Logo American Journal of Case Reports

Call: 1.631.629.4328
Mon-Fri 10 am - 2 pm EST

Contact Us

Logo American Journal of Case Reports Logo American Journal of Case Reports Logo American Journal of Case Reports

10 April 2025: Articles  Taiwan

Navigating Fertility Preservation in Epithelioid Peritoneal Mesothelioma: A Case Study

Challenging differential diagnosis, Unusual setting of medical care, Rare disease

Daniel Wang Qiu ORCID logo1BCEF, Yi-Jin Lin Lee ORCID logo1BCEF, Garima Tripathi123BCEF, Dito Anurogo ORCID logo4BCEF, Cheng-Fu Ni5CD, Wei-Chin Chang ORCID logo6BCD, Hao-Sheng Fu3BCD, J. Timothy Qiu137ABCDEFG*

DOI: 10.12659/AJCR.946585

Am J Case Rep 2025; 26:e946585

0 Comments

Abstract

0:00

BACKGROUND: Malignant peritoneal mesothelioma (MPM) is a rare and aggressive neoplasm, with low-grade epithelioid subtypes presenting diagnostic and therapeutic challenges. With nonspecific symptoms and diagnostic challenges, definitive diagnosis relies on histopathological and immunohistochemical analysis. This study reviews the current research on the epidemiology, diagnosis, and treatment of MPM, focusing on preserving fertility and achieving successful childbirth following surgery, chemotherapy, and immunotherapy, despite immune-related adverse events.

CASE REPORT: We report the case of a 44-year-old nulliparous woman, initially diagnosed with a uterine leiomyoma, who was incidentally found to have MPM during surgery. Diagnostic laparoscopy and subsequent imaging revealed extensive peritoneal involvement, confirmed by histopathology and immunohistochemistry. The patient’s desire to preserve fertility complicated the treatment approach, leading to chemotherapy followed by immunotherapy, with complete disease control. Without evidence of disease recurrence or metastasis, the patient proceeded with in vitro fertilization (IVF) and embryo transfer. During an emergency cesarean section at 30 weeks 2 days of gestation, performed due to preterm premature rupture of membranes, intraabdominal residual tumors were discovered and excised. No evidence of malignancy was found elsewhere.

CONCLUSIONS: MPM is a rare disease, often with a delayed diagnosed due to nonspecific symptoms. Treatments like cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) improve survival for operable cases, while systemic therapies and immune-checkpoint inhibitors (ICIs) are options for inoperable cases; however, further research on early-detection biomarkers and the impact of ICIs on fertility is needed to enhance clinical practice. This case highlights the intricate balance between oncologic control and fertility preservation in MPM, illustrating the need for individualized treatment strategies.

Keywords: Chemotherapy, Adjuvant, fertility preservation, hyperthermic intraperitoneal chemotherapy, Immunohistochemistry, Immunotherapy, Mesothelioma, Malignant

Introduction

Malignant peritoneal mesothelioma (MPM) is a rare but aggressive neoplasm arising from the serosal membranes lining the peritoneal cavity. Although it accounts for only a small percentage of all mesothelioma cases, MPM’s clinical and pathological challenges make it a formidable disease with significant morbidity and mortality [1,2]. The incidence of MPM has shown a relatively stable trend, with a higher prevalence noted in females, contrasting with the male predominance observed in pleural mesothelioma [3,4]. The etiological role of asbestos exposure, although well-established in pleural mesothelioma, appears to be less pronounced in peritoneal cases, suggesting a multifactorial pathogenesis, including genetic predispositions such as BAP1 mutations [1,5].

The clinical presentation of MPM is often insidious and non-specific, leading to frequent delays in diagnosis. Symptoms such as abdominal distension, pain, and ascites are common but are not unique to this malignancy, often resulting in late-stage detection. Diagnostic modalities such as computed tomography (CT) and magnetic resonance imaging (MRI) are useful for identifying suspicious lesions, yet they lack specificity, necessitating histopathological confirmation through biopsy [2]. The advent of immunohistochemistry (IHC) has greatly enhanced the diagnostic accuracy of MPM, particularly in distinguishing it from other malignancies that present with similar clinical features [6,7]. However, despite advances in diagnostic techniques and therapeutic approaches, including CRS and HIPEC, the prognosis for patients with MPM remains poor, underscoring the need for ongoing research and development of more effective treatment strategies [8,9]. Therefore, an increasing number of studies have focused on the immunotherapies used in mesothelioma, with growing utilization and attention to the treatment outcomes of immune-checkpoint inhibitors (Table 1).

Case Report

INCIDENTAL DISCOVERY OF MALIGNANT PERITONEAL MESOTHELIOMA:

A 44-year-old asymptomatic nulliparous woman presented with a palpable pelvic mass. The physical examination revealed an enlarged uterus of about 10- to 12-week size, which was mobile, firm in consistency, with no uterine or forniceal tenderness, and mild swelling in the right pubic area. The primary diagnosis is an anterior wall uterine leiomyoma, measuring 112×94×108 mm, accompanied by ascites measuring 65×42 mm and a 20×14 mm mass, as identified via gynecological sonography on January 30, 2010. During a laparotomic myomectomy performed on February 17, 2020, an incidental discovery of a pelvic tumor was made.

