ArticlePDF Available

Case report: Electrocardiographic changes in pembrolizumab-induced fatal myocarditis

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Immune checkpoint inhibitor (ICI)-induced myocarditis is rare but fatal. Because of the rapid course of ICI-induced myocarditis, understanding of clinical course is only possible through information from case reports. We report a case of pembrolizumab-induced myocarditis in which we were able to document the course of electrocardiographic changes from onset to death. A 58-year-old woman with stage IV lung adenocarcinoma, who had completed her first cycle of pembrolizumab, carboplatin, and pemetrexed, was admitted with pericardial effusion. She underwent pericardiocentesis after admission. A second cycle of chemotherapy was administered 3 weeks after the first cycle. Twenty-two days after admission, she developed a mild sore throat and tested positive for SARS-CoV-2 antigen. She was diagnosed with mild coronavirus disease 2019 (COVID-19), isolated, and treated with sotrovimab. Thirty-two days after admission, an electrocardiogram showed monomorphic ventricular tachycardia (VT). Suspecting myocarditis caused by pembrolizumab, the patient was started on daily methylprednisolone after coronary angiography and endocardial biopsy. Eight days after the start of methylprednisolone administration, she was considered to have passed the acute stage. However, four days later, R-on-T phenomenon triggered polymorphic VT and she died. The impact of viral infections such as COVID-19 on patients be treated with immune checkpoint inhibitors is still unknown and we need to be careful with systemic management after viral infections.
This content is subject to copyright.
Case report:
Electrocardiographic changes
in pembrolizumab-induced
fatal myocarditis
Kazuhiro Nishiyama
1
*, Kei Morikawa
1
, Yusuke Shinozaki
1
,
Junko Ueno
1
, Satoshi Tanaka
1
, Hajime Tsuruoka
1
,
Shinya Azagami
1
, Atsuko Ishida
1
, Nobuyuki Yanagisawa
2
,
Yoshihiro J. Akashi
3
and Masamichi Mineshita
1
1
Division of Respiratory Medicine, Department of Internal Medicine, St Marianna University School of
Medicine, Kawasaki, Kanagawa, Japan,
2
Department of Pathology, St. Marianna University School of
Medicine, Kawasaki, Kanagawa, Japan,
3
Division of Cardiology, Department of Internal Medicine, St.
Marianna University School of Medicine, Kawasaki, Kanagawa, Japan
Immune checkpoint inhibitor (ICI)-induced myocarditis is rare but fatal. Because
of the rapid course of ICI-induced myocarditis, understanding of clinical course
is only possible through information from case reports. We report a case of
pembrolizumab-induced myocarditis in which we were able to document the
course of electrocardiographic changes from onset to death. A 58-year-old
woman with stage IV lung adenocarcinoma, who had completed her rst cycle of
pembrolizumab, carboplatin, and pemetrexed, was admitted with pericardial
effusion. She underwent pericardiocentesis after admission. A second cycle of
chemotherapy was administered 3 weeks after the rst cycle. Twenty-two days
after admission, she developed a mild sore throat and tested positive for SARS-
CoV-2 antigen. She was diagnosed with mild coronavirus disease 2019 (COVID-
19), isolated, and treated with sotrovimab. Thirty-two days after admission, an
electrocardiogram showed monomorphic ventricular tachycardia (VT).
Suspecting myocarditis caused by pembrolizumab, the patient was started on
daily methylprednisolone after coronary angiography and endocardial biopsy.
Eight days after the start of methylprednisolone administration, she was
considered to have passed the acute stage. However, four days later, R-on-T
phenomenon triggered polymorphic VT and she died. The impact of viral
infections such as COVID-19 on patients be treated with immune checkpoint
inhibitors is still unknown and we need to be careful with systemic management
after viral infections.
KEYWORDS
irAE, lung cancer, myocarditis, pembrolizumab, COVID-19
Frontiers in Immunology frontiersin.org01
OPEN ACCESS
EDITED BY
Emanuele Bobbio,
Sahlgrenska University Hospital, Sweden
REVIEWED BY
Piero Gentile,
Niguarda CaGranda Hospital,
Italy
Gopal Chandra Ghosh,
Rabindranath Thakur Diagnostic and
Medical Care Center, India
Entela Bollano,
Sahlgrenska University Hospital, Sweden
*CORRESPONDENCE
Kazuhiro Nishiyama
kazuhiro.nishiyama@marianna-u.ac.jp
SPECIALTY SECTION
This article was submitted to
Cancer Immunity
and Immunotherapy,
a section of the journal
Frontiers in Immunology
RECEIVED 24 October 2022
ACCEPTED 06 February 2023
PUBLISHED 16 February 2023
CITATION
Nishiyama K, Morikawa K, Shinozaki Y,
Ueno J, Tanaka S, Tsuruoka H, Azagami S,
Ishida A, Yanagisawa N, Akashi YJ and
Mineshita M (2023) Case report:
Electrocardiographic changes in
pembrolizumab-induced fatal myocarditis.
Front. Immunol. 14:1078838.
doi: 10.3389/fimmu.2023.1078838
COPYRIGHT
© 2023 Nishiyama, Morikawa, Shinozaki,
Ueno, Tanaka, Tsuruoka, Azagami, Ishida,
Yanagisawa, Akashi and Mineshita. This is an
open-access article distributed under the
terms of the Creative Commons Attribution
License (CC BY). The use, distribution or
reproduction in other forums is permitted,
provided the original author(s) and the
copyright owner(s) are credited and that
the original publication in this journal is
cited, in accordance with accepted
academic practice. No use, distribution or
reproduction is permitted which does not
comply with these terms.
TYPE Case Report
PUBLISHED 16 February 2023
DOI 10.3389/fimmu.2023.1078838
Introduction
The advent of immune checkpoint inhibitors (ICIs) has
revolutionized cancer treatment. ICIs sustain T-cell activation and
exert the anti-tumor effects by blocking immunosuppressive
signaling from antigen-presenting cells and tumor cells (1).
Currently, seven ICIs are approved for the treatment of cancer.
