Introduction: The development of immune-related adverse events (irAEs) has been associated with improved survival from various solid tumors. Given that immunotherapy has not been widely used in ovarian cancer and has only been applied to patients with high tumor mutational burden or microsatellite instability, studies exploring the effects of irAEs on ovarian cancer have been limited. Case Presentation: A 47-year-old woman was diagnosed with International Federation of Gynecology and Obstetrics stage III ovarian cancer in 2013. Between 2013 and 2021, she underwent palliative chemotherapy comprising paclitaxel liposomes, cisplatin/nedaplatin, S-1/raltitrexed, irinotecan, doxorubicin, vinorebine, toripalimab, apatinib, gemcitabine, oxaliplatin, and capecitabine, as well as two debulking surgeries. From November 2021, she received six cycles of tislelizumab (a PD-1 inhibitor), paclitaxel (albumin-bound), and carboplatin, to which a partial response was observed according to the Response Evaluation Criteria in Solid Tumors. From May 2022, the patient was switched to maintenance therapy with tislelizumab plus olaparib. However, all antitumor treatments were discontinued from April 2023 due to multiple irAEs, including hypothyroidism, adrenal insufficiency, and pneumonitis, with the tumor remaining stable until November 2023. Progression-free survival (PFS) was approximately 24 months with tislelizumab-containing therapy but was 18 months with tislelizumab/olaparib maintenance therapy. Conclusions: We report a case involving a patient with highly pretreated recurrent ovarian cancer who exhibited prolonged PFS after developing three irAEs. The distinctly prolonged PFS observed, along with the reviewed literature, suggests that irAEs may be correlated with improved survival in ovarian cancer.

Ovarian cancer, one of the most common malignancies among females, has remained the leading cause of gynecological cancer-related deaths. Given that approximately half of patients are diagnosed with advanced disease, ovarian cancer has a 5-year survival rate of approximately 49%. Historically, debulking surgery and platinum-taxane-based chemotherapy have been the standard therapies for advanced or recurrent epithelial ovarian cancer. Over the last decade, however, poly (ADP-ribose) polymerase inhibitors (PARPi) and anti-angiogenic drugs have been proven effective against recurrent ovarian cancer, thereby altering the treatment landscape. Immune checkpoint inhibitors (ICIs) have shown limited efficacy for recurrent ovarian cancer and have only been approved for mismatch repair-deficient/microsatellite instability-high or high tumor mutational burden (≥10 mutations/Mb) ovarian cancer. Although current evidence fails to validate ICI-chemotherapy combinations, emerging evidence demonstrates synergistic antitumor efficacy when combining ICIs with either PARP inhibitors or antiangiogenic [1]. This therapeutic potential is supported by the Phase III DUO-O trial (NCT03737643), which demonstrated significantly improved progression-free survival (PFS) among newly diagnosed non-BRCAm advanced ovarian cancer patients receiving paclitaxel/carboplatin/bevacizumab/durvalumab induction followed by bevacizumab/durvalumab/olaparib maintenance than among those receiving paclitaxel/carboplatin/bevacizumab with bevacizumab maintenance alone [2]. Additionally, preliminary evidence indicates that combining dual immunotherapy with chemotherapy may confer survival benefits [3]. Currently, multiple phase III randomized trials are investigating various ICI-based combination regimens for ovarian cancer management, though definitive results remain pending.

Immune-related adverse events (irAEs) have been associated with longer overall survival (OS) and PFS, as well as better objective response rates across various tumor types. However, these studies have mainly focused on non-small cell lung cancers (NSCLCs), melanomas, esophageal carcinomas, and hepatocellular carcinomas (HCCs), for which ICIs are widely used. Only a few studies have explored the effects of irAEs on ovarian cancer. In this report, we present a case involving a patient with recurrent ovarian cancer who showed notably prolonged PFS after developing multisystem irAEs.

In July 2013, a 47-year-old woman presented to our institution with recurrent lower abdominal pain. She underwent exploratory laparotomy, which revealed her unsuitability for debulking surgery. The patient was then diagnosed with FIGO stage III poorly differentiated ovarian adenocarcinoma. Thereafter, palliative chemotherapy with paclitaxel liposomes, cisplatin/nedaplatin, S-1/raltitrexed, irinotecan, doxorubicin, and vinorelbine was administered successfully from 2013 to 2019 (Fig. 1). The first debulking surgery was performed in March 2015, followed by a second one in February 2019. Histopathological examination revealed high-grade serous papillary carcinoma of the ovary.

