collapse
|
27
|
Power JR, Dolladille C, Ozbay B, Procureur AMD, Ederhy S, Palaskas NL, Lehmann LH, Cautela J, Courand PY, Hayek SS, Zhu H, Zaha VG, Cheng RK, Alexandre J, Roubille F, Baldassarre LA, Chen YC, Baik AH, Laufer-Perl M, Tamura Y, Asnani A, Francis S, Gaughan EM, Rainer PP, Bailly G, Flint D, Arangalage D, Cariou E, Florido R, Narezkina A, Liu Y, Sandhu S, Leong D, Issa N, Piriou N, Heinzerling L, Peretto G, Crusz SM, Akhter N, Levenson JE, Turker I, Eslami A, Fenioux C, Moliner P, Obeid M, Chan WT, Ewer SM, Kassaian SE, International ICI-Myocarditis Registry, Johnson DB, Nohria A, Zadok OIB, Moslehi JJ, Salem JE. Predictors and Risk Score for Immune Checkpoint-Inhibitor-Associated Myocarditis Severity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.02.24308336. [PMID: 38883792 PMCID: PMC11177901 DOI: 10.1101/2024.06.02.24308336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Background Immune-checkpoint inhibitors (ICI) are associated with life-threatening myocarditis but milder presentations are increasingly recognized. The same autoimmune process that causes ICI-myocarditis can manifest concurrent generalized myositis, myasthenia-like syndrome, and respiratory muscle failure. Prognostic factors for this "cardiomyotoxicity" are lacking. Methods A multicenter registry collected data retrospectively from 17 countries between 2014-2023. A multivariable cox regression model (hazard-ratio(HR), [95%confidence-interval]) was used to determine risk factors for the primary composite outcome: severe arrhythmia, heart failure, respiratory muscle failure, and/or cardiomyotoxicity-related death. Covariates included demographics, comorbidities, cardio-muscular symptoms, diagnostics, and treatments. Time-dependent covariates were used and missing data were imputed. A point-based prognostic risk score was derived and externally validated. Results In 748 patients (67% male, age 23-94), 30-days incidence of the primary composite outcome, cardiomyotoxic death, and overall death were 33%, 13%, and 17% respectively. By multivariable analysis, the primary composite outcome was associated with active thymoma (HR=3.60[1.93-6.72]), presence of cardio-muscular symptoms (HR=2.60 [1.58-4.28]), low QRS-voltage on presenting electrocardiogram (HR for ≤0.5mV versus >1mV=2.08[1.31-3.30]), left ventricular ejection fraction (LVEF) <50% (HR=1.78[1.22-2.60]), and incremental troponin elevation (HR=1.86 [1.44-2.39], 2.99[1.91-4.65], 4.80[2.54-9.08], for 20, 200 and 2000-fold above upper reference limit, respectively). A prognostic risk score developed using these parameters showed good performance; 30-days primary outcome incidence increased gradually from 3.9%(risk-score=0) to 81.3%(risk-score≥4). This risk-score was externally validated in two independent French and US cohorts. This risk score was used prospectively in the external French cohort to identify low risk patients who were managed with no immunosuppression resulting in no cardio-myotoxic events. Conclusions ICI-myocarditis can manifest with high morbidity and mortality. Myocarditis severity is associated with magnitude of troponin, thymoma, low-QRS voltage, depressed LVEF, and cardio-muscular symptoms. A risk-score incorporating these features performed well. Trial registration number NCT04294771 and NCT05454527.
Collapse
Affiliation(s)
| | - Charles Dolladille
- Sorbonne University, APHP, INSERM, CIC-1901, Hopital Pitié-Salpétrière, Paris, France
| | | | - Adrien MD Procureur
- Sorbonne University, APHP, INSERM, CIC-1901, Hopital Pitié-Salpétrière, Paris, France
| | - Stephane Ederhy
- Sorbonne University, APHP, INSERM, CIC-1901, Hopital Pitié-Salpétrière, Paris, France
| | | | | | - Jennifer Cautela
- Aix-Marseille University; University Mediterranean Center of Cardio-Oncology ; North Hospital, Assistance Publique – Hôpitaux de Marseille
| | | | | | | | | | | | - Joachim Alexandre
- Normandie Univ, UNICAEN, INSERM U1086 ANTICIPE; Caen-Normandy University Hospital
| | | | | | | | | | - Michal Laufer-Perl
- Tel Aviv Sourasky Medical Center, School of Medicine, Tel Aviv University
| | | | | | | | | | - Peter P. Rainer
- Medical University of Graz; BioTechMed Graz; St. Johann in Tirol General Hospital
| | | | | | | | | | | | | | - Yan Liu
- University of Texas at Austin Ascension Seton
| | | | | | | | | | | | - Giovanni Peretto
- Disease Unit for Myocarditis and Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Scientific Institute
| | | | | | | | | | - Assié Eslami
- Hôpital Européen Georges Pompidou, Université Paris Cité
| | | | - Pedro Moliner
- Bellvitge University Hospital - Catalan Institute of Oncology, IDIBELL, CIBERCV
| | | | | | - Stephen M. Ewer
- University of Wisconsin School of Medicine and Public Health
| | | | | | | | | | | | | | - Joe-Elie Salem
- Sorbonne University, APHP, INSERM, CIC-1901, Hopital Pitié-Salpétrière, Paris, France
| |
Collapse
|
28
|
Abstract
In addition to conventional chemoradiation and targeted cancer therapy, the use of immune based therapies, specifically immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T cell therapy (CAR-T), has increased exponentially across a wide spectrum of cancers. This has been paralleled by recognition of off-target immune related adverse events that can affect almost any organ system including the cardiovascular system. The use of ICIs has been associated with myocarditis, a less common but highly fatal adverse effect, pericarditis and pericardial effusions, vasculitis, thromboembolism, and potentially accelerated atherosclerosis. CAR-T resulting in a systemic cytokine release syndrome has been associated with myriad cardiovascular consequences including arrhythmias, myocardial infarction, and heart failure. This review summarizes the current state of knowledge regarding adverse cardiovascular effects associated with ICIs and CAR-T.