This tumor appeared as a cauliflower-like lesion disseminated across the right pelvic wall, cul-de-sac, anterior uterine surface, bladder, and colon (Figure 1). A biopsy of the sigmoid colon serosa yielded a 0.8 cm sample, which pathologically demonstrated proliferative mesothelial cells without significant atypia or necrosis. A post-myomectomy CT scan (Figure 2) revealed seeding in the lower pelvis, characterized by multiple small, heterogeneous, enhancing nodules on the peritoneum and a prominent soft-tissue mass in the right adnexa.

Histopathological examination identified bland mesothelial cells and no invasions (Figure 3A–3C), with immunohistochemistry showing no evidence of significant staining loss (Figure 3D, 3E). The differential diagnosis included well-differentiated papillary mesothelioma, epithelioid peritoneal mesothelioma, and florid mesothelial hyperplasia, and a well-differentiated papillary mesothelioma was highly suspected. The management plan recommending debulking surgery followed by HIPEC was proposed by the multidiscipline tumor board. However, the patient declined the suggested treatment due to her wish to preserve fertility. Six months after the initial presentation, she returned to the clinic for embryo transfer surgery, aiming to address her fertility aspirations. During the preoperative evaluation, the incidental finding of ascites prompted a referral to Taipei Medical University Hospital for further assessment. A CT scan performed on July 8, 2020 revealed a newly-enlarged right inguinal lymph node measuring 1.8 cm, along with a persistent soft-tissue density in the adnexa and a stable peritoneal nodule, with no significant interval changes compared to the CT scan conducted on February 26, 2020.

Subsequent diagnostic laparoscopy included the excision of tumors from the small intestine and omentum, with the tumors measuring 1 cm and 2 cm, respectively. Notably, the tumor in the omentum exhibited invasion into the muscularis propria. Additionally, miliary tumors were observed on the right pelvic peritoneum, cul-de-sac, and diaphragm, accompanied by the presence of dark-brownish ascites. Histopathological examination of specimens (Figure 4) from the peritoneum, large intestine, and small intestine revealed proliferative mesothelial cells with a finely granular chromatin pattern. Invasion into the subserosa and muscularis propria of the large intestine and omentum was confirmed. Immunohistochemical staining was positive for calretinin and BAP1, consistent with a final diagnosis of low-grade epithelioid peritoneal mesothelioma, staged as pT3NxMx (IIIB).

ONCOLOGICAL COURSE AND TREATMENT:

Following the diagnostic procedure, the patient elected to undergo chemotherapy instead of debulking surgery. She subsequently received 5 cycles of adjuvant chemotherapy, consisting of carboplatin at 5 AUC and paclitaxel at a dose of 175 mg/m2, administered every 3 weeks between August 2020 and November 2020. The chemotherapy regimen was generally well-tolerated, with the patient experiencing only minor adverse effects, notably alopecia. No significant adverse effects such as pancytopenia, nausea, or vomiting were reported during treatment.

However, a follow-up fluorodeoxyglucose-positron emission tomography (FDG-PET CT) scan performed on October 15, 2020 (Figure 5), during the chemotherapy course indicated disease progression. The scan revealed an increase in the size of the right inguinal lymph node, suggesting progression of the underlying pathology.

On November 5, 2020, the patient underwent a laparoscopic fertility-sparing surgery aimed at excising malignant tumors from the abdominal wall and omentum. This procedure included HIPEC and excision of the right inguinal tumor (Figure 6). During the operation, multiple miliary tumors, ranging from 0.1 to 1 cm in size, were identified and excised from the abdominal wall and peritoneum. The right inguinal lymph node was also excised, and HIPEC was performed with paclitaxel (175 mg/m2) for 90 minutes. Pathological analysis of the excised specimens confirmed the presence of epithelioid-type mesothelioma involving the right pelvic cavity and omentum (Figure 7).

After chemotherapy proved ineffective, the patient underwent surgery again, combined with HIPEC (hyperthermic intraperitoneal chemotherapy) to remove visible tumors. During shared decision-making regarding treatment options, the patient declined further chemotherapy (including pemetrexed) and opted for dual immunotherapy instead. She was started on immunotherapy, receiving 3 cycles of nivolumab (100 mg) and ipilimumab (50 mg) every 4 weeks from December 2020 to February 2021. Throughout the immunotherapy course, she tolerated the treatment well, without experiencing significant adverse effects such as nausea, vomiting, or fever. Additionally, her estrogen and follicle-stimulating hormone (FSH) levels remained within normal ranges, suggesting preserved ovarian function despite the aggressive treatment regimen. Subsequent follow-up imaging studies, including CT and MRI scans, demonstrated stable soft-tissue densities in the bilateral adnexa. Importantly, there were no signs of malignancy or metastasis in the lungs or brain, providing reassurance of disease stability.

IN VITRO FERTILIZATION AND OBSTETRIC COURSE:

With no evidence of disease recurrence or metastasis, the patient proceeded with IVF and embryo transfer. This intervention culminated in a successful pregnancy in May 2022. Comprehensive prenatal evaluations, conducted at local hospital, confirmed the absence of gestational diabetes mellitus, preeclampsia, or proteinuria. Additionally, high-resolution ultrasound examinations confirmed normal fetal anatomical development.