Fig. 1.

Chronology of the patient’s medical history.

Fig. 1.

Chronology of the patient’s medical history.

Close modal

Liver metastases were first detected in May 2020. Core biopsy of the lesion confirmed the presence of high-grade serous papillary carcinoma of the ovary. Genetic testing revealed low microsatellite instability, a programmed cell death ligand 1 combined positive score of <1, a tumor mutational burden of 6.70 mutations/Mb, and no BRCA mutations. Two cycles of toripalimab (a PD-1 inhibitor) combined with apatinib, gemcitabine, and oxaliplatin were then administered, with subsequent computed tomography (CT) confirming a partial response based on the Response Evaluation Criteria in Solid Tumors (RECIST). After liver metastasis ablation, the patient received three cycles of toripalimab with apatinib and 1 year of maintenance therapy with toripalimab and capecitabine until tumor progression (enlargement of the left iliac paravascular lymph node) in November 2021. The patient developed hypothyroidism (grade 2) after 2 days of toripalimab use, for which thyroid hormone supplementation with levothyroxine was initiated.

From November 2021, she received six cycles of tislelizumab (a PD-1 inhibitor), paclitaxel (albumin-bound), and carboplatin, which promoted a partial response according to the RECIST. Maintenance therapy with tislelizumab combined with olaparib was initiated in May 2022, which stabilized the tumor. However, in February 2023, the patient complained of loss of appetite, general fatigue, and poor sleep and was subsequently diagnosed with adrenal insufficiency based on the examinations showing normal blood adrenocorticotropic hormone levels (2.59 pmol/L, reference range: 1.60–13.90 pmol/L), decreased blood cortisol levels (1.71 μg/dL, reference range: 3.7–19.40 μg/dL), and decreased urine cortisol (1 µg/24 h, reference range: 4.3–176 μg/24 h). Hydrocortisone administration gradually ameliorated the patient’s clinical symptoms. In April 2023, the patient presented with chest tightness, a cough, and a small amount of white phlegm, with CT revealing interstitial lung disease. She was then diagnosed with pneumonitis (grade 4), which prompted the discontinuation of all antitumor treatments. Thereafter, the patient was treated with methylprednisolone, which improved her condition. Since then, the patient had been followed-up through CA125 tests and chest, abdominal, and pelvic CT every 3–4 months. Serum CA125 levels remained low 4 months after discontinuing antitumor treatment (Fig. 2), with CT confirming that the tumor remained stable until November 2023 (Fig. 3). The patient expressed that, since the onset of her cancer, she was the happiest she had ever been over the 7-month period in which her antitumor treatment was stopped.

Fig. 2.

Serum CA125 levels were stable during tislelizumab-containing therapy and remained low for 4 months after stopping antitumor treatment.

Fig. 2.

Serum CA125 levels were stable during tislelizumab-containing therapy and remained low for 4 months after stopping antitumor treatment.

Close modal
Fig. 3.

a Computed tomography (CT) in November 2021 conducted before tislelizumab combination therapy revealing liver metastasis in segment 2/3 (arrowheads). b CT in August 2023 conducted after stopping antitumor treatment for 4 months revealing shrinkage of the liver metastasis in segment 2/3. c CT in November 2023 revealing new liver metastases.

Fig. 3.

a Computed tomography (CT) in November 2021 conducted before tislelizumab combination therapy revealing liver metastasis in segment 2/3 (arrowheads). b CT in August 2023 conducted after stopping antitumor treatment for 4 months revealing shrinkage of the liver metastasis in segment 2/3. c CT in November 2023 revealing new liver metastases.