Specically, they are pembrolizumab, nivolumab (PD-1 inhibitors),
atezolizumab, durvalumab, avelumab (PD-L1 inhibitors),
ipilimumab, and tremelimumab (CTLA-4 inhibitors). ICIs have
shown efcacy in the treatment of lung cancer, but they also cause
various immune-related adverse events (irAEs). Among them,
myocarditis is rare but has the highest mortality rate among all
irAEs (2). In cancer therapy, the incidence of myocarditis has been
reported to be 1.14% for all ICIs, 0.5% for PD-1 inhibitors, 2.4% for
PD-L1 inhibitors, and 3.3% for CTLA-4 inhibitors (3). In the
KEYNOTE-189 trial, which evaluated the efcacy and safety of
platinum doublet and pembrolizumab combination chemotherapy
in patients with non-squamous non-small cell lung cancer,
myocarditis was reported in only one case (0.2%) (4).
Recently, the coronavirus disease 2019 (COVID-19) pandemic
has had a major impact on healthcare. COVID-19 is an acute
respiratory illness caused by infection with severe acute respiratory
syndrome coronavirus 2 (SARS-CoV-2). It is known that some
COVID-19 patients develop cytokine release syndrome (CRS), in
which inammation-inducing cytokines are increased and the
immune system is activated (5-7). Theoretically, COVID-19
infection could further activate the immune system of cancer
patients being treated with ICI, resulting in severe irAEs. We report
a case of pembrolizumab-induced myocarditis that developed after
COVID-19 infection, in which we were able to document the course
of electrocardiographic changes from onset to death.
Case presentation
A 58-year-old female with a smoking history of at least 35 pack
years had no medical history of dyslipidemia, diabetes mellitus,
hypertension or other medical conditions, and no family history of
coronary artery disease. She received her second COVID-19
vaccination 6 manths ago and no other vaccinations. She was
diagnosed with left lower lobular adenocarcinoma of the lung that
had metastasized to the left hilar and right mediastinal lymph
nodes, invading the pericardium. The tumor was negative for
epidermal growth factor receptor (EGFR), anaplastic lymphoma
kinase (ALK), with a programmed death ligand 1 (PD-L1) tumor
proportion score (TPS) of 25%. She visited her previous physician
complaining of dyspnea after completing her rst cycle of
pembrolizumab, carboplatin, and pemetrexed 9 days earlier.
Subsequently, she was referred to our hospital due to a worsening
pericardial effusion on computed tomography (CT) scan (Figure 1).
On initial examination, she was afebrile with a blood
pressure of 125/78 mm Hg, a heart rate of 124 beats/min, and an
oxygen saturation of 97% on 3 liters per minute of oxygen
administration. Blood tests showed no elevation of creatine kinase
(CK), creatine kinasemyocardial band (CK-MB) or troponin T.
Electrocardiogram showed sinus tachycardia and low-voltage QRS
complexes (Figure 2A), while transthoracic echocardiography (TTE)
revealed pericardial effusion. We diagnosed her with cardiac
tamponade, and she underwent pericardiocentesis, removing
500 ml of bloody uid by drainage tube. Subsequently, her
symptoms and tachycardia improved, and her oxygen saturation
was 96% without oxygen administration. Cytology from the
pericardial uid revealed class V and neoplastic cells consistent
with metastatic lung adenocarcinoma but no genetic mutation was
detected by highly sensitive next-generation sequencing gene panel
assay. The pericardial uid drainage tube was removed 6 days later
since there was no re-accumulation of pericardial uid.
A second cycle of chemotherapy was administered 3 weeks after
the rst cycle since her blood tests revealed declining tumor markers,
and no regrowth of the primary tumor on CT scan. Twenty-two days
after admission, she developed a mild sore throat and tested positive
for SARS-CoV-2 antigen. She was diagnosed with mild COVID-19,
isolated, and treated with sotrovimab. Twenty-seven days after
admission, blood tests showed elevated CK, CK-MB and troponin
T, and a negative T wave appeared on electrocardiogram (Figure 2B).
AB
FIGURE 1
Chest computed tomography on admission. (A) Lung window shows a mass shadow in the lower lobe of the left lung. (B) Mediastinal window shows
pericardial effusion.
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org02
She did not complain of palpitations or chest pain. TTE showed a left
ventricular ejection fraction of about 70%, with no re-accumulation
of pericardial uid, ventricular wall thickening, or ventricular
hypokinesis. However, CK and CK-MB continued to rise on blood
tests, and palpitations appeared 5 days later. She was afebrile with a
blood pressure of 118/82 mm Hg, a heart rate of 111 beats/min, and
an oxygen saturation of 93% on 3 liters per minute of oxygen
administration. Differential diagnoses were considered, with
myocarditis most concerning, followed by acute coronary
syndrome, takotsubo cardiomyopathy, pericardial effusion, and
pulmonary embolism. An electrocardiogram showed monomorphic
VT (Figure 2C). Blood tests revealed CK 5906 U/l, CK-MB 141.7 ng/
ml, troponin T 0.721 ng/ml, and N-terminal prohormone of
brain natriuretic peptide (NT-proBNP) 1368 pg/ml. Pulmonary
embolism was subsequently ruled out with computed tomography
angiography of the chest. Her hemodynamics had been stable, and
she was started on continuous intravenous amiodarone. The next
day, coronary angiography and endomyocardial biopsy (EMB)
were performed. Her coronary arteries were found to be normal.
Cardiac magnetic resonance (CMR) was not performed due to
infection control.
We suspected pembrolizumab-induced myocarditis and
initiated daily methylprednisolone (1 mg/kg/day) immediately
after EMB. The following day, CK and CK-MB decreased on
blood test (Figure 3) and ventricular tachycardia disappeared on
electrocardiogram. Eight days after the start of methylprednisolone
administration, negative T waves remained on electrocardiogram,
but ST-segment elevation was no longer present (Figure 2D);
therefore, she was considered to have passed the acute stage and
amiodarone administration was terminated. Nine days after EMB,
she was diagnosed histologically as having acute lymphocytic
myocarditis. Myocardial tissue collected at the EMB showed an
inltrate of inammatory cells predominantly composed of
lymphocytes (Figure 4A) and granulation brosis of the stroma
(Figure 4B). Immunostaining of the tissue showed an inammatory
cell inltrate predominantly composed of CD8-positive T
lymphocytes (Figure 4C). A viral genome study of the tissue was
not available at our institution. Twelve days after the start of
methylprednisolone administration, R-on-T phenomenon
triggered polymorphic VT (Figure 2E). It immediately
degenerated into ventricular brillation and cardiopulmonary
resuscitation was attempted, but she died.