However, at 29 weeks of gestation, she had preterm premature rupture of membranes (PPROM). She was subsequently admitted to our hospital for tocolytic therapy, receiving ritodrine and nifedipine to inhibit uterine contractions. To prevent infection, antibiotics were administered alongside beta-methasone to accelerate fetal lung maturation and magnesium sulfate for neuroprotection. Despite 5 days of intensive tocolysis, the patient continued to experience frequent uterine contractions. Concurrently, the amniotic fluid index (AFI) decreased significantly from an initial measurement of 4.82 at admission to 0, accompanied by bloody vaginal discharge and uterine contraction. Given these developments, an emergency cesarean section was performed at a gestational age of 30 weeks 2 days on Dec. 12, 2022. She delivered a female infant who weighed 1378 g, with Apgar score 7/9 at 1 and 5 mins. The neonate was kept in the intensive care unit with oxygen therapy, which was withdrawn when the oxygen saturation level was greater than 90%. She continued to feed well and grow appropriately during her stay. No imaging studies were required and she was subsequently discharged.

Following the delivery, a secondary surgical exploration by gynecologic oncologists was conducted to evaluate any potential tumor progression or metastasis within the abdominal cavity. Intraoperatively, multiple tumor seedings were identified, ranging from 0.5 to 1.5 cm in size, distributed across the small intestine, mesentery, and peritoneum. These lesions were excised, along with residual tumors located on the rectum, peritoneal wall, and bladder base. Biopsies were obtained, and pathological examination confirmed the presence of epithelioid peritoneal mesothelioma. Postoperatively, the patient remained stable without any anticancer treatment and was discharged with a plan for outpatient follow-up. The most recent imaging studies, including CT and MRI scans, revealed a uterine myoma measuring 4.5 cm, with no further evidence of malignancy elsewhere. The treatment timeline is presented in Figure 8.

Discussion

CLINICAL PRESENTATION, DIAGNOSIS AND MANAGEMENT OF MALIGNANT PERITONEAL MESOTHELIOMA (MPM):

Malignant mesothelioma is a rare but highly aggressive neoplasm originating from the serosal membranes. Its prevalence varies by anatomical site, with the pleura being the most commonly affected, followed by the peritoneum, pericardium, and testis, in descending order of frequency [4,10]. In contrast to the male predominance observed in pleural mesothelioma, the prevalence of malignant peritoneal mesothelioma (MPM) in females ranges between 30% to 50% in the United States [11]. While MPM is often associated with industrial pollutants, particularly asbestos exposure, this association is less pronounced than in pleural mesothelioma [12–14]. This suggests that MPM can develop even in the absence of significant asbestos exposure, potentially due to germline BAP1 heterozygous mutations, which are hypothesized to increase susceptibility to low levels of asbestos exposure [15]. MPM is histologically classified into 3 subtypes: epithelioid, sarcomatous, and biphasic. Among these, the epithelioid subtype is the most prevalent and is associated with the most favorable prognosis [11,16]. MPM is classified into 2 grades: low-grade and high-grade [14].

However, the clinical presentation of MPM is often nonspecific, with symptoms such as abdominal distension, pain, early satiety, anorexia, nausea, and weight loss, which do not distinctly point to this malignancy [17]. As a result, MPM is frequently discovered incidentally during routine examinations or unrelated surgical procedures, leading to delayed diagnosis and, consequently, advanced disease with abdominal seeding in many patients [11,18].

When malignancy is suspected, initial imaging typically involves computed tomography (CT) or magnetic resonance imaging (MRI). While these modalities effectively identify areas of concern, they lack the specificity required to definitively diagnose the type of cancer [19].

Positron emission tomography (PET) scans are often employed for staging purposes but do not offer definitive identification of MPM. Given the low diagnostic specificity of imaging studies, the definitive diagnosis of MPM relies on tissue sampling via CT-guided core-needle biopsy or laparoscopic biopsy, followed by histopathological examination and immunohistochemical analysis [18].

Hematoxylin-eosin (H&E) staining provides initial identification of the MPM subtype but is insufficient for distinguishing MPM from other malignancies such as non-small cell lung carcinoma, renal cell carcinoma, and ovarian carcinoma [20]. Consequently, immunohistochemistry (IHC) is essential in differentiating mesothelioma from its malignant mimics and other mesothelial proliferations [14].

A comprehensive IHC panel, typically including at least 2 mesothelial markers and 2 carcinoma-related markers, is employed to confirm the diagnosis [14]. Commonly utilized mesothelial markers include calretinin, WT-1, podoplanin, BAP1, MTAP, and CDKN2A. These markers are crucial for confirming the mesothelial origin of the tumor. Additionally, carcinoma-related markers such as CEA, claudin-4, Ber-EP4, MOC-31, and TTF-1 are utilized to further refine the diagnosis, distinguishing MPM from other carcinomas [14,16]. This immunohistochemical approach not only aids in accurate diagnosis but also has prognostic implications, particularly in the context of determining the most appropriate therapeutic strategy.