Collapse
Affiliation(s)
| | - Hyeon-Ju Ali
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Sarju Ganatra
- Lahey Hospital and Medical Center, Burlington, MA 01805
| | - Anita Deswal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA 01805
| |
Collapse
|
29
|
Adhikari J, Sharma P, Karnabi E, Merchan JH, Arshad H. Case: A Case of Pembrolizumab Induced Myocarditis and Complete Heart Block. J Community Hosp Intern Med Perspect 2024; 14:77-80. [PMID: 39036583 PMCID: PMC11259476 DOI: 10.55729/2000-9666.1329] [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: 07/04/2023] [Revised: 01/23/2024] [Accepted: 02/07/2024] [Indexed: 07/23/2024] Open
Abstract
Use of checkpoint inhibitor pembrolizumab has continued to grow since its approval in 2014. Rare instances of conduction side effects have been described including brady and tachyarrhythmias, heart block and even cardiac arrest. We present a case of Pembrolizumab induced myocarditis and persistent third-degree heart block.
Collapse
Affiliation(s)
- Janak Adhikari
- Department of Internal Medicine, Eastern Maine Medical Center, Bangor, ME, USA
| | | | - Eddy Karnabi
- Department of Cardiology, Eastern Maine Medical Center, Bangor, ME, USA
| | - Juan H Merchan
- Department of Cardiology, Eastern Maine Medical Center, Bangor, ME, USA
| | - Hasnain Arshad
- Department of Neurology, Eastern Maine Medical Center, Bangor, ME, USA
| |
Collapse
|
30
|
Zheng Y, Chen Z, Song W, Xu Y, Zhao Z, Sun Y, Wang Y, Geng X, Zhao J, Zhang X, Xu Y, Chan JSK, Tse G, Li G, Hong L, Liu T. Cardiovascular adverse events associated with immune checkpoint inhibitors: A retrospective multicenter cohort study. Cancer Med 2024; 13:e7233. [PMID: 38752474 PMCID: PMC11097245 DOI: 10.1002/cam4.7233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Over the past decade, immune checkpoint inhibitors (ICIs) have significantly transformed cancer treatment. However, ICIs inevitably may cause a spectrum of immune-related adverse events, among which cardiovascular toxicity, particularly myocarditis, while infrequent, has garnered increasing attention due to its high fatality rate. METHODS We conducted a multicenter retrospective study to characterize ICI-associated cardiovascular adverse events. Logistic regression was performed to explore the risk factors for the development of myocarditis and severe myocarditis. Receiver operating characteristic curves were conducted to assess the diagnostic abilities of cardiac biomarkers to distinguish different cardiovascular toxicities, and the performance and calibration were evaluated using Hosmer-Lemeshow test. RESULTS Forty-four patients were identified, including thirty-five myocarditis, five heart failure, three arrhythmias, and one myocardial infarction. Compared with other patients, myocarditis patients had higher cardiac troponin-I (cTnI) levels (p < 0.001), higher creatine kinase levels (p = 0.003), higher creatine kinase isoenzyme-MB (CK-MB) levels (p = 0.013), and shorter time to the incidence of adverse cardiovascular events (p = 0.022) after ICI treatment. Twenty-one patients (60%) were classified as severe myocarditis, and they presented higher cardiac troponin I (cTnI) levels (p = 0.013), higher N-terminal pro-B-type natriuretic peptide levels (p = 0.031), higher creatine kinase levels (p = 0.018), higher CK-MB levels (p = 0.026), and higher neutrophil to lymphocyte ratio (NLR) levels (p = 0.016) compared to non-severe myocarditis patients after ICI treatment. Multivariate logistic regression showed that CK-MB (adjusted odds ratio [OR]: 1.775, 95% confidence interval [CI]: 1.055-2.984, p = 0.031) was the independent risk factor of the development of ICI-associated myocarditis, and cTnI (adjusted OR: 1.021, 95% CI: 1.002-1.039, p = 0.03) and NLR (adjusted OR: 1.890, 95% CI: 1.026-3.483, p = 0.041) were the independent risk factors of ICI-associated severe myocarditis. The receiver operating characteristic curve showed an area under curve of 0.785 (95% CI: 0.642 to 0.928, p = 0.013) for CK-MB, 0.765 (95% CI: 0.601 to 0.929, p = 0.013) for cTnI, and 0.773 for NLR (95% CI: 0.597 to 0.948, p = 0.016). CONCLUSIONS Elevated CK-MB after ICI treatment is the independent risk factor for the incidence of ICI-associated myocarditis, and elevated cTnI and NLR after ICI treatment are the independent risk factors for the development of ICI-associated severe myocarditis. CK-MB, cTnI, and NLR demonstrated a promising predictive utility for the identification of ICI-associated myocarditis and severe myocarditis.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Ziliang Chen
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Wenhua Song
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yu Xu
- Department of OncologyTianjin Huanghe HospitalTianjinChina
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yihong Sun
- Department of CardiologyChina‐Japan Friendship HospitalBeijingChina
| | - Yuanyuan Wang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Xuhong Geng
- Department of FunctionFourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jun Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Xiaowei Zhang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yanmin Xu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | | | - Gary Tse
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
- School of Nursing and Health StudiesHong Kong Metropolitan UniversityHong KongChina
- Cardiac Electrophysiology Unit, Cardiovascular Analytics GroupPowerHealth LimitedHong KongChina
| | - Guangping Li
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Lili Hong
- Department of OncologyTianjin Huanghe HospitalTianjinChina
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
31
|
Liang Y, Maeda O, Ando Y. Biomarkers for immune-related adverse events in cancer patients treated with immune checkpoint inhibitors. Jpn J Clin Oncol 2024; 54:365-375. [PMID: 38183211 PMCID: PMC11771318 DOI: 10.1093/jjco/hyad184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Although immune checkpoint inhibitors have greatly improved cancer therapy, they also cause immune-related adverse events, including a wide range of inflammatory side effects resulting from excessive immune activation. Types of immune-related adverse events are diverse and can occur in almost any organ, with different frequencies and severities. Furthermore, immune-related adverse events may occur within the first few weeks after treatment or even several months after treatment discontinuation. Predictive biomarkers include blood cell counts and cell surface markers, serum proteins, autoantibodies, cytokines/chemokines, germline genetic variations and gene expression profiles, human leukocyte antigen genotype, microRNAs and the gut microbiome. Given the inconsistencies in research results and limited practical utility, there is to date no established biomarker that can be used in routine clinical practice, and additional investigations are essential to demonstrate efficacy and subsequently facilitate integration into routine clinical use.