AB
D
E
C
FIGURE 2
Changes in electrocardiographic waveforms during hospitalization. The admission electrocardiogram showed (A) sinus tachycardia and low-voltage
QRS complexes. (B) Negative T waves appeared on electrocardiogram 27 days after admission, and (C) monomorphic VT appeared 5 days later.
Eight days after the start of methylprednisolone administration, (D) negative T waves remained but ST-segment elevation was no longer present.
Pre-death electrocardiogram showed (E) R-on-T phenomenon triggered polymorphic VT. QT/QTc intervals: (A) 313/443 ms, (B) 395/507 ms,
(C) 433/572 ms, (D) 408/482 ms, (E) 400/408 ms.
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org03
Discussion
This is the rst report of pembrolizumab-induced myocarditis
after COVID-19 infection and is also a valuable case in which
electrocardiographic changes of myocarditis could be recorded in
detail. Although the patient died as a result of arrhythmia, we were
able to conrm that corticosteroids are markedly effective in the
acute phase of pembrolizumab-induced myocarditis.
Myocarditis is an inammatory disease of the myocardium
caused by viral infection, autoimmunity, or drugs (8). The denitive
diagnosis of myocarditis is made by EMB. Myocarditis is classied
as eosinophilic, lymphocytic, giant cell, granulomatous or
pleomorphic based on the type of cells inltrating the
myocardium. In recent years, ICI-induced myocarditis has been
reported with the spread of ICI, and COVID-19-associated
myocarditis with the COVID-19 pandemic.
ICI-induced myocarditis occurs when ICIs maintain T
lymphocyte activity, T lymphocytes inltrate the myocardium,
and the immune response is excessive (9). The median time of
onset was reported to be 34 days after the rst ICI administration
(3). Histological ndings of EMB have been reported to show
myocardial inltration of CD4- positive lymphocytes, CD8-
positive lymphocytes, and CD68-positive macrophages (1012).
Corticosteroids are often used in the initial treatment of ICI-
induced myocarditis, and other immunosuppressive agents are
also considered in corticosteroid-resistant patients (11,12,13).
Guidelines published in 2018 by the American Society of Clinical
Oncology and the National Comprehensive Cancer Center Network
recommend treatment with 1 to 2 mg/kg of prednisone for ICI-
induced myocarditis (15).
On the other hand, COVID-19-associated myocarditis is
thought to result from direct damage to the myocardium by the
virus and myocardial damage by the hosts immune response (16).
The exact incidence of COVID-19-associated myocarditis is
unknown because of diagnostic difculties; some reports indicate
that 5.0% of COVID-19 patients developed new onset myocarditis
(17). Fulminant myocarditis caused by COVID-19 has been
reported to produce ventricular dysfunction and heart failure
within 2 to 3 weeks after infection with SARS-CoV-2 (18,19).
Histological ndings of EMB shows inltration of CD4- and CD8-
positive lymphocytes in myocardial tissue, as well as CD68-positive
macrophages in patients with severe clinical symptoms, such as
fulminant myocarditis (20,21). There are reports that the SARS-
CoV-2 genome was detected in myocardial tissue from some
COVID-19 patients (2225). However, there have been reports of
virus-negative COVID-19-associated myocarditis, and the
authenticity of the SARS-CoV-2 genome remains uncertain (26).
Although the treatment of COVID-19-associated myocarditis has
not yet been established, corticosteroids are not recommended in
viral myocarditis (27).
This patient developed myocarditis 41 days after the rst dose
of pembrolizumab and 11 days after SARS-CoV-2 infection.
Myocardial tissue showed histological ndings of acute lymphocytic
myocarditis. The timing of onset and histological ndings of
myocarditis were consistent with both pembrolizumab-induced
myocarditis and COVID-19-associated myocarditis. We considered
pembrolizumab-induced myocarditis most likely since that
myocarditis improved markedly after corticosteroid administration.
However, COVID-19-associated myocarditis also causes myocardial
damage due to the immune response, so it cannot be completely ruled
out. COVID-19 infection has been reported to increase the risk of
serious irAEs and may have triggered the development of
pembrolizumab-induced myocarditis in this case (28).
An electrocardiogram is a simple test that records the hearts
electrical signals and is often used to detect arrhythmias and
myocardial disorders. In this case, symptoms of myocarditis, such
as palpitations and chest pain, were not present at rst, and it was
difcult to suspect myocarditis from the symptoms alone. However,
we were able to suspect myocarditis at an early stage based on
elevated CK and electrocardiographic changes. In addition, frequent
ECG testing after the onset of myocarditis made it possible to
document ECG changes during the course of treatment for
myocarditis. Poor prognostic factors in electrocardiograms of
acute myocarditis have been reported as pathological Q wave,
wide QRS complex, QRS/T angle 100°, prolonged QT interval,
high-degree atrioventricular block and malignant ventricular
tachyarrhythmia (2932). There are also reports of a high
incidence of heart block, such as complete atrioventricular block
and right bundle branch block, in electrocardiograms of patients
with ICI-induced myocarditis (33). Her ECG showed no heart
block, but a wide QRS complex, QRS/T angle 100°, prolonged
QT interval and malignant ventricular tachyarrhythmia, which
predicted a poor prognosis.
There have been several case reports of successful treatment of
pembrolizumab-induced myocarditis (3436). However, in this
case, she survived the acute stage of myocarditis without the use
of an extracorporeal circulatory device, but the resulting arrhythmia
FIGURE 3
Cardiac biomarkers and electrolytes in blood tests after admission.
CK and CK-MB decreased initially after methylprednisolone
administration, and troponin T and NT-proBNP decreased later.
Electrolytes values at the rst occurrence of VT were K 4.1 mEq/L,
Ca 9.1 mg/dL, Mg 1.8 mg/dL, and at the second occurrence of VT
were K 4.7 mEq/L, Ca 8.9 mg/dL, Mg 2.0 mg/dL. Drugs that induce
VT are not used.
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org04
in the post-acute stage resulted in her death. It is suggested that the
arrhythmia was caused by severe myocardial damage due to acute
myocarditis. The reason for the severe myocardial damage may be
related to COVID-19 infection and pericardial invasion of lung
cancer. This patient had been infected with COVID-19 prior to the
onset of myocarditis, so infection control measures were necessary.