Close modal

This report presents a case involving a patient with highly pretreated, non-gBRCAm recurrent ovarian cancer who exhibited prolonged PFS after developing three irAEs, namely, hypothyroidism, adrenal insufficiency, and pneumonitis. The patient exhibited a PFS of approximately 24 months with tislelizumab-containing therapy and 18 months with tislelizumab plus olaparib maintenance, which were notably superior outcomes than those reported in comparable studies. For context, despite employing a similar design involving six cycles of platinum-based chemotherapy plus atezolizumab followed by maintenance niraparib plus atezolizumab for recurrent ovarian cancer patients, the phase III ANITA trial (NCT03598270) reported a median PFS of 11.2 months in the intention-to-treat population and 11.1 months in the non-gBRCAm cohort, with maintenance PFS lasting only 6.7 months [4]. Similarly, the MEDIOLA phase II trial (NCT02734004), which administered olaparib plus durvalumab in two cohorts, yielded a median PFS of 15.0 months (gBRCAm cohort) and 5.5 months (non-gBRCAm cohort) [5]. Although emerging evidence suggests that irAEs may confer survival benefits across multiple cancer types, neither the ANITA nor MEDIOLA trial included preplanned irAE subgroup analyses, leaving the relationship between irAEs and clinical benefit unresolved. However, exploratory analyses in these studies indicated that cohorts with higher irAE rates had longer PFS (Table 1). To date, the relationship between irAE development and survival in ovarian cancer remains unclear. Here, we review available literature on the association between survival outcomes and irAE development, irAE types, irAE numbers, and irAE severity.

Table 1.

Incidence of irAEs and PFS in ovarian cancer patients treated with PARP inhibitors and immunotherapy

StudyGroupAE rateIrAE rateMedian PFS, months
ANITA Atezolizumab + CT → atezolizumab + niraparib (n = 208) 97% 23% 11.2 
 Placebo + CT → placebo + niraparib (n = 209) 97% 9% 10.1 
MEDIOLA Olaparib + durvalumab, gBRCAm cohorts (n = 51) 100% 29.4% 15.0 
Olaparib + durvalumab, non-gBRCAm cohorts (n = 32) 100% 15.6% 5.5 
StudyGroupAE rateIrAE rateMedian PFS, months
ANITA Atezolizumab + CT → atezolizumab + niraparib (n = 208) 97% 23% 11.2 
 Placebo + CT → placebo + niraparib (n = 209) 97% 9% 10.1 
MEDIOLA Olaparib + durvalumab, gBRCAm cohorts (n = 51) 100% 29.4% 15.0 
Olaparib + durvalumab, non-gBRCAm cohorts (n = 32) 100% 15.6% 5.5 

AE, adverse event; irAE, immune-related adverse events; PFS, progression-free survival; CT, chemotherapy; gBRCAm, germline BRCA mutation.

Association between irAE Development and Survival

Patients with locally advanced or metastatic NSCLC with irAEs have demonstrated better treatment outcomes than those without irAEs. In fact, a retrospective observational cohort study of 803 patients suggested that irAEs were correlated with improved OS, with a median OS of 23.7 and 9.8 months in those with and without irAEs, respectively (p < 0.001) [6]. Moreover, a study on patients with urothelial cancer found that those who developed irAEs had a significantly better OS than those who did not (hazard ratio 0.53, 95% CI: 0.43–0.66) and that the use of systemic corticosteroids had no effect on this benefit [7]. Similarly, among patients with esophageal carcinoma, those with irAEs had longer OS and PFS than those without irAEs [8]. Another study showed that the development of irAEs following atezolizumab combined with bevacizumab, which has become the first-line therapy for advanced HCC, was associated with improved OS and PFS in HCC patients [9]. Likewise, melanoma patients who experienced irAEs had significantly better OS than those who did not [10]. Taken together, these findings suggest that irAEs were often associated with enhanced clinical outcomes across a range of cancers, although the underlying mechanisms have remained an area of active research.

Association between irAE Types and Survival

Endocrine and cutaneous irAEs have been widely reported to be associated with improved survival outcomes. One study found that both irAEs were associated with longer PFS and OS in patients with esophageal squamous cell carcinoma [8]. Another study found that HCC patients with hypothyroidism showed longer OS and time to progression than did those without irAEs [11]. Two retrospective studies on metastatic NSCLC demonstrated that patients who developed endocrine irAEs had significantly better OS and PFS than those who did not and that cutaneous irAEs were notably correlated with longer PFS but were not significantly associated with OS [12]. A systematic review of studies involving 3,815 patients with metastatic lung cancer or head and neck cancer who received ICIs identified a significant positive correlation between OS and endocrine irAEs [13]. Overall, these findings suggest that while endocrine and cutaneous irAEs may serve as markers for better clinical outcomes in certain types of cancers, their associations with survival can vary depending on the type of irAE and cancer.