In recent years, advances in molecular diagnostics have further enhanced our understanding of malignant peritoneal mesothelioma (MPM), providing deeper insights into its pathogenesis and potential therapeutic targets. Genetic alterations, particularly involving the BAP1 gene, have emerged as significant contributors to the development and progression of MPM. BAP1 (BRCA1-associated protein 1) is a tumor-suppressor gene that plays a crucial role in various cellular processes, including DNA repair, cell cycle regulation, and chromatin remodeling [21]. Loss-of-function mutations in BAP1 are found in many MPM cases, especially in those without a history of asbestos exposure, highlighting its role in the tumorigenesis of mesothelioma [22].

The identification of BAP1 mutations has not only provided insights into the molecular underpinnings of MPM but also opened avenues for personalized medicine. Patients with BAP1 mutations may benefit from targeted therapies aimed at exploiting specific vulnerabilities associated with this genetic alteration [23]. For instance, recent studies have explored the potential of PARP inhibitors, which target DNA repair pathways, in BAP1-mutated mesotheliomas [24]. These developments are a significant step forward in the treatment of MPM, which has historically been challenging due to the disease’s resistance to conventional chemotherapy and radiation therapy.

Another promising area of research in MPM management is the use of immunotherapy, particularly immune-checkpoint inhibitors. Immune-checkpoint blockade, which involves targeting inhibitory receptors such as PD-1, PD-L1, and CTLA-4 on T cells, has revolutionized the treatment of various solid tumors [25]. The potential synergy between immunotherapy and other modalities, such as cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (HIPEC), is also under investigation, with the goal of improving patient outcomes and prolonging survival [26].

Additionally, comprehensive genomic profiling can identify actionable mutations and inform the selection of targeted therapies, moving towards a more personalized treatment approach. Despite these advancements, the management of MPM remains complex and multifaceted, requiring a multidisciplinary approach. Optimal patient care involves not only accurate diagnosis and staging but also consideration of various treatment modalities, including surgery, chemotherapy, radiation, and emerging therapies such as targeted agents and immunotherapy. The ongoing development of clinical trials and translational research is critical to further refining therapeutic strategies and improving the prognosis of patients with MPM.

CRS AND HIPEC IN TREATMENT OF MALIGNANT PERITONEAL MESOTHELIOMA:

Malignant peritoneal mesothelioma (MPM) remains a challenging and aggressive malignancy with limited treatment options. The current therapeutic strategies for MPM include surgical intervention and systemic therapies [27], with cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) emerging as the cornerstone of treatment in select patients. This approach is particularly recommended for individuals with medically operable, diffuse peritoneal mesothelioma and those demonstrating good performance status, following a comprehensive multidisciplinary evaluation [27–29].

CRS aims to surgically remove all visible tumor deposits within the peritoneal cavity, thereby reducing the tumor burden to the lowest possible level. This is followed by HIPEC, which involves the circulation of heated chemotherapeutic agents directly within the abdominal cavity to target residual microscopic disease. The rationale behind HIPEC lies in the enhanced penetration of chemotherapeutic agents at elevated temperatures, which may also potentiate their cytotoxic effects on cancer cells. This combined modality treatment has been shown to significantly improve survival outcomes in patients with MPM [28,29].

Several studies have demonstrated the efficacy of CRS and HIPEC, with median overall survival for patients undergoing these procedures ranging from 38.4 to 63.2 months [29–33]. The 3-year and 5-year survival rates reported in the literature vary between 42% and 60% and 42% and 50.2%, respectively. These findings underscore the potential of CRS and HIPEC not only to extend survival but also to offer durable control of disease in a subset of patients with MPM [30–33].

The decision to pursue CRS and HIPEC is based on multiple factors, including the depth of tumor invasion, histological subtype, patient age, and overall health status [29]. The success of this approach is highly dependent on the completeness of cytoreduction, with more extensive tumor debulking correlating with better outcomes. However, the procedure is not without risks, and patient selection remains critical to optimizing results. The potential benefits of this aggressive therapy must be weighed against the risks, particularly in patients with comorbidities or advanced age.

SYSTEMIC THERAPY IN THE MANAGEMENT OF MALIGNANT PERITONEAL MESOTHELIOMA:

For patients with malignant peritoneal mesothelioma (MPM) who are not candidates for cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC), systemic therapy remains the cornerstone of treatment. This group includes individuals who are deemed inoperable due to factors such as extensive disease involvement, including bicavitary dissemination, or those with histological subtypes associated with a poorer prognosis, such as sarcomatoid or biphasic histology [34–36].

Systemic therapy typically involves use of chemotherapeutic agents aimed at controlling tumor growth, alleviating symptoms, and prolonging survival. Pemetrexed, a multi-targeted antifolate, has emerged as a key agent in the systemic treatment of MPM, particularly when combined with platinum-based chemotherapy such as cisplatin [37]. This regimen is commonly employed as a first-line therapy, especially for patients who are not eligible for surgical intervention [38].

Several clinical trials have evaluated the efficacy of pemetrexed-based regimens in MPM, both as monotherapy and in combination with cisplatin. These studies have reported median overall survival ranging from approximately 13 to 16.6 months, demonstrating the potential of systemic therapy to offer meaningful clinical benefits in this challenging patient population [34,36,39–41]. Additionally, the 1-year survival rates for patients receiving these regimens have been reported to range between 41.5% and 51.4% [42]. These outcomes, while modest compared to those achieved with CRS and HIPEC, underscore the importance of systemic therapy in managing patients with advanced or inoperable MPM.

In recent years, ongoing research has sought to optimize systemic treatment strategies for MPM, including the exploration of novel combinations and the integration of emerging therapies such as immunotherapy. Immune-checkpoint inhibitors targeting PD-1, PD-L1, and CTLA-4 have shown promise in other mesothelioma subtypes and are now being investigated in the context of peritoneal mesothelioma [43,44].

Despite these advances, the management of MPM with systemic therapy remains complex, and response rates vary depending on factors such as tumor biology and patient performance status. Personalized approaches that incorporate molecular profiling and the identification of predictive biomarkers are increasingly recognized as essential in guiding treatment decisions and maximizing therapeutic efficacy.

THE ROLE OF IMMUNE-CHECKPOINT INHIBITORS IN TREATMENT OF PERITONEAL MESOTHELIOMA:

The exploration of ICIs in the treatment of peritoneal mesothelioma, although constrained by small sample sizes, has shown promising potential. Immune-checkpoint blockade, particularly targeting the PD-1/PD-L1 axis, has garnered significant attention due to its effectiveness in other malignancies, and recent studies have sought to extend these benefits to peritoneal mesothelioma. Atezolizumab, a PD-L1 inhibitor, has been evaluated in combination with bevacizumab, an anti-VEGF agent, as a second-line therapy for patients with peritoneal mesothelioma. In this setting, the combination therapy demonstrated a 1-year overall survival rate of 85% and a median response duration of 12.8 months [45]. This regimen appears particularly promising given the typically poor prognosis associated with peritoneal mesothelioma and the limited efficacy of existing therapies.

Avelumab, another PD-L1 inhibitor, has also shown potential as a second-line treatment. Notably, its efficacy appears to be enhanced in tumors that express PD-L1, with these patients exhibiting a higher response rate compared to those without PD-L1 expression [46]. This finding underscores the importance of biomarker-driven therapy in optimizing treatment outcomes for peritoneal mesothelioma.

In addition to PD-1/PD-L1 inhibitors, CTLA-4 inhibitors such as ipilimumab have been investigated for their therapeutic potential. Ipilimumab has been studied both as a monotherapy and in combination with other ICIs such as nivolumab. These combination regimens have shown significant clinical benefit, with median overall survival ranging from 10.2 to 19 months and a 1-year survival rate of 68% [47,48]. The combination of ipilimumab and nivolumab, in particular, is a synergistic approach that enhances the anti-tumor immune response, offering hope for improved outcomes in this challenging malignancy.

Despite these encouraging results, the limited data available on the use of ICIs in peritoneal mesothelioma preclude their widespread adoption as first-line therapies. Current evidence has not conclusively demonstrated that ICIs alone or in combination are superior to traditional chemotherapy in the initial treatment setting. However, the U.S. Food and Drug Administration (FDA) approved the combination of the PD-1 inhibitor nivolumab and the CTLA-4 inhibitor ipilimumab for unresectable pleural mesothelioma in 2020, suggesting a potential future role for similar strategies in peritoneal mesothelioma.

This approval for pleural mesothelioma may pave the way for broader applications in peritoneal mesothelioma, particularly as more data become available. Ongoing clinical trials and expanded studies are crucial to determining the optimal use of ICIs in this context and may eventually establish these therapies as a standard part of the treatment arsenal against peritoneal mesothelioma.

IMMUNE-RELATED ADVERSE EVENTS (IRAES) AND FERTILITY CONSIDERATIONS IN IMMUNOTHERAPY:

While ICIs have revolutionized the treatment landscape for various malignancies, the potential for irAEs remains a significant concern that must be carefully managed [49,50]. irAEs arise from the immune system’s hyperactivation, leading to off-target effects that can impact multiple organ systems. The incidence and severity of irAEs vary, with dermatologic toxicities such as pruritus and rash being among the most common manifestations [51].

Other common manifestations include lower digestive tract issues [52,53] (eg, diarrhea, colitis), hepatotoxicity [54] and endocrinopathies [55,56] (eg, hypophysitis, thyroid dysfunction). Despite the clinical effectiveness of treatment, irAEs cannot be ignored. The impact of irAEs is broad, affecting various organ systems through immunological enhancement and suppression. One of the most common irAEs is dermatologic toxicity, such as pruritus and rash [51].

The cardiovascular system can also be affected by irAEs, but these events are less common than dermatologic or gastrointestinal manifestations. Cardiotoxicity associated with ICIs may present as myocarditis, pericarditis, or even arrhythmias, and while rare, these conditions can be severe and potentially life-threatening. Cardiovascular irAEs require prompt recognition and management, often necessitating discontinuation of ICIs and initiation of immunosuppressive therapy [57,58].

Renal toxicity is another important irAE that clinicians must monitor for. Nephritis induced by ICIs can lead to acute kidney injury, which may necessitate cessation of therapy and the use of corticosteroids or other immunosuppressants to mitigate damage [59]. The renal system’s vulnerability to irAEs underscores the need for regular monitoring of renal function throughout the course of ICI therapy.

Pulmonary toxicity, including pneumonitis, is a well-documented irAE and can range from mild interstitial changes to severe, life-threatening inflammation requiring hospitalization. Patients presenting with respiratory symptoms such as cough, dyspnea, or chest pain during ICI therapy should be promptly evaluated for pneumonitis, with imaging studies and pulmonary function tests as necessary. The management of pneumonitis often involves corticosteroids and discontinuation of ICIs, with the decision to resume therapy carefully weighed against the risks of recurrence [60,61].

In the context of endocrinopathies, irAEs can lead to a wide spectrum of dysfunctions involving the pituitary, thyroid, adrenal glands, and pancreas. Hypophysitis, a common endocrine-related irAE, may present with symptoms of headache, fatigue, and visual disturbances and can result in secondary adrenal insufficiency [62]. Thyroid dysfunction, including hypothyroidism and hyperthyroidism, is frequently observed and may necessitate lifelong thyroid hormone replacement therapy [63]. Furthermore, adrenalitis can manifest as adrenal insufficiency, requiring prompt glucocorticoid replacement. The impact of these endocrine irAEs can be profound, affecting the patient’s quality of life and requiring ongoing management even after discontinuation of ICIs [64].

The reproductive system is not immune to the effects of irAEs. While data on the direct impact of ICIs on fertility are still emerging, there is growing concern about potential gonadal toxicity, particularly in women of childbearing age. Fertility preservation strategies, including sperm banking for men and oocyte or embryo cryopreservation for women, are recommended before initiating ICI therapy in patients with reproductive concerns. These proactive measures can help mitigate the potential long-term consequences of treatment on fertility [65].

For pregnant patients or those planning pregnancy, the use of ICIs poses unique challenges. Given the crucial role of the immune system in maintaining fetomaternal tolerance, the administration of ICIs during pregnancy raises concerns about the potential for immune-mediated pregnancy complications such as spontaneous abortion, preterm labor, or fetal growth restriction. Recently, human studies have shown that ICI use during pregnancy may be better tolerated than previously suspected [66].

In clinical practice, the decision to use ICIs in pregnant patients must be made on a case-by-case basis, with a thorough assessment of the risks and benefits. For women undergoing ICI therapy who are considering pregnancy, it is generally advised to delay conception until at least 6 months after the completion of treatment to allow for resolution of irAEs and stabilization of the immune system [67]. This precaution helps to minimize potential risks to both the mother and the fetus. Additionally, healthcare providers should engage in discussions with patients about fertility preservation options before starting ICI therapy, especially in younger patients who may wish to have children in the future.

Conclusions

This report presents the case of a 44-year-old nulliparous woman diagnosed with low-grade epithelioid MPM, discovered incidentally during a laparotomic myomectomy. Initial imaging and histopathological assessments confirmed the diagnosis, with immunohistochemistry revealing positivity for calretinin and BAP1. Despite the recommendation for debulking surgery and HIPEC, the patient opted for fertility preservation, influencing her treatment decisions. Following chemotherapy and immunotherapy, the patient delivered a premature female baby with healthy outcome. This review illustrates the delicate balance between managing a serious cancer diagnosis and preserving fertility. It highlights the importance of personalized treatment plans, especially when patients face challenging decisions that impact both their health and future family plans. The successful outcome in this case, with the patient delivering a healthy baby, underscores the potential for favorable results of immunotherapy even in complex situations.

Figures

Schematic diagram of the tumor invasion site. The stage IIIb tumors have invaded deeper tissues in nearby organs and distant lymph nodes. Sites involved by peritoneal mesothelioma: peritoneum, abdominal wall, omentum, lymph nodes, large intestine, mesentery and bladder.Figure 1.. Schematic diagram of the tumor invasion site. The stage IIIb tumors have invaded deeper tissues in nearby organs and distant lymph nodes. Sites involved by peritoneal mesothelioma: peritoneum, abdominal wall, omentum, lymph nodes, large intestine, mesentery and bladder. Postoperative pelvic CT scan (February 26th, 2020). (A) The scan reveals moderate ascites (white arrow) and nonspecific peritoneal thickening. (B) The scan shows small enhancing nodules on the peritoneal membrane, with notable soft-tissue thickening in the right adnexa compared to the left.Figure 2.. Postoperative pelvic CT scan (February 26th, 2020). (A) The scan reveals moderate ascites (white arrow) and nonspecific peritoneal thickening. (B) The scan shows small enhancing nodules on the peritoneal membrane, with notable soft-tissue thickening in the right adnexa compared to the left. Pathology report of pelvic tissue (February 17th, 2020). (A–C). Histology showed bland mesothelial cells and no invasions. The diagnosis of well-differentiated papillary mesothelioma was highly suspected. (D, E) Immunohistochemical analysis demonstrated diffuse positivity for (D) calretinin and (E) BAP1. Notably, the BAP1 stain indicated a preserved nuclear staining pattern.Figure 3.. Pathology report of pelvic tissue (February 17th, 2020). (A–C). Histology showed bland mesothelial cells and no invasions. The diagnosis of well-differentiated papillary mesothelioma was highly suspected. (D, E) Immunohistochemical analysis demonstrated diffuse positivity for (D) calretinin and (E) BAP1. Notably, the BAP1 stain indicated a preserved nuclear staining pattern. Pathology report (July 16th, 2020). (A) Histopathological examination of the peritoneal specimen revealed mild nuclear atypia with a finely granular chromatin pattern. (B, C) Immunohistochemical (IHC) staining demonstrated both nuclear and cytoplasmic positivity for calretinin, with preserved nuclear staining observed for BAP1. (D, E) The specimen from the large intestine exhibited papillary growth and glandular differentiation. Notably, there was evidence of stromal invasion extending into the subserosa and muscularis propria, indicative of malignant transformation.Figure 4.. Pathology report (July 16th, 2020). (A) Histopathological examination of the peritoneal specimen revealed mild nuclear atypia with a finely granular chromatin pattern. (B, C) Immunohistochemical (IHC) staining demonstrated both nuclear and cytoplasmic positivity for calretinin, with preserved nuclear staining observed for BAP1. (D, E) The specimen from the large intestine exhibited papillary growth and glandular differentiation. Notably, there was evidence of stromal invasion extending into the subserosa and muscularis propria, indicative of malignant transformation. FDG-PET CT interpretation (October 15th, 2020). (A–C) The PET CT shows metabolic activity in the pelvis and a 1.2-cm low-FDG nodule in the umbilical area, consistent with post-treatment response in peritoneal mesothelioma.Figure 5.. FDG-PET CT interpretation (October 15th, 2020). (A–C) The PET CT shows metabolic activity in the pelvis and a 1.2-cm low-FDG nodule in the umbilical area, consistent with post-treatment response in peritoneal mesothelioma. Laparoscopic fertility-sparing surgery with HIPEC (November 5th, 2020): Intraoperative findings and surgical notes. (A) The PCI score was 14, indicating extensive peritoneal involvement, with tumors mainly in the pelvis and lower abdomen. The diagram shows numerous nodules over 1 cm. (B) Surgical exploration revealed multiple miliary tumors (0.1–1 cm) scattered across the peritoneum, mesentery, omentum, and abdominal wall, indicating widespread tumor dissemination. (C) The images show a hydatidiform lesion in the upper quadrant and tumor spread into the pelvic region, highlighting extensive abdominal disease (ovaries).Figure 6.. Laparoscopic fertility-sparing surgery with HIPEC (November 5th, 2020): Intraoperative findings and surgical notes. (A) The PCI score was 14, indicating extensive peritoneal involvement, with tumors mainly in the pelvis and lower abdomen. The diagram shows numerous nodules over 1 cm. (B) Surgical exploration revealed multiple miliary tumors (0.1–1 cm) scattered across the peritoneum, mesentery, omentum, and abdominal wall, indicating widespread tumor dissemination. (C) The images show a hydatidiform lesion in the upper quadrant and tumor spread into the pelvic region, highlighting extensive abdominal disease (ovaries). Pathological findings (November 6th, 2020). (A) The pelvic specimen shows marked adhesion and stromal invasion with a papillary growth pattern, consistent with epithelioid mesothelioma. (B) Immunohistochemical staining confirmed the diagnosis, showing strong, diffuse positivity for calretinin, indicating a mesothelial origin of the tumor. (C) WT-1 and (D) BAP1 were positive. WT-1 is linked to mesotheliomas, while BAP1 positivity, uncommon in aggressive cases, indicates a less aggressive tumor subtype.Figure 7.. Pathological findings (November 6th, 2020). (A) The pelvic specimen shows marked adhesion and stromal invasion with a papillary growth pattern, consistent with epithelioid mesothelioma. (B) Immunohistochemical staining confirmed the diagnosis, showing strong, diffuse positivity for calretinin, indicating a mesothelial origin of the tumor. (C) WT-1 and (D) BAP1 were positive. WT-1 is linked to mesotheliomas, while BAP1 positivity, uncommon in aggressive cases, indicates a less aggressive tumor subtype. Treatment timeline of the patient.Figure 8.. Treatment timeline of the patient.

References:

1.. Sousa B, Silva J, Monteiro N, Malignant peritoneal mesothelioma: A case report: Cureus, 2023; 15(8); e42902

2.. Levy M, Boublikova L, Buchler T, Simsa J, Treatment of malignant peritoneal mesothelioma: Klin Onkol, 2019; 32(5); 333-37

3.. Pandav V, Patel J, Inampudi JV, Incidence of additional primary malignancies in patients with malignant peritoneal mesothelioma: J Clin Oncol, 2023; 41(16 Suppl.); e20535-e35

4.. van Kooten JP, Belderbos RA, von der Thusen JH, Incidence, treatment and survival of malignant pleural and peritoneal mesothelioma: A population-based study: Thorax, 2022; 77(12); 1260-67

5.. Idkedek M, Tahayneh KS, Abu-Akar F, Bakri IA, Case report and review of literature: familial malignant pleural mesothelioma in a 39 years old patient with an inconclusive (18)F-FDG PET/CT result: Front Surg, 2022; 9; 819596

6.. Zhao F, Zhang YL, Liu X, [A case of malignant peritoneal mesothelioma.]: Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi, 2023; 41(4); 307-9 [in Chinese]

7.. Marques de Sousa S, Pereira F, Duarte M, Malignant peritoneal mesothelioma as a rare cause of dyspeptic complaints and ascites: A diagnostic challenge: GE Port J Gastroenterol, 2020; 27(3); 197-202

8.. Salo SAS, Ilonen I, Laaksonen S, Malignant peritoneal mesothelioma: Treatment options and survival: Anticancer Res, 2019; 39(2); 839-45

9.. de Boer NL, van Kooten JP, Burger JWA, Adjuvant dendritic cell based immunotherapy (DCBI) after cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) for peritoneal mesothelioma, a phase II single centre open-label clinical trial: rationale and design of the MESOPEC trial: BMJ Open, 2019; 9(5); e026779

10.. Mezei G, Chang ET, Mowat FS, Moolgavkar SH, Epidemiology of mesothelioma of the pericardium and tunica vaginalis testis: Ann Epidemiol, 2017; 27(5); 348-59.e11

11.. Kim J, Bhagwandin S, Labow DM, Malignant peritoneal mesothelioma: A review: Ann Transl Med, 2017; 5(11); 236

12.. Zhao Z, Li J, Tan F, Assessing the global burden of mesothelioma: Trends, socioeconomic influences, and asbestos exposure: A retrospective cohort study.: Int J Surg., 2024 [Online ahead of print]

13.. Boffetta P, Epidemiology of peritoneal mesothelioma: A review: Ann Oncol, 2007; 18(6); 985-90

14.. Husain AN, Chapel DB, Attanoos R, Guidelines for pathologic diagnosis of mesothelioma: 2023 Update of the consensus statement from the International Mesothelioma Interest Group: Arch Pathol Lab Med, 2024; 148(11); 1251-71

15.. Napolitano A, Pellegrini L, Dey A, Minimal asbestos exposure in germline BAP1 heterozygous mice is associated with deregulated inflammatory response and increased risk of mesothelioma: Oncogene, 2016; 35(15); 1996-2002

16.. Robinson BW, Musk AW, Lake RA, Malignant mesothelioma: Lancet, 2005; 366(9483); 397-408

17.. Acherman YI, Welch LS, Bromley CM, Sugarbaker PH, Clinical presentation of peritoneal mesothelioma: Tumori, 2003; 89(3); 269-73

18.. Sugarbaker PH, Yan H, Grazi RV, Shmookler BM, Early localized peritoneal mesothelioma as an incidental finding at laparoscopy. Report of a case and implications regarding natural history of the disease: Cancer, 2000; 89(6); 1279-84

19.. Kato K, Gemba K, Fujimoto N, Computed tomographic features of malignant peritoneal mesothelioma: Anticancer Res, 2016; 36(3); 1067-72

20.. Carbone M, Shimizu D, Napolitano A, Positive nuclear BAP1 immunostaining helps differentiate non-small cell lung carcinomas from malignant mesothelioma: Oncotarget, 2016; 7(37); 59314-21

21.. Kwon J, Lee D, Lee S-A, BAP1 as a guardian of genome stability: Implications in human cancer: Exp Mol Med, 2023; 55(4); 745-54

22.. Rossi G, Righi L, Barbisan F, BAP1 loss, nuclear grading, and nonepithelioid features in the diagnosis of mesothelioma in Italy: Nevermore without the pathology report: J Pers Med, 2024; 14(4); 394

23.. Carbone M, Minaai M, Takinishi Y, Preventive and therapeutic opportunities: Targeting BAP1 and/or HMGB1 pathways to diminish the burden of mesothelioma: J Transl Med, 2023; 21(1); 749

24.. Ghafoor A, Mian I, Wagner C, Phase 2 study of olaparib in malignant mesothelioma and correlation of efficacy with germline or somatic mutations in BAP1 gene: JTO Clin Res Rep, 2021; 2(10); 100231

25.. Calabrò L, Bronte G, Grosso F, Immunotherapy of mesothelioma: The evolving change of a long-standing therapeutic dream: Front Immunol, 2023; 14; 1333661

26.. Broeckx G, Pauwels P, Malignant peritoneal mesothelioma: A review: Transl Lung Cancer Res, 2018; 7(5); 537-42

27.. Kusamura S, Baratti D, De Simone M, Diagnostic and therapeutic pathway in diffuse malignant peritoneal mesothelioma: Cancers (Basel), 2023; 15(3); 662

28.. Feldman AL, Libutti SK, Pingpank JF, Analysis of factors associated with outcome in patients with malignant peritoneal mesothelioma undergoing surgical debulking and intraperitoneal chemotherapy: J Clin Oncol, 2003; 21(24); 4560-67

29.. Yan TD, Brun EA, Cerruto CA, Prognostic indicators for patients undergoing cytoreductive surgery and perioperative intraperitoneal chemotherapy for diffuse malignant peritoneal mesothelioma: Ann Surg Oncol, 2007; 14(1); 41-49

30.. Yan TD, Deraco M, Baratti D, Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for malignant peritoneal mesothelioma: Multi-institutional experience: J Clin Oncol, 2009; 27(36); 6237-42

31.. Baratti D, Kusamura S, Cabras AD, Diffuse malignant peritoneal mesothelioma: long-term survival with complete cytoreductive surgery followed by hyperthermic intraperitoneal chemotherapy (HIPEC): Eur J