This limited the types of tests that could be performed and delayed
the diagnosis of myocarditis. It also took longer to respond to
emergencies, making it difcult to deal with fatal arrhythmias. In
addition, the possibility of COVID-19-associated myocarditis was
considered at the pre-treatment stage, which caused a delay in the
initiation of corticosteroid administration. There is a report of
pembrolizumab-induced myocarditis in a patient with pericardial
inltration of lung cancer (37). Thus, the administration of ICI to
patients with pericardial inltration of tumor may have resulted in
excessive lymphocyte inltration into the myocardium.
There are several limitations in the present case report. First, it
was difcult to perform an CMR on COVID-19-infected patients at
our institution, and second, we were unable to perform a viral
genome study of myocardial tissue. A viral genome study of
myocardial tissue might have brought us closer to identifying the
cause of myocarditis.
AB
C
FIGURE 4
Histological ndings of endocardial biopsy. Histological ndings of the myocardium showed (A) an inltrate of inammatory cells predominantly
composed of lymphocytes by hematoxylin-eosin (HE) staining and (B) granulation brosis of the stroma by Massons trichrome (MT) staining.
Immunostaining of the tissue showed (C) CD3-positive T cells inltrated more than CD20-positive B cells. CD8-positive T lymphocytes inltrated
more than CD4-positive T lymphocytes. Inltration of CD68-positive macrophages and CD138-positive plasma cells was also observed. (HE ×200,
MT ×200 and CD3/CD4/CD8/CD20/CD68/CD138 immunostaining ×200).
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org05
Conclusion
We report a case of pembrolizumab-induced myocarditis that
developed after COVID-19 infection, in which we were able to
document the course of electrocardiographic changes from onset
to death. In myocarditis, elevated myocardial markers
and electrocardiographic changes may precede clinical
symptoms, so regular myocardial marker measurements and
electrocardiographic testing are important. In addition, ECG
examination is useful even after the start of treatment, since the
prognosis may be inferred from ECG changes. Early diagnosis of
pembrolizumab-induced myocarditis is important because early
administration of corticosteroids may improve the prognosis. The
impact of viral infections such as COVID-19 on patients with ICIs
is unknown, and the appearance of irAEs after infection should
be noted.
Data availability statement
The original contributions presented in the study are
included in the article. Further inquiries can be directed to the
corresponding author.
Author contributions
The manuscript was drafted by KN and KM. KN, KM, YS, JU,
ST, HT, SA, AI, and MM examined and treated the patient. NY
performed the histopathological assessment. YA gave clinical
advice. All authors contributed to the article and approved the
submitted version.
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations,
or those of the publisher, the editors and the reviewers. Any product
that may be evaluated in this article, or claim that may be made by its
manufacturer, is not guaranteed or endorsed by the publisher.
References
1. Das S, Johnson DB. Immune-related adverse events and anti-tumor efcacy of
immune checkpoint inhibitors. J Immunother Cancer (2019) 7(1):306. doi: 10.1186/
s40425-019-0805-8
2. Wang DY, Salem JE, Cohen JV, Chandra S, Menzer C, Ye F, et al. Fatal toxic
effects associated with immune checkpoint inhibitors: A systematic review and meta-
analysis. JAMA Oncol (2018) 4(12):17218. doi: 10.1001/jamaoncol.2018.3923
3. Mahmood SS, Fradley MG, Cohen JV, Nohria A, Reynolds KL, Heinzerling LM,
et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll
Cardiol (2018) 71(16):175564. doi: 10.1016/j.jacc.2018.02.037
4. Rodrı
guez-Abreu D, Powell SF, Hochmair MJ, Gadgeel S, Esteban E, Felip E,
et al. Pemetrexed plus platinum with or without pembrolizumab in patients with
previously untreated metastatic nonsquamous NSCLC: Protocol-specied nal
analysis from KEYNOTE-189. Ann Oncol (2021) 32(7):88195. doi: 10.1016/
j.annonc.2021.04.008
5. Ammirati E, Frigerio M, Adler ED, Basso C, Birnie DH, Brambatti M, et al.
Management of acute myocarditis and chronic inammatory cardiomyopathy: An
expert consensus document. Circ Heart Fail (2020) 13(11):e007405. doi: 10.1161/
CIRCHEARTFAILURE.120.007405
6. Jimenez-Alejandre R, Ruiz-Fernandez I, Martı
n P. Pathophysiology of immune
checkpoint inhibitor-induced myocarditis. Cancers (Basel) (2022) 14(18):4494.
doi: 10.3390/cancers14184494
7. Palaskas N, Lopez-Mattei J, Durand JB, Iliescu C, Deswal A. Immune checkpoint
inhibitor myocarditis: Pathophysiological characteristics, diagnosis, and treatment. J
Am Heart Assoc (2020) 9(2):e013757. doi: 10.1161/JAHA.119.013757
8. Matzen E, Bartels LE, Løgstrup B, Horskær S, Stilling C, Donskov F. Immune
checkpoint inhibitor-induced myocarditis in cancer patients: a case report and review
of reported cases. Cardiooncology (2021) 7(1):27. doi: 10.1186/s40959-021-00114-x
9. Sobol I, Chen CL, Mahmood SS, Borczuk AC. Histopathologic characterization of
myocarditis associated with immune checkpoint inhibitor therapy. Arch Pathol Lab
Med (2020) 144(11):13926. doi: 10.5858/arpa.2019-0447-OA
10. De Luca G, Campochiaro C, Sartorelli S, Peretto G, Dagna L. Therapeutic
strategies for virus-negative myocarditis: A comprehensive review. Eur J Intern Med
(2020) 77:917. doi: 10.1016/j.ejim.2020.04.050
11. Frustaci A, Russo MA, Chimenti C. Randomized study on the efcacy of
immunosuppressive therapy in patients with virus-negative inammatory
cardiomyopathy: The TIMIC study. Eur Heart J (2009) 30(16):19952002.
doi: 10.1093/eurheartj/ehp249
12. Brahmer JR, Lacchetti C, Schneider BJ, Atkins MB, Brassil KJ, Caterino JM, et al.
Management of immune-related adverse events in patients treated with immune
checkpoint inhibitor therapy: American society of clinical oncology clinical practice
guideline. J Clin Oncol (2018) 36(17):171468. doi: 10.1200/JCO.2017.77.6385
13. Agdamag ACC, Edmiston JB, Charpentier V, Chowdhury M, Fraser M, Maharaj
VR, et al. Update on COVID-19 myocarditis. Medicina (Kaunas). (2020) 56(12):678.
doi: 10.3390/medicina56120678
14. Buckley BJR, Harrison SL, Fazio-Eynullayeva E, Underhill P, Lane DA, Lip
GYH. Prevalence and clinical outcomes of myocarditis and pericarditis in 718,365
COVID-19 patients. Eur J Clin Invest. (2021) 51(11):e13679. doi: 10.1111/eci.13679
15.KociolRD,CooperLT,FangJC,MoslehiJJ,PangPS,SabeMA,etal.