Association between irAE Severity and Survival

Several studies have shown that mild irAEs can serve as predictive markers of improved immunotherapy efficacy across various tumor types. Among patients with HCC, grade 1/2 irAEs were strongly correlated with better OS than were grade 3/4 irAEs [9]. A pooled analysis of the IMpower130, IMpower132, and IMpower150 trials only patients with metastatic NSCLC who experienced low-grade irAEs demonstrated an increase in OS [14]. However, a recent retrospective study of 803 patients with metastatic NSCLC reported no significant difference in OS between outpatients and those being treated for irAEs in the hospital [6]. Another study found no significant difference in OS or PFS between patients with esophageal squamous cell carcinoma who experienced grade 1/2 and grade ≥3 irAEs [8]. Collectively, these results suggest that while low-grade irAEs may be associated with improved survival outcomes in certain types of cancers, the relationship between irAE grade and survival benefits can vary depending on cancer type and treatment context.

Association between irAE Number and Survival

Two studies have consistently demonstrated that irAE number was positively correlated with increased OS and PFS in patients with advanced NSCLC [12, 15]. Similarly, 1 study reported that patients with multisystem irAEs, one irAE, and no irAEs had a median PFS of 10.9, 5.1, and 2.8 months (p < 0.001) and an OS of 21.8, 12.3, and 8.7 months, respectively [15]. Among patients with advanced melanoma, those who experienced ≥3 irAEs had longer OS than did those who experienced no or only one irAE [10]. However, 1 study on patients with esophageal cancer found no significant difference in OS or PFS between those with multisystem irAEs and those with only one irAE [8]. Altogether, these findings suggest that although a higher number of irAEs was generally linked to improved survival in certain types of cancers, the association may vary depending on cancer type.

This report presents a case involving a patient with highly pretreated non-gBRCAm recurrent ovarian cancer who demonstrated prolonged PFS after developing three irAEs. The markedly extended PFS observed in this case, along with the findings of our literature review, suggests a potential correlation between irAE occurrence and improved survival outcomes in patients with ovarian cancer. We recommend that future clinical trials incorporate irAE-based stratification to validate this relationship in larger populations. Additional mechanistic studies are warranted to elucidate the underlying biological mechanisms for this relationship.

The CARE Checklist has been completed by the authors for this case report, attached as online supplementary material (for all online suppl. material, see https://doi.org/10.1159/000546423).

Written informed consent was obtained from the patient for publication of this case report and any accompanying images. Ethical approval to report this case was obtained from the Ethics Committee of The Seventh Affiliated Hospital of Sun Yat-sen University (KY-2025-020-01).

The authors have no conflicts of interest to declare.

This work was supported by the National Natural Science Foundation of China (32200583) and Shenzhen Science and Technology Program (Grant No. JCYJ20230807110312026).

Writing: M.L.; review and editing: S.F. and H.D.; investigation: M.L., S.F., and H.D.; data curation: T.L. and C.G.; and supervision: B.W. All authors have read and agreed to the published version of the manuscript.

Additional Information

Minling Liu, Shuo Fang, and Huiru Dai contributed equally to this work and share first authorship.

All data supporting the findings in this case report are contained within the text. Additional data are available upon request from the corresponding author.