Collapse
Affiliation(s)
- Yao Liang
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Osamu Maeda
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan
| | - Yuichi Ando
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital, Nagoya, Aichi, Japan
| |
Collapse
|
32
|
Vasbinder A, Anderson E, Catalan T, Ismail A, Banerjee M, Pizzo I, Machado K, Blakely P, Salem JE, Hayek SS. Incidence of Immune Checkpoint Inhibitor-Induced Myocarditis During the COVID-19 Pandemic. J Am Heart Assoc 2024; 13:e032667. [PMID: 38497479 PMCID: PMC11010026 DOI: 10.1161/jaha.123.032667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 02/23/2024] [Indexed: 03/19/2024]
Affiliation(s)
- Alexi Vasbinder
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Elizabeth Anderson
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Tonimarie Catalan
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Anis Ismail
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Mousumi Banerjee
- Department of Biostatistics, School of Public Health University of Michigan Ann Arbor MI USA
| | - Ian Pizzo
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Kristen Machado
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Pennelope Blakely
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| | - Joe-Elie Salem
- Department of Pharmacology Sorbonne Université, INSERM, AP-HP, CIC-1901, Pitié-Salpêtrière Hospital Paris France
| | - Salim S Hayek
- Department of Internal Medicine, Division of Cardiology University of Michigan Ann Arbor MI USA
| |
Collapse
|
33
|
Zhuang Y, An Q, Wang F, Han D, Qiao Z, Jiang Q, Liu M, Li Y, Shangguan J, Bi X, Shen D. The role of circulating biomarkers in predicting the 30-day mortality of immune checkpoint inhibitors-related myocarditis: a retrospective cohort study. Intern Emerg Med 2024; 19:377-389. [PMID: 38085435 DOI: 10.1007/s11739-023-03481-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 11/07/2023] [Indexed: 03/21/2024]
Abstract
Immune checkpoint inhibitors-related myocarditis (ICIs-M) is a rare and highly lethal immune-related adverse events (irAEs) in common irAEs. This study aims to find circulating biomarkers that can reflect disease state and prognosis accurately. 48 patients with ICIs-M were enrolled according to the diagnostic criteria for ICIs-related myocarditis. For all enrolled patients, valuable information was extracted retrospectively from the medical system, mainly including demographic information, tumor information and laboratory examination. The follow-up period was defined as 30 days after the first diagnosis of ICIs-M. In this study, the 30-day mortality rate of ICIs-M was 24.4%. After adjusting for potential confounding factors using multivariate analysis tools, we demonstrated the excellent performance of biomarkers in predicting 30-day mortality in patients with ICIs-M, including PLT (hazard ratio (HR), 1.07; 95% confidence interval (95%CI), 1.01-1.14; p = 0.028), ALT (HR, 1.23; 95%CI, 1.06-1.41; p = 0.005), AST(HR, 1.06; 95%CI, 1.01-1.10; p = 0.015), LDH (HR, 1.15; 95%CI, 1.04-1.26; p = 0.004), troponin I(HR, 1.44; 95%CI, 1.09-1.89; p = 0.009), PLR (blood plate/lymphocyte) (HR, 1.04; 95% CI, 1.01-1.07; p = 0.024), LAR (lactate dehydrogenase/albumin) (HR, 1.05; 95%CI, 1.01-1.09; p = 0.012), and AAR (aspartate transaminase/albumin) (HR, 1.18; 95%CI, 1.00-1.39; p = 0.048). The analysis of the receiver operating characteristic showed that biomarkers with area under curve (AUC) greater than or equal to 0.80 were LDH (cutoff value, 724.5; AUC, 0.86; 95%CI, 0.75-0.97), LAR (cutoff value, 18.11; AUC, 0.87; 95%CI, 0.76-0.97), troponin I (cutoff value, 0.87; AUC, 0.80; 95%CI, 0.62-0.99), and AAR(cutoff value, 1.52; AUC, 0.80; 95%CI, 0.61-0.98). LDH, LAR, troponin I, and AAR are a group of promising biomarkers that demonstrate excellent predictive ability in predicting the 30-day mortality rate of immune-related myocarditis.
Collapse
Affiliation(s)
- Yuansong Zhuang
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Quanxu An
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Fuhang Wang
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Dongjian Han
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhentao Qiao
- Department of Vascular and Endovascular Surgery, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Qingjiao Jiang
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Miaomiao Liu
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Yuhang Li
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Jiahong Shangguan
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Xuanye Bi
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China
| | - Deliang Shen
- Cardiology Department, Key Laboratory of Cardiac Injury and Repair of Henan Province, First Affiliated Hospital of Zhengzhou University, Henan, China.
| |
Collapse
|
34
|
Maayah M, Grubman S, Allen S, Ye Z, Park DY, Vemmou E, Gokhan I, Sun WW, Possick S, Kwan JM, Gandhi PU, Hu JR. Clinical Interpretation of Serum Troponin in the Era of High-Sensitivity Testing. Diagnostics (Basel) 2024; 14:503. [PMID: 38472975 DOI: 10.3390/diagnostics14050503] [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: 01/01/2024] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Cardiac troponin (Tn) plays a central role in the evaluation of patients with angina presenting with acute coronary syndrome. The advent of high-sensitivity assays has improved the analytic sensitivity and precision of serum Tn measurement, but this advancement has come at the cost of poorer specificity. The role of clinical judgment is of heightened importance because, more so than ever, the interpretation of serum Tn elevation hinges on the careful integration of findings from electrocardiographic, echocardiographic, physical exam, interview, and other imaging and laboratory data to formulate a weighted differential diagnosis. A thorough understanding of the epidemiology, mechanisms, and prognostic implications of Tn elevations in each cardiac and non-cardiac etiology allows the clinician to better distinguish between presentations of myocardial ischemia and myocardial injury-an important discernment to make, as the treatment of acute coronary syndrome is vastly different from the workup and management of myocardial injury and should be directed at the underlying cause.
Collapse
Affiliation(s)
- Marah Maayah
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Scott Grubman
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Stephanie Allen
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Zachary Ye
- Department of Internal Medicine, Temple University Medical Center, Philadelphia, PA 19140, USA
| | - Dae Yong Park
- Department of Internal Medicine, Cook County Hospital, Chicago, IL 60612, USA
| | - Evangelia Vemmou
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Ilhan Gokhan
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Wendy W Sun
- Department of Emergency Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Stephen Possick
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Parul U Gandhi
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Cardiology, Veterans Affairs Connecticut Health Care System, West Haven, CT 06516, USA
| | - Jiun-Ruey Hu
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
35
|
Murtagh G, deFilippi C, Zhao Q, Barac A. Circulating biomarkers in the diagnosis and prognosis of immune checkpoint inhibitor-related myocarditis: time for a risk-based approach. Front Cardiovasc Med 2024; 11:1350585. [PMID: 38410245 PMCID: PMC10894940 DOI: 10.3389/fcvm.2024.1350585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that block immune checkpoints and therefore activate immune cells, allowing them to recognize and attack cancer cells. ICIs have revolutionized oncology practice, but their use has been complicated by immune-related adverse events (irAEs). Of cardiovascular (CV) irAEs, ICI-related myocarditis has received significant attention due to high mortality rates, ranging from 25% to 50%, despite its overall low incidence. Establishing the early diagnosis of ICI-myocarditis is important for early initiation of steroids and consideration of hospitalization in patients who are at risk for hemodynamic compromise and need high acuity care in a tertiary setting. In this review, we summarize the diagnostic and prognostic tools for ICI-myocarditis, including electrocardiography, echocardiography, cardiac magnetic resonance imaging, with emphasis on circulating biomarkers. Cardiac troponins (cTns) are an essential component of the diagnosis of ICI-myocarditis, and we provide a summary of the recent studies that utilized different assays (cTnI vs. cTnT) and outcomes (diagnosis vs. prognosis including major adverse cardiac outcomes). With the exponential increase in ICI use across different oncology indications, there is a major need to include biomarkers in risk stratification to guide diagnosis and treatment. Our review proposes a framework for future multisite registries, including cTn evaluation at baseline and at the time of irAE suspicion, with development of central biobanking to allow head-to-head evaluation and clinical validation of different biomarker assays in ICI-myocarditis. This approach, with the inclusion of CV biomarkers into clinical and pragmatic oncology trials, holds promise to improve the early recognition and management of ICI-myocarditis and CV irAEs, thus leading to better outcomes.
Collapse
Affiliation(s)
- Gillian Murtagh
- Core Diagnostics, Abbott Laboratories, Abbott Park, IL, United States
| | | | - Qiong Zhao
- Inova Schar Heart and Vascular, Falls Church, VA, United States
| | - Ana Barac
- Inova Schar Heart and Vascular, Falls Church, VA, United States
| |
Collapse
|
36
|
Rong Y, Bentley JP, Bhattacharya K, Yang Y, Chang Y, Earl S, Ramachandran S. Incidence and risk factors of immune-related adverse events induced by immune checkpoint inhibitors among older adults with non-small cell lung cancer. Cancer Med 2024; 13:e6879. [PMID: 38164655 PMCID: PMC10807682 DOI: 10.1002/cam4.6879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/31/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) treatment has been linked to a variety of immune-related adverse events (irAEs), which can affect any organ system. The incidence and risk factors of irAEs have not been adequately evaluated among older adults with NSCLC. METHODS A cohort study was conducted using 1999-2019 SEER-Medicare data among beneficiaries aged ≥65 years with a diagnosis of NSCLC who received nivolumab, pembrolizumab, or atezolizumab. Incident irAEs were identified post-ICI initiation. Demographic, cancer-related characteristics, and clinical history risk factors of irAEs were evaluated with competing events considered. RESULTS A total of 8175 older NSCLC patients were included (with 46.8% experiencing irAEs). Pneumonitis (16.5%), hypothyroidism (10.5%), arrhythmia (11.18%), and acute kidney injury (AKI) (5.8%) were the most common irAEs. The median time to first irAE was 82 days (IQR: 29-182 days). The earliest onset of irAE occurrence was for hematologic irAEs, while the latest were gastrointestinal, dermatologic, and musculoskeletal irAEs. Fine-Gray regression modeling revealed significantly greater hazards of irAE occurrence in patients who received pembrolizumab at index, did not have CNS metastases, had a history of autoimmune disorder, and had chemotherapy in combination with ICI. Race, socioeconomic status, previous radiation therapy, and comorbidity burden were found to be associated with the occurrence of certain type of irAEs. CONCLUSION A significant proportion of older patients with NSCLC develop an irAE after receiving ICI treatment. Factors related to cancer and treatment as well as demographics contribute to the increased risk of irAEs. Close monitoring and prediction of irAE among older patients receiving ICI is warranted.
Collapse
Affiliation(s)
- Yiran Rong
- Department of Pharmacy AdministrationUniversity of MississippiUniversityMississippiUSA
| | - John P. Bentley
- Department of Pharmacy AdministrationUniversity of MississippiUniversityMississippiUSA
- Center for Pharmaceutical Marketing and ManagementCenter for Pharmaceutical Marketing and ManagementUniversity of MississippiUniversityMississippiUSA
| | - Kaustuv Bhattacharya
- Department of Pharmacy AdministrationUniversity of MississippiUniversityMississippiUSA
- Center for Pharmaceutical Marketing and ManagementCenter for Pharmaceutical Marketing and ManagementUniversity of MississippiUniversityMississippiUSA
| | - Yi Yang
- Department of Pharmacy AdministrationUniversity of MississippiUniversityMississippiUSA
| | - Yunhee Chang
- Department of Nutrition and Hospitality ManagementUniversity of MississippiUniversityMississippiUSA
| | - Sally Earl
- Department of Pharmacy PracticeUniversity of MississippiUniversityMississippiUSA
| | - Sujith Ramachandran
- Department of Pharmacy AdministrationUniversity of MississippiUniversityMississippiUSA
- Center for Pharmaceutical Marketing and ManagementCenter for Pharmaceutical Marketing and ManagementUniversity of MississippiUniversityMississippiUSA
| |
Collapse
|
37
|
Talukder S, Ghose A, Chakraborty T, Olsson-Brown A, Ramalingam S, Rosen SD, Young K, Lyon AR, Ghosh AK. Evolving cardiac biomarkers for immune checkpoint inhibitor related myocarditis in cancer patients. IJC HEART & VASCULATURE 2023; 49:101278. [PMID: 37842144 PMCID: PMC10570005 DOI: 10.1016/j.ijcha.2023.101278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/04/2023] [Indexed: 10/17/2023]
Affiliation(s)
- Suprateeka Talukder
- Norfolk and Norwich University Hospital, Norfolk and Norwich University Hospitals NHS Foundation Trust, Norwich, UK
- Faculty of Medicine and Health Sciences, University of East Anglia, Norwich, UK
| | - Aruni Ghose
- Barts Cancer Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Cardio-Oncology Service, Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, London, UK
- Medical Oncology, Medway NHS Foundation Trust, Gillingham, Kent, UK
- Immuno-Oncology Clinical Network, UK
| | - Turja Chakraborty
- London Northwest University Healthcare NHS Trust, London, UK
- National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, UK
| | - Anna Olsson-Brown
- Immuno-Oncology Clinical Network, UK
- Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, UK
- Department of Molecular and Clinical Pharmacology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Stuart D. Rosen
- Immuno-Oncology Clinical Network, UK
- London Northwest University Healthcare NHS Trust, London, UK
- Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, UK
| | - Kate Young
- The Royal Marsden Hospital, The Royal Marsden NHS Foundation Trust, London, UK
| | - Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, UK
| | - Arjun K. Ghosh
- Cardio-Oncology Service, Barts Heart Centre, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
- Cardio-Oncology Service, University College London Hospitals NHS Foundation Trust, London, UK
- Hatter Cardiovascular Institute, UCL Institute of Cardiovascular Science, University College London, London, UK
| |
Collapse
|
38
|
Fenioux C, Abbar B, Boussouar S, Bretagne M, Power JR, Moslehi JJ, Gougis P, Amelin D, Dechartres A, Lehmann LH, Courand PY, Cautela J, Alexandre J, Procureur A, Rozes A, Leonard-Louis S, Qin J, Cheynier R, Charmeteau-De Muylder B, Redheuil A, Tubach F, Cadranel J, Milon A, Ederhy S, Similowski T, Johnson DB, Pizzo I, Catalan T, Benveniste O, Hayek SS, Allenbach Y, Rosenzwajg M, Dolladille C, Salem JE. Thymus alterations and susceptibility to immune checkpoint inhibitor myocarditis. Nat Med 2023; 29:3100-3110. [PMID: 37884625 DOI: 10.1038/s41591-023-02591-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 09/15/2023] [Indexed: 10/28/2023]
Abstract
Immune checkpoint inhibitors (ICI) have transformed the therapeutic landscape in oncology. However, ICI can induce uncommon life-threatening autoimmune T-cell-mediated myotoxicities, including myocarditis and myositis. The thymus plays a critical role in T cell maturation. Here we demonstrate that thymic alterations are associated with increased incidence and severity of ICI myotoxicities. First, using the international pharmacovigilance database VigiBase, the Assistance Publique Hôpitaux de Paris-Sorbonne University data warehouse (Paris, France) and a meta-analysis of clinical trials, we show that ICI treatment of thymic epithelial tumors (TET, and particularly thymoma) was more frequently associated with ICI myotoxicities than other ICI-treated cancers. Second, in an international ICI myocarditis registry, we established that myocarditis occurred earlier after ICI initiation in patients with TET (including active or prior history of TET) compared to other cancers and was more severe in terms of life-threatening arrythmias and concurrent myositis, leading to respiratory muscle failure and death. Lastly, we show that presence of anti-acetylcholine-receptor antibodies (a biological proxy of thymic-associated autoimmunity) was more prevalent in patients with ICI myocarditis than in ICI-treated control patients. Altogether, our results highlight that thymic alterations are associated with incidence and seriousness of ICI myotoxicities. Clinico-radio-biological workup evaluating the thymus may help in predicting ICI myotoxicities.
Collapse
Affiliation(s)
- Charlotte Fenioux
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
- Hôpitaux de Paris, Henri Mondor Hospital, Department of Oncology, Créteil, France
| | - Baptiste Abbar
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
- Hôpitaux de Paris, Pitié Salpêtrière Hospital, Department of Oncology, Paris, France
| | - Samia Boussouar
- Hôpitaux de Paris, Pitié Salpêtrière Hospital, Department of Radiology, Paris, France
| | - Marie Bretagne
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - John R Power
- Department of Medecine, University of California, San Diego, San Diego, CA, USA
| | - Javid J Moslehi
- Department of Medecine, University of California, San Francisco, San Francisco, CA, USA
| | - Paul Gougis
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - Damien Amelin
- Sorbonne Université, INSERM, Association Institut de Myologie, Center of Research in Myology, UMRS 974, Paris, France
| | - Agnès Dechartres
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Unité de Recherche Clinique PSL-CFX, CIC-1901, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Lorenz H Lehmann
- Department of Cardiology, University Hospital Heidelberg; German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Pierre-Yves Courand
- Fédération de Cardiologie, IMMUCARE, Hôpital de La Croix-Rousse Et Hôpital Lyon Sud, Hospices Civils de Lyon; Université de Lyon, CREATIS UMR INSERM U1044, INSA, Lyon, France
| | - Jennifer Cautela
- Aix-Marseille University, University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Center for Cardiovascular and Nutrition Research, INSERM 1263, INRAE 1260, Nord Hospital, Assistance Publique-Hôpitaux de Marseille, Paris, France
| | - Joachim Alexandre
- CHU de Caen Normandie, Department of Pharmacology, Pharmacoepidemiology Unit; Normandie Université, UNICAEN, INSERM U1086 ANTICIPE Centre François Baclesse, Caen, France
| | - Adrien Procureur
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - Antoine Rozes
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Unité de Recherche Clinique PSL-CFX, CIC-1901, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Sarah Leonard-Louis
- Hôpitaux de Paris, Pitié Salpêtrière Hospital, Laboratoire de Neuropathologie, Paris, France
| | - Juan Qin
- Department of Medecine, University of California, San Francisco, San Francisco, CA, USA
| | - Rémi Cheynier
- Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | | | - Alban Redheuil
- Hôpitaux de Paris, Pitié Salpêtrière Hospital, Department of Radiology, Paris, France
| | - Florence Tubach
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie et de Santé Publique, Département de Santé Publique, Centre de Pharmacoépidémiologie (Cephepi), Unité de Recherche Clinique PSL-CFX, CIC-1901, AP-HP, Hôpital Pitié Salpêtrière, Paris, France
| | - Jacques Cadranel
- Hôpitaux de Paris, Tenon Hospital, Department of Pulmonology and Thoracic Oncology and GRC Theranoscan Sorbonne University, Paris, France
| | - Audrey Milon
- Hôpitaux de Paris, Tenon Hospital, Department of Radiology, Paris, France
| | - Stéphane Ederhy
- Hôpitaux de Paris, Saint-Antoine Hospital, Department of Cardiology, Paris, France
| | - Thomas Similowski
- Sorbonne Université, INSERM, UMRS 1158 Neurophysiologie respiratoire expérimentale et clinique'; Assistance Publique -Hôpitaux de Paris, Pitié-Salpêtrière Hospital, 'Département R3S', Paris, France
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ian Pizzo
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Toniemarie Catalan
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Olivier Benveniste
- Department of Internal Medicine, Sorbonne University, AP-HP, INSERM UMRS 974, Pitié-Salpêtrière Hospital, Paris, France
| | - Salim S Hayek
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI, USA
| | - Yves Allenbach
- Department of Internal Medicine, Sorbonne University, AP-HP, INSERM UMRS 974, Pitié-Salpêtrière Hospital, Paris, France
| | - Michelle Rosenzwajg
- Hôpitaux de Paris, Pitié Salpêtrière Hospital, Department of Immunology, Paris, France
| | - Charles Dolladille
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France
| | - Joe-Elie Salem
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France.
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
39
|
Lasica R, Djukanovic L, Savic L, Krljanac G, Zdravkovic M, Ristic M, Lasica A, Asanin M, Ristic A. Update on Myocarditis: From Etiology and Clinical Picture to Modern Diagnostics and Methods of Treatment. Diagnostics (Basel) 2023; 13:3073. [PMID: 37835816 PMCID: PMC10572782 DOI: 10.3390/diagnostics13193073] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/22/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Although the frequency of myocarditis in the general population is very difficult to accurately determine due to the large number of asymptomatic cases, the incidence of this disease is increasing significantly due to better defined criteria for diagnosis and the development of modern diagnostic methods. The multitude of different etiological factors, the diversity of the clinical picture, and the variability of the diagnostic findings make this disease often demanding both for the selection of the diagnostic modality and for the proper therapeutic approach. The previously known most common viral etiology of this disease is today overshadowed by new findings based on immune-mediated processes, associated with diseases that in their natural course can lead to myocardial involvement, as well as the iatrogenic cause of myocarditis, which is due to use of immune checkpoint inhibitors in the treatment of cancer patients. Suspecting that a patient with polymorphic and non-specific clinical signs and symptoms, such as changes in ECG and echocardiography readings, has myocarditis is the starting point in the diagnostic algorithm. Cardio magnetic resonance imaging is non-invasive and is the gold standard for diagnosis and clinical follow-up of these patients. Endomyocardial biopsy as an invasive method is the diagnostic choice in life-threatening cases with suspicion of fulminant myocarditis where the diagnosis has not yet established or there is no adequate response to the applied therapeutic regimen. The treatment of myocarditis is increasingly demanding and includes conservative methods of treating heart failure, immunomodulatory and immunospressive therapy, methods of mechanical circulatory support, and heart transplantation. The goal of developing new diagnostic and therapeutic methods is to reduce mortality from this complex disease, which is still high.
Collapse
Affiliation(s)
- Ratko Lasica
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.D.); (L.S.); (G.K.); (M.A.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Lazar Djukanovic
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.D.); (L.S.); (G.K.); (M.A.)
| | - Lidija Savic
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.D.); (L.S.); (G.K.); (M.A.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Gordana Krljanac
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.D.); (L.S.); (G.K.); (M.A.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Marija Zdravkovic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Department of Cardiology, University Medical Center Bezanijska Kosa, 11000 Belgrade, Serbia
| | - Marko Ristic
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| | | | - Milika Asanin
- Department of Cardiology, Emergency Center, University Clinical Center of Serbia, 11000 Belgrade, Serbia; (L.D.); (L.S.); (G.K.); (M.A.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
| | - Arsen Ristic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia;
- Department of Cardiology, University Clinical Center of Serbia, 11000 Belgrade, Serbia;
| |
Collapse
|
40
|
Vasbinder A, Ismail A, Salem JE, Hayek SS. Role of Biomarkers in the Management of Immune-Checkpoint Inhibitor-Related Myocarditis. Curr Cardiol Rep 2023; 25:959-967. [PMID: 37436648 PMCID: PMC11729503 DOI: 10.1007/s11886-023-01915-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitor (ICI)-related myocarditis poses a major clinical challenge given its non-specific presentation, rapid progression, and high mortality rate. Here, we review the role of blood-based biomarkers in the clinical management of patients with ICI-related myocarditis. RECENT FINDINGS Myocardial injury, its unique pattern, and the co-occurrence with myositis are defining features of ICI-related myocarditis. Non-cardiac biomarkers, specifically creatinine phosphokinase, precedes the symptomatic presentation and is highly sensitive for diagnosing ICI-related myocarditis, making them useful screening biomarkers. Combined elevations in cardiac troponins and non-cardiac biomarkers improve the confidence of an ICI myocarditis diagnosis. High troponin and creatinine phosphokinase levels are strongly associated with severe outcomes. We propose biomarker-based algorithms for the monitoring and diagnosis of ICI-related myocarditis. Biomarkers, such as cardiac troponins and creatine phosphokinase, can be used in combination in the monitoring, diagnosis, and prognostication of patients with ICI-related myocarditis.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA
| | - Anis Ismail
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA
| | - Joe-Elie Salem
- Department of Pharmacology and Clinical Investigation Centre, Pitié-Salpetriere Hospital, Sorbonne Universite, Paris, France
| | - Salim S Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
41
|
Lehmann LH, Heckmann MB, Bailly G, Finke D, Procureur A, Power JR, Stein F, Bretagne M, Ederhy S, Fenioux C, Hamwy O, Funck-Brentano E, Romano E, Pieroni L, Münster J, Allenbach Y, Anquetil C, Leonard-Louis S, Palaskas NL, International ICI-myocarditis Registry, Hayek SS, Katus HA, Giannitsis E, Frey N, Kaya Z, Moslehi J, Prifti E, Salem JE. Cardiomuscular Biomarkers in the Diagnosis and Prognostication of Immune Checkpoint Inhibitor Myocarditis. Circulation 2023; 148:473-486. [PMID: 37317858 PMCID: PMC10527069 DOI: 10.1161/circulationaha.123.062405] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 05/19/2023] [Indexed: 06/16/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are approved for multiple cancers but can result in ICI-associated myocarditis, an infrequent but life-threatening condition. Elevations in cardiac biomarkers, specifically troponin-I (cTnI), troponin-T (cTnT), and creatine kinase (CK), are used for diagnosis. However, the association between temporal elevations of these biomarkers with disease trajectory and outcomes has not been established. METHODS We analyzed the diagnostic accuracy and prognostic performances of cTnI, cTnT, and CK in patients with ICI myocarditis (n=60) through 1-year follow-up in 2 cardio-oncology units (APHP Sorbonne, Paris, France and Heidelberg, Germany). A total of 1751 (1 cTnT assay type), 920 (4 cTnI assay types), and 1191 CK sampling time points were available. Major adverse cardiomyotoxic events (MACE) were defined as heart failure, ventricular arrhythmia, atrioventricular or sinus block requiring pacemaker, respiratory muscle failure requiring mechanical ventilation, and sudden cardiac death. Diagnostic performance of cTnI and cTnT was also assessed in an international ICI myocarditis registry. RESULTS Within 72 hours of admission, cTnT, cTnI, and CK were increased compared with upper reference limits (URLs) in 56 of 57 (98%), 37 of 42 ([88%] P=0.03 versus cTnT), and 43 of 57 ([75%] P<0.001 versus cTnT), respectively. This increased rate of positivity for cTnT (93%) versus cTnI ([64%] P<0.001) on admission was confirmed in 87 independent cases from an international registry. In the Franco-German cohort, 24 of 60 (40%) patients developed ≥1 MACE (total, 52; median time to first MACE, 5 [interquartile range, 2-16] days). The highest value of cTnT:URL within the first 72 hours of admission performed best in terms of association with MACE within 90 days (area under the curve, 0.84) than CK:URL (area under the curve, 0.70). A cTnT:URL ≥32 within 72 hours of admission was the best cut-off associated with MACE within 90 days (hazard ratio, 11.1 [95% CI, 3.2-38.0]; P<0.001), after adjustment for age and sex. cTnT was increased in all patients within 72 hours of the first MACE (23 of 23 [100%]), whereas cTnI and CK values were less than the URL in 2 of 19 (11%) and 6 of 22 (27%) of patients (P<0.001), respectively. CONCLUSIONS cTnT is associated with MACE and is sensitive for diagnosis and surveillance in patients with ICI myocarditis. A cTnT:URL ratio <32 within 72 hours of diagnosis is associated with a subgroup at low risk for MACE. Potential differences in diagnostic and prognostic performances between cTnT and cTnI as a function of the assays used deserve further evaluation in ICI myocarditis.
Collapse
Affiliation(s)
- Lorenz H. Lehmann
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Markus B. Heckmann
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Guillaume Bailly
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | - Daniel Finke
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Adrien Procureur
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | - John R. Power
- University of California San Diego, San Diego, California, USA
| | - Frederic Stein
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | - Marie Bretagne
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | - Stephane Ederhy
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris,, INSERM, Department of Cardiology, CIC-1901, UNICO-GRECO Cardio-oncology program, Saint-Antoine Hospital, Paris, France
| | - Charlotte Fenioux
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | - Omar Hamwy
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| | | | - Emanuela Romano
- Center for Cancer Immunotherapy, Department of Oncology, PSL Research University, Institut Curie, Paris, France
| | - Laurence Pieroni
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Biochimie- Hormonologie, Hôpital Tenon, Paris, France
| | - Jan Münster
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Yves Allenbach
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of internal medecine, Hôpital Pitié-Salpêtrière, Paris, France
| | - Céline Anquetil
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of internal medecine, Hôpital Pitié-Salpêtrière, Paris, France
| | - Sarah Leonard-Louis
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Neuropathology, Hôpital Pitié-Salpêtrière, F-75013 Paris, France
| | - Nicolas L. Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| | | | - Salim S. Hayek
- Department of Internal Medicine, Division of Cardiology, University of Michigan, Ann Arbor, MI
| | - Hugo A. Katus
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Evangelos Giannitsis
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Norbert Frey
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Ziya Kaya
- Department of Cardiology, Angiology, and Pneumology, Heidelberg University Hospital, Heidelberg, Germany; German Centre for Cardiovascular Research (DZHK), Heidelberg/Mannheim partner site, Heidelberg, Germany
| | - Javid Moslehi
- Section of Cardio-Oncology & Immunology, Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Edi Prifti
- IRD, Sorbonne University, UMMISCO, 32 Avenue Henri Varagnat, F-93143 Bondy, France; Sorbonne Université, INSERM (NutriOmics), Paris, France
| | - Joe-Elie Salem
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, INSERM, Department of Pharmacology, CIC-1901, UNICO-GRECO Cardio-oncology program, Pitié-salpétrière Hospital, Paris, France
| |
Collapse
|
42
|
Matsumori A. Myocarditis and Autoimmunity. Expert Rev Cardiovasc Ther 2023. [PMID: 37243585 DOI: 10.1080/14779072.2023.2219895] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 05/20/2023] [Accepted: 05/26/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Autoimmune myocarditis may develop due to heterogeneous causes. Myocarditis is often caused by viral infections, but it can also be caused by systemic autoimmune diseases. Immune checkpoint inhibitors and virus vaccines induce immune activation, and they can cause the development of myocarditis, as well as several immune-related adverse events. The development of myocarditis is dependent on the genetic factors of the host, and the major histocompatibility complex (MHC) may be an important determinant of the type and severity of the disease. However, non-MHC immunoregulatory genes may also play a role in determining susceptibility. AREA COVERED This review summarizes the current knowledge of the etiology, pathogenesis, diagnosis and treatment of autoimmune myocarditis with a particular focus on viral infection and autoimmunity, and biomarkers of myocarditis. EXPERT OPINION An endomyocardial biopsy may not be the gold standard for the diagnosis of myocarditis. Cardiac magnetic resonance imaging is useful in diagnosing autoimmune myocarditis. Recently identified biomarkers of inflammation and myocyte injury are promising for the diagnosis of myocarditis when measured simultaneously. Future treatments should focus on the appropriate diagnosis of the etiologic agent, as well as on the specific stage of the evolution of immune and inflammatory processes.
Collapse
Affiliation(s)
- Akira Matsumori
- Clinical Research Institute, National Hospital Organization Kyoto Medical Center, Kyoto 612-8555, Japan
| |
Collapse
|
43
|
Delombaerde D, De Sutter J, Croes L, Vervloet D, Moerman V, Van de Veire N, Willems AM, Wouters K, Peeters M, Prenen H, Vulsteke C. Extensive CArdioVAscular Characterization and Follow-Up of Patients Receiving Immune Checkpoint Inhibitors: A Prospective Multicenter Study. Pharmaceuticals (Basel) 2023; 16:ph16040625. [PMID: 37111382 PMCID: PMC10142365 DOI: 10.3390/ph16040625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The increasing use of immune checkpoint inhibitors (ICIs) in the treatment of both advanced and early stages of various malignancies has resulted in a substantial increase in the incidence of cardiovascular (CV) immune-related adverse events (irAEs). The current follow-up guidelines are based on anecdotal evidence and expert opinions, due to a lack of solid data and prospective studies. As many questions remain unanswered, cardiac monitoring, in patients receiving ICIs, is not always implemented by oncologists. Hence, an urgent need to investigate the possible short- and long-term CV effects of ICIs, as ICI approval is continuing to expand to the (neo)adjuvant setting. METHODS We have initiated a prospective, multicenter study, i.e., the CAVACI trial, in which a minimum of 276 patients with a solid tumor, eligible for ICI treatment, will be enrolled. The study consists of routine investigations of blood parameters (troponin and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels, in particular) and a thorough CV follow-up (electrocardiograms, transthoracic echocardiograms, and coronary calcium scoring) at fixed time points for a total period of two years. The primary endpoint is the cumulative incidence of troponin elevation in the first three months of ICI treatment, compared to baseline levels. Furthermore, secondary endpoints include incidence above the upper limit of normal of both troponin and NT-proBNP levels, evolution in troponin and NT-proBNP levels, the incidence of CV abnormalities/major adverse cardiac events, evaluation of associations between patient characteristics/biochemical parameters and CV events, transthoracic echocardiography parameters, electrocardiography parameters, and progression of coronary atherosclerosis. Recruitment of patients started in January 2022. Enrolment is ongoing in AZ Maria Middelares, Antwerp University Hospital, AZ Sint-Vincentius Deinze, and AZ Sint-Elisabeth Zottegem. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05699915, registered 26 January 2023.
Collapse
Affiliation(s)
- Danielle Delombaerde
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | - Johan De Sutter
- Department of Cardiology, AZ Maria Middelares, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lieselot Croes
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | | | | | - Nico Van de Veire
- Department of Cardiology, AZ Maria Middelares, 9000 Ghent, Belgium
- Department of Cardiology, Free University Brussels, 1000 Brussels, Belgium
| | | | - Kristien Wouters
- Antwerp University Hospital, Clinical Trial Center (CTC), CRC Antwerp, 2650 Edegem, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
- Multidisciplinary Oncologic Center Antwerp (MOCA), Antwerp University Hospital, 2650 Edegem, Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
- Multidisciplinary Oncologic Center Antwerp (MOCA), Antwerp University Hospital, 2650 Edegem, Belgium
| | - Christof Vulsteke
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
44
|
PCSK9 Inhibitors in Cancer Patients Treated with Immune-Checkpoint Inhibitors to Reduce Cardiovascular Events: New Frontiers in Cardioncology. Cancers (Basel) 2023; 15:cancers15051397. [PMID: 36900189 PMCID: PMC10000232 DOI: 10.3390/cancers15051397] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Cancer patients treated with immune checkpoint inhibitors (ICIs) are exposed to a high risk of atherosclerosis and cardiometabolic diseases due to systemic inflammatory conditions and immune-related atheroma destabilization. Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a key protein involved in metabolism of low-density lipoprotein (LDL) cholesterol. PCSK9 blocking agents are clinically available and involve monoclonal antibodies, and SiRNA reduces LDL levels in high-risk patients and atherosclerotic cardiovascular disease events in multiple patient cohorts. Moreover, PCSK9 induces peripheral immune tolerance (inhibition of cancer cell- immune recognition), reduces cardiac mitochondrial metabolism, and enhances cancer cell survival. The present review summarizes the potential benefits of PCSK9 inhibition through selective blocking antibodies and siRNA in patients with cancer, especially in those treated with ICIs therapies, in order to reduce atherosclerotic cardiovascular events and potentially improve ICIs-related anticancer functions.
Collapse
|