Recognition and initial management of fulminant myocarditis: A scientic statement
from the American heart association. Circulation (2020) 141(6):e6992. doi: 10.1161/
CIR.0000000000000745
16. Siripanthong B, Nazarian S, Muser D, Deo R, Santangeli P, Khanji MY, et al.
Recognizing COVID-19-related myocarditis: The possible pathophysiology and
proposed guideline for diagnosis and management. Heart Rhythm (2020) 17
(9):146371. doi: 10.1016/j.hrthm.2020.05.001
17. Jaiswal V, Sarfraz Z, SarfrazA, Mukherjee D, BatraN, Hitawala G, et al. COVID-19
infection and myocarditis: A state-of-the-Art systematic review. J Prim Care Community
Health (2021) 12:21501327211056800. doi: 10.1177/21501327211056800
18. Fox SE, Li G, Akmatbek ov A, Harbert JL, Lameira FS, Brown JQ, et al.
Unexpected features of cardiac pathology in COVID-19 infection. Circulation (2020)
142(11):11235. doi: 10.1161/CIRCULATIONAHA.120.049465
19. Castiello T, Georgiopoulos G, Finocchiaro G, Claudia M, Gianatti A, Delialis D,
et al. COVID-19 and myocarditis: A systematic review and overview of current
challenges. Heart Fail Rev (2022) 27(1):25161. doi: 10.1007/s10741-021-10087-9
20. Escher F, Pietsch H, Aleshcheva G, Bock T, Baumeier C, Elsaesser A, et al.
Detection of viral SARS-CoV-2 genomes and histopathological changes in
endomyocardial biopsies. ESC Heart Fail (2020) 7(5):24407. doi: 10.1002/ehf2.12805
21. Peretto G, Villatore A, Rizzo S, Esposito A, De Luca G, Palmisano A, et al. The
spectrum of COVID-19-Associated myocarditis: A patient-tailored multidisciplinary
approach. J Clin Med (2021) 10(9):1974. doi: 10.3390/jcm10091974
22. Bearse M, Hung YP, Krauson AJ, Bonanno L, Boyraz B, Harris CK, et al. Factors
associated with myocardial SARS-CoV-2 infection, myocarditis, and cardiac
inammation in patients with COVID-19. Mod Pathol (2021) 34(7):134557.
doi: 10.1038/s41379-021-00790-1
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org06
23. Del Nonno F, Frustaci A, Verardo R, Chimenti C, Nicastri E, Antinori A, et al.
Virus-negative myopericarditis in huma n coronavirus infection: Report from an
autopsy series. Circ Heart Fail (2020) 13(11):CIRCHEARTFAILURE120007636.
doi: 10.1161/CIRCHEARTFAILURE.120.007636
24. Chen HS, Wang W, Wu SN,Liu JP. Corticosteroids for viralmyocarditis. Cochrane
Database Syst Rev (2013) 2013 (10):CD004471. doi: 10.1002/14651858.CD004471.pub3
25. Guo M, Liu J, Miao R, Ahmed Z, Yu J, Guan J, et al. A single center retrospective
study of the impact of COVID-19 infection on immune-related adverse events in
cancer patients receiving immune checkpoint inhibitors. J Immunother (2022) 45
(9):38995. doi: 10.1097/CJI.0000000000000440
26. Buttà C, Zappia L, Laterra G, Roberto M. Diagnostic and prognostic role of
electrocardiogram in acute myocarditis: A comprehensive review. Ann Noninvasive
Electrocardiol (2020) 25(3):e12726. doi: 10.1111/anec.12726
27. Ammirati E, Veronese G, Brambatti M, Merlo M, Cipriani M, Potena L, et al.
Fulminantversus acute nonfulminant myocarditis in patients with left ventricular systolic
dysfunction. J Am Coll Cardiol (2019) 74(3):299311. doi: 10.1016/j.jacc.2019.04.063
28. Hung Y, Lin WH, Lin CS, Cheng SM, Tsai TN, Yang SP, et al. The prognostic
role of QTc interval in acute myocarditis. Acta Cardiol Sin (2016) 32(2):22330.
doi: 10.6515/acs20150226a
29. Kindermann I, Barth C, Mahfoud F, Ukena C, Lenski M, Yilmaz A, et al. Update
on myocarditis. J Am Coll Cardiol (2012) 59(9):77992. doi: 10.1016/j.jacc.2011.09.074
30. Power JR, Alexandre J, Choudhary A, Ozbay B, Hayek S, Asnani A, et al.
Electrocardiographic manifestations of immune checkpoint inhibitor myocarditis.
Circulation (2021) 144(18):15213. doi: 10.1161/CIRCULATIONAHA.121.055816
31. Su L, Liu C, Wu W, Cui Y, Wu M, Chen H. Successful therapy for myocarditis
concomitant with complete heart block after pembrolizumab treatment for head and
neck squamous cell carcinoma: A case report with literature review. Front Cardiovasc
Med (2022) 9:898756. doi: 10.3389/fcvm.2022.898756
32. Schiopu SRI, Käsmann L, Schönermarck U, Fischereder M, Grabmaier U,
Manapov F, et al. Pembrolizumab-induced myocarditis in a patient with malignant
mesothelioma: plasma exchange as a successful emerging therapy-case report. Transl
Lung Cancer Res (2021) 10(2):103946. doi: 10.21037/tlcr-20-1095
33. Wang Q, Hu B. Successful therapy for autoimmune myocarditis with
pembrolizumab treatment for nasopharyngeal carcinoma. Ann Transl Med (2019) 7
(11):247. doi: 10.21037/atm.2019.04.73
34. Bersanelli M. Controversies about COVID-19 and anticancer treatment with
immune checkpoint inhibitors. Immunotherapy (2020) 12(5):26973. doi: 10.2217/imt-
2020-0067
35. Yongzhi X. COVID-19-associated cytokine storm syndrome and diagnostic
principles: an old and new issue. Emerg Microbes Infect (2021) 10(1):26676.
doi: 10.1080/22221751.2021.1884503
36. Pasrija R, Naime M. The deregulated immune reaction and cytokines release
storm (CRS) in COVID-19 disease. Int Immunopharmacol (2021) 90:107225.
doi: 10.1016/j.intimp.2020.107225
37. Takashi O, Akira S, Yoshiki S, Tsuyoshi T, Shunzo H. Fulminant myocarditis
after initial pembrolizumab treatment for squamous cell carcinoma of the lung. an
autopsy case report. Japanese J Lung Cancer (2020) 60:33540. doi: 10.2482/
haigan.60.335
Nishiyama et al. 10.3389/mmu.2023.1078838
Frontiers in Immunology frontiersin.org07
... Therefore, it is necessary to carry out medical suspicion when the clinical condition deteriorates after receiving these additional immunosuppressive therapies to correct irAEs [26]. [46][47][48][49][50][51][52]. Of them, 65.4% were male, and the median age was 62.0 years. ...
Article
Full-text available
Cancer patients are susceptible to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Different antitumor treatments have attracted wide attention in the context of coronavirus disease 2019 (COVID-19), especially immune checkpoint inhibitors (ICIs) that have revolutionized oncology changes. It may also have protective and therapeutic roles in viral infections. In this article, we collected 26 cases of SARS-CoV-2 infection during ICIs therapy and 13 related to COVID-19 vaccination from Pubmed, EMBASE, and Wed of Science. Of these 26 cases, 19 (73.1%) presented mild cases and 7 (26.9%) were severe cases. Melanoma (47.4%) was a common cancer type in mild cases and lung cancer (71.4%) in severe cases (P = 0.016). The results showed that their clinical outcomes varied widely. Although there are similarities between the immune checkpoint pathway and COVID-19 immunogenicity, ICIs therapy overactivated T cells, which often leads to immune-related adverse events. In fact, the COVID-19 vaccine has been shown to be safe and effective in patients treated with ICIs. In this review, we report the vital clinical observations of SARS-CoV-2 infection or vaccination in cancer patients treated with ICIs and explore the potential interaction between them.
Article
Full-text available
Immune checkpoint inhibitors (ICIs) have recently emerged as strong therapies for a broad spectrum of cancers being the first-line treatment for many of them, even improving the prognosis of malignancies that were considered untreatable. This therapy is based on the administration of monoclonal antibodies targeting inhibitory T-cell receptors, which boost the immune system and prevent immune evasion. However, non-specific T-cell de-repression can result in a wide variety of immune-related adverse events (irAEs), including gastrointestinal, endocrine, and dermatologic, with a smaller proportion of these having the potential for fatal outcomes such as neurotoxicity, pulmonary toxicity, and cardiotoxicity. In recent years, alarm has been raised about cardiotoxicity as it has the highest mortality rate when myocarditis develops. However, due to the difficulty in diagnosing this cardiac condition and the lack of clinical guidelines for the management of cardiovascular disease in patients on therapy with ICIs, early detection of myocarditis has become a challenge in these patients. In this review we outline the mechanisms of tolerance by which this fatal cardiomyopathy may develop in selected cancer patients treated with ICIs, summarize preclinical models of the disease that will allow the development of more accurate strategies for its detection and treatment, and discuss the challenges in the future to decrease the risks of its development with better decision making in susceptible patients.
Article
Full-text available
Immune checkpoint inhibitors (ICIs) can cause a variety of immune-related adverse events (irAEs). The coronavirus disease 2019 (COVID-19) is associated with increased amounts of pro-inflammatory cytokines, which may affect the outcome of irAEs. Data are limited regarding the impact of COVID-19 on irAEs in ICI-treated cancer patients. Hence, in this study, we retrospectively analyzed ICI-treated adult patients with malignant solid tumors at a single-institution between August 2020 and August 2021. Patients who had the most recent ICI treatment over 1-month before or after the positive COVID-19 test were excluded from the study. For the COVID-19 positive group, only the irAEs that developed after COVID-19 infection were considered as events. A total of 579 patients were included in our study, with 46 (7.9%) in the COVID-19 positive group and 533 (92.1%) in the COVID-19 negative group. The baseline characteristics of patients in the two groups were similar. With a median follow-up of 331 days (range: 21–2,226), we noticed a non-significant higher incidence of all-grade irAEs in the COVID-19 positive group (30.4% vs. 19.9%, p=0.18). The incidence of grade 3 and 4 irAEs was significantly higher in the COVID-19 positive group (10.9% vs. 3.2%, p=0.02). Multivariate analysis confirmed the association between COVID-19 infection and increased risk of severe irAE development (odds ratio: 1.08, 95% confidence interval: 1.02-1.14, p= 0.01). Our study suggested that COVID-19 may pose a risk of severe irAEs in cancer patients receiving ICIs. Close monitoring and possibly delaying ICI administration could be considered when cancer patients are infected with COVID-19.
Article
Full-text available
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy over the past decade. Despite their beneficial effects on treating numerous types of tumors, cardiotoxicity resulting from ICIs is a rare side effect but a concerning one due to its high mortality rate. We herein describe a case of an 80-year-old woman with recurrent head and neck squamous cell cancer (HNSCC), who presented with myocarditis complicated by complete atrioventricular block (CAVB) after second infusion of pembrolizumab. After quickly ruling out myocardial infarction and viral myocarditis, the strong relationship between the onset time and pembrolizumab therapy suggested that ICI-induced myocarditis was the most possible diagnosis. Though CAVB frequently presents with fulminant myocarditis in the setting of ICI-related cardiotoxicity, the patients kept a stable hemodynamic status and had normal myocardial function with just a slightly low global longitudinal strain (GLS) at−16.4%, which implied myocardial injury but was highly related to good prognosis based on the existing literature. Besides, elderly patients are vulnerable to adverse outcomes of steroid therapy, notably opportunistic infections. To balance beneficial effects and adverse effects of immune suppression, she accepted high-dose steroids without pulse methylprednisolone. Excitingly, she had a dramatic clinical and laboratory improvement, and heart block quickly returned to normal sinus rhythm. Another interesting finding was that the patient's tumor remained stable during the half-year follow-up from the termination of immunotherapy. Besides, we here firstly review previously reported cases in terms of their clinical characteristics and prognosis of ICI-induced myocarditis with CAVB, in particular the reversibility of heart block. In conclusion, ICI-induced myocarditis can be life-threatening and it therefore warrants efforts to increase awareness, facilitate early detection, and initiate prompt intervention. Importantly, CAVB secondary to ICIs-induced myocarditis may not always present with fulminant myocarditis and more than 50% of these surviving patients might recover to normal sinus rhythm. For patients with ICI-induced myocarditis with contraindication for cardiac magnetic resonance (CMR), speckle-tracking echocardiography is a reliable and sensitive alternative to CMR for detecting myocardial injury, and GLS may be an important prognostic indicator.
Article
Full-text available
Background COVID-19 was initially considered to be a respiratory illness, but current findings suggest that SARS-CoV-2 is increasingly expressed in cardiac myocytes as well. COVID-19 may lead to cardiovascular injuries, resulting in myocarditis, with inflammation of the heart muscle. Objective This systematic review collates current evidence about demographics, symptomatology, diagnostic, and clinical outcomes of COVID-19 infected patients with myocarditis. Methods In accordance with PRISMA 2020 guidelines, a systematic search was conducted using PubMed, Cochrane Central, Web of Science and Google Scholar until August, 2021. A combination of the following keywords was used: SARS-CoV-2, COVID-19, myocarditis. Cohorts and case reports that comprised of patients with confirmed myocarditis due to COVID-19 infection, aged >18 years were included. The findings were tabulated and subsequently synthesized. Results In total, 54 case reports and 5 cohorts were identified comprising 215 patients. Hypertension (51.7%), diabetes mellitus type 2 (46.4%), cardiac comorbidities (14.6%) were the 3 most reported comorbidities. Majority of the patients presented with cough (61.9%), fever (60.4%), shortness of breath (53.2%), and chest pain (43.9%). Inflammatory markers were raised in 97.8% patients, whereas cardiac markers were elevated in 94.8% of the included patients. On noting radiographic findings, cardiomegaly (32.5%) was the most common finding. Electrocardiography testing obtained ST segment elevation among 44.8% patients and T wave inversion in 7.3% of the sample. Cardiovascular magnetic resonance imaging yielded 83.3% patients with myocardial edema, with late gadolinium enhancement in 63.9% patients. In hospital management consisted of azithromycin (25.5%), methylprednisolone/steroids (8.5%), and other standard care treatments for COVID-19. The most common in-hospital complication included acute respiratory distress syndrome (66.4%) and cardiogenic shock (14%). On last follow up, 64.7% of the patients survived, whereas 31.8% patients did not survive, and 3.5% were in the critical care unit. Conclusion It is essential to demarcate COVID-19 infection and myocarditis presentations due to the heightened risk of death among patients contracting both myocardial inflammation and ARDS. With a multitude of diagnostic and treatment options available for COVID-19 and myocarditis, patients that are under high risk of suspicion for COVID-19 induced myocarditis must be appropriately diagnosed and treated to curb co-infections.
Article
Full-text available
Background Immune checkpoint inhibitor (ICI) induced myocarditis is a rare, severe, and often fatal adverse event. Evidence to guide appropriate immunosuppressive therapy is scarce. We present a case of ICI-induced myocarditis and a review of ICI-induced myocarditis cases to determine the most effective immunosuppressive therapeutic strategy for ICI-induced myocarditis. Methods A systematic search of PubMed was carried out for treatment of ICI-induced myocarditis. Reference lists from identified articles were manually reviewed for additional cases. Results A total of 87 cases with ICI-induced myocarditis were identified. The majority were melanoma ( n = 39), lung cancer ( n = 19), renal cell cancer ( n = 10), and thymoma cancer patients ( n = 4). In 38 (44%) cases, patients received high-dose steroid treatment only. A total of 49 (56%) cases were treated with immunosuppressive agents other than steroid; a total of 13 different immunosuppressive agents were used, including alemtuzumab or abatacept. The median time to onset of symptoms after initiation of ICI was 16 days (range, 1–196 days); cardiotoxic symptoms developed after 2 cycles of ICI (range, 1–13 cycles). A total of 48% of cases were fatal. In cases treated with high-dose steroids only vs. cases treated with other immunosuppressive agents, fatality was 55% and 43% respectively. In 64 out of the 87 cases, tumor control was not described. In patients treated with high-dose steroids only, two patients had stable disease as best tumor response; in patients treated with other immunosuppressive agents, one complete response, one partial response and seven stable disease were noted as best tumor response. Overall, 11 studies were at low risk of bias (12.6%), 38 at moderate risk of bias (43.7%) and 38 at high risk of bias (43.7%). Conclusion Immune checkpoint inhibitor induced myocarditis is a serious and often fatal adverse event. High-dose prednisolone, alemtuzumab or abatacept are all possible treatments options for ICI-induced myocarditis, whereas infliximab increases the risk of death from cardiovascular causes, and should be avoided. Further research is needed.
Article
Full-text available
Background: Myocarditis lacks systematic characterization in COVID-19 patients. Methods: We enrolled consecutive patients with newly diagnosed myocarditis in the context of COVID-19 infection. Diagnostic and treatment strategies were driven by a dedicated multidisciplinary disease unit for myocarditis. Multimodal outcomes were assessed during prospective follow-up. Results: Seven consecutive patients (57% males, age 51 ± 9 y) with acute COVID-19 infection received a de novo diagnosis of myocarditis. Endomyocardial biopsy was of choice in hemodynamically unstable patients (n = 4, mean left ventricular ejection fraction (LVEF) 25 ± 9%), whereas cardiac magnetic resonance constituted the first exam in stable patients (n = 3, mean LVEF 48 ± 10%). Polymerase chain reaction (PCR) analysis revealed an intra-myocardial SARS-CoV-2 genome in one of the six cases undergoing biopsy: in the remaining patients, myocarditis was either due to other viruses (n = 2) or virus-negative (n = 3). Hemodynamic support was needed for four unstable patients (57%), whereas a cardiac device implant was chosen in two of four cases showing ventricular arrhythmias. Medical treatment included immunosuppression (43%) and biological therapy (29%). By the 6-month median follow-up, no patient died or experienced malignant arrhythmias. However, two cases (29%) were screened for heart transplantation. Conclusions: Myocarditis associated with acute COVID-19 infection is a spectrum of clinical manifestations and underlying etiologies. A multidisciplinary approach is the cornerstone for tailored management.
Article
Full-text available
Myocardial inflammation in COVID-19 has been documented. Its pathogenesis is not fully elucidated, but the two main theories foresee a direct role of ACE2 receptor and a hyperimmune response, which may also lead to isolated presentation of COVID-19-mediated myocarditis. The frequency and prognostic impact of COVID-19-mediated myocarditis is unknown. This review aims to summarise current evidence on this topic. We performed a systematic review of MEDLINE and Cochrane Library (1/12/19–30/09/20). We also searched clinicaltrials.gov for unpublished studies testing therapies with potential implication for COVID-19-mediated cardiovascular complication. Eligible studies had laboratory confirmed COVID-19 and a clinical and/or histological diagnosis of myocarditis by ESC or WHO/ISFC criteria. Reports of 38 cases were included (26 male patients, 24 aged < 50 years). The first histologically proven case was a virus-negative lymphocytic myocarditis; however, biopsy evidence of myocarditis secondary to SARS-CoV-2 cardiotropism has been recently demonstrated. Histological data was found in 12 cases (8 EMB and 4 autopsies) and CMR was the main imaging modality to confirm a diagnosis of myocarditis (25 patients). There was a substantial variability in biventricular systolic function during the acute episode and in therapeutic regimen used. Five patients died in hospital. Cause-effect relationship between SARS-CoV-2 infection and myocarditis is difficult to demonstrate. However, current evidence demonstrates myocardial inflammation with or without direct cardiomyocyte damage, suggesting different pathophysiology mechanisms responsible of COVID-mediated myocarditis. Established clinical approaches should be pursued until future evidence support different actions. Large multicentre registries are advisable to elucidate further.
Article
Immune checkpoint inhibitors (ICIs) have transformed oncology care by unleashing T-cells to achieve anti-tumor effects but can cause inflammatory adverse events including myocarditis. This study shows that ICI-myocarditis is highly arrhythmogenic, presenting with new conduction blocks, decreased voltage, and repolarization abnormalities that frequently degenerate to malignant arrhythmias. Further studies are needed to evaluate how these ECG changes can facilitate screening, prognostication, and monitoring strategies in ICI-myocarditis.
Article
Background COVID-19 has a wide spectrum of cardiovascular sequelae including myocarditis and pericarditis; however, the prevalence and clinical impact is unclear. We investigated the prevalence of new-onset myocarditis/pericarditis and associated adverse cardiovascular events in patients with COVID-19. Methods and Results A retrospective cohort study was conducted using electronic medical records from a global federated health research network. Patients were included based on a diagnosis of COVID-19 and new-onset myocarditis or pericarditis. Patients with COVID-19 and myocarditis/pericarditis were 1:1 propensity score matched for age, sex, race and co-morbidities to patients with COVID-19 but without myocarditis/pericarditis. The outcomes of interest were 6-month all-cause mortality, hospitalisation, cardiac arrest, incident heart failure, incident atrial fibrillation, and acute myocardial infarction, comparing patients with and without myocarditis/pericarditis. Of 718,365 patients with COVID-19, 35,820 (5.0%) developed new onset myocarditis and 10,706 (1.5%) developed new onset pericarditis. Six-month all-cause mortality was 3.9% (n=702) in patients with myocarditis and 2.9% (n=523) in matched controls (P<0.0001), odds ratio 1.36 (95% confidence interval (CI): 1.21-1.53). Six-month all-cause mortality was 15.5% (n=816) for pericarditis and 6.7% (n=356) in matched controls (P<0.0001), odds ratio 2.55 (95% CI: 2.24-2.91). Receiving critical care was associated with significantly higher odds of mortality for patients with myocarditis and pericarditis. Patients with pericarditis seemed to associate with more new-onset cardiovascular sequelae than those with myocarditis. This finding was consistent when looking at pre-COVID-19 data with pneumonia patients. Conclusions Patients with COVID-19 who present with myocarditis/pericarditis associate with increased odds of major adverse events and new-onset cardiovascular sequelae.
Article
Background: In the phase 3 KEYNOTE-189 study (NCT02578680), pembrolizumab plus pemetrexed and platinum-based chemotherapy (pemetrexed-platinum) significantly improved overall survival (OS) and progression-free survival (PFS) in patients with previously untreated metastatic nonsquamous NSCLC versus placebo plus pemetrexed-platinum. We report updated efficacy outcomes from the protocol-specified final analysis, including outcomes in patients who crossed over to pembrolizumab from pemetrexed-platinum and in patients who completed 35 cycles (approximately 2 years) of pembrolizumab. Patients and methods: Eligible patients were randomized 2:1 to pembrolizumab 200 mg (n=410) or placebo (n=206) every 3 weeks (for up to 35 cycles, approximately 2 years) plus 4 cycles of pemetrexed (500 mg/m2) and investigators' choice of cisplatin (75 mg/m2) or carboplatin (AUC 5 mg/ml/min) every 3 weeks, followed by pemetrexed until progression. Patients assigned to placebo plus pemetrexed-platinum could crossover to pembrolizumab upon progression if eligibility criteria were met. The primary endpoints were OS and PFS. Results: After median follow-up of 31.0 months, pembrolizumab plus pemetrexed-platinum continued to improve OS (hazard ratio [HR], 0.56; 95% CI, 0.46‒0.69), and PFS (HR, 0.49; 95% CI, 0.41‒0.59) over placebo plus pemetrexed-platinum regardless of PD-L1 expression. ORR (48.3% versus 19.9%) and time to second/subsequent tumor progression on next-line treatment (PFS2; HR, 0.50; 95% CI, 0.41‒0.61) were improved in patients who received pembrolizumab plus pemetrexed-platinum. 84 patients (40.8%) from the placebo plus pemetrexed-platinum group crossed over to pembrolizumab on-study. Grade 3‒5 adverse events occurred in 72.1% of patients receiving pembrolizumab plus pemetrexed-platinum and 66.8% of patients receiving placebo plus pemetrexed-platinum. 56 patients completed 35 cycles (approximately 2 years) of pembrolizumab; ORR was 85.7% and 53 (94.6%) were alive at data cutoff. Conclusion: Pembrolizumab plus pemetrexed-platinum continued to show improved efficacy outcomes compared with placebo plus pemetrexed-platinum, with manageable toxicity. These findings support first-line pembrolizumab plus pemetrexed-platinum in patients with previously untreated metastatic nonsquamous NSCLC.