1.
Bogani
G
,
Moore
KN
,
Ray-Coquard
I
,
Lorusso
D
,
Matulonis
UA
,
Ledermann
JA
, et al
.
Incorporating immune checkpoint inhibitors in epithelial ovarian cancer
.
Gynecol Oncol
.
2025
;
193
:
30
40
.
2.
Harter
P
,
Trillsch
F
,
Okamoto
A
,
Reuss
A
,
Kim
J-W
,
Rubio-Pérez
MJ
, et al
.
Durvalumab with paclitaxel/carboplatin (PC) and bevacizumab (bev), followed by maintenance durvalumab, bev, and olaparib in patients (pts) with newly diagnosed advanced ovarian cancer (AOC) without a tumor BRCA1/2 mutation (non-tBRCAm): results from the randomized, placebo (pbo)-controlled phase III DUO-O trial
.
J Clin Oncol
.
2023
;
41
(
17_Suppl l
):
LBA5506
.
3.
Kim
SI
,
Joung
JG
,
Kim
YN
,
Park
J
,
Park
E
,
Kim
JW
, et al
.
Durvalumab with or without tremelimumab plus chemotherapy in HRR non-mutated, platinum-resistant ovarian cancer (KGOG 3045): a phase II umbrella trial
.
Gynecol Oncol
.
2024
;
182
:
7
14
.
4.
González-Martín
A
,
Rubio
MJ
,
Heitz
F
,
Depont Christensen
R
,
Colombo
N
,
Van Gorp
T
, et al
.
Atezolizumab combined with platinum and maintenance niraparib for recurrent ovarian cancer with a platinum-free interval >6 months: ENGOT-OV41/GEICO 69-O/ANITA phase III trial
.
J Clin Oncol
.
2024
;
42
(
36
):
4294
304
.
5.
Drew
Y
,
Kim
JW
,
Penson
RT
,
O’Malley
DM
,
Parkinson
C
,
Roxburgh
P
, et al
.
Olaparib plus durvalumab, with or without bevacizumab, as treatment in PARP inhibitor-naïve platinum-sensitive relapsed ovarian cancer: a phase II multi-cohort study
.
Clin Cancer Res
.
2024
;
30
(
1
):
50
62
.
6.
Cook
S
,
Samuel
V
,
Meyers
DE
,
Stukalin
I
,
Litt
I
,
Sangha
R
, et al
.
Immune-related adverse events and survival among patients with metastatic NSCLC treated with immune checkpoint inhibitors
.
JAMA Netw Open
.
2024
;
7
(
1
):
e2352302
.
7.
Maher
VE
,
Fernandes
LL
,
Weinstock
C
,
Tang
S
,
Agarwal
S
,
Brave
M
, et al
.
Analysis of the association between adverse events and outcome in patients receiving a programmed death protein 1 or programmed death ligand 1 antibody
.
J Clin Oncol
.
2019
;
37
(
30
):
2730
7
.
8.
Cao
R
,
Wu
C
,
Lv
Y
,
Xu
S
,
Li
C
,
Yin
F
, et al
.
Correlation between immune-related adverse events and treatment efficacy of anti-PD1 immunotherapy in patients with esophageal squamous cell carcinoma
.
Oncol Lett
.
2023
;
25
(
2
):
55
.
9.
Fukushima
T
,
Morimoto
M
,
Kobayashi
S
,
Ueno
M
,
Uojima
H
,
Hidaka
H
, et al
.
Association between immune-related adverse events and survival in patients with hepatocellular carcinoma treated with atezolizumab plus bevacizumab
.
Oncologist
.
2023
;
28
(
7
):
e526
33
.
10.
Freeman-Keller
M
,
Kim
Y
,
Cronin
H
,
Richards
A
,
Gibney
G
,
Weber
JS
.
Nivolumab in resected and unresectable metastatic melanoma: characteristics of immune-related adverse events and association with outcomes
.
Clin Cancer Res
.
2016
;
22
(
4
):
886
94
.
11.
Zhou
JM
,
Xiong
HF
,
Chen
XP
,
Zhang
ZW
,
Zhu
LP
,
Wu
B
.
Correlation between immune-related adverse events and long-term outcomes in pembrolizumab-treated patients with unresectable hepatocellular carcinoma: a retrospective study
.
World J Gastrointest Oncol
.
2023
;
15
(
4
):
689
99
.
12.
Ricciuti
B
,
Genova
C
,
De Giglio
A
,
Bassanelli
M
,
Dal Bello
MG
,
Metro
G
, et al
.
Impact of immune-related adverse events on survival in patients with advanced non-small cell lung cancer treated with nivolumab: long-term outcomes from a multi-institutional analysis
.
J Cancer Res Clin Oncol
.
2019
;
145
(
2
):
479
85
.
13.
Gomes-Lima
CJ
,
Kwagyan
J
,
King
F
,
Fernandez
SJ
,
Burman
KD
,
Veytsman
I
.
A comprehensive meta-analysis of endocrine immune-related adverse events of immune checkpoint inhibitors and outcomes in head and neck cancer and lung cancer
.
J Clin Oncol
.
2019
;
37
(
15_Suppl l
):
e14096
.
14.
Socinski
MA
,
Jotte
RM
,
Cappuzzo
F
,
Nishio
M
,
Mok
TSK
,
Reck
M
, et al
.
Association of immune-related adverse events with efficacy of atezolizumab in patients with non-small cell lung cancer: pooled analyses of the phase 3 IMpower130, IMpower132, and IMpower150 randomized clinical trials
.
JAMA Oncol
.
2023
;
9
(
4
):
527
35
.
15.
Shankar
B
,
Zhang
J
,
Naqash
AR
,
Forde
PM
,
Feliciano
JL
,
Marrone
KA
, et al
.
Multisystem immune-related adverse events associated with immune checkpoint inhibitors for treatment of non-small cell lung cancer
.
JAMA Oncol
.
2020
;
6
(
12
):
1952
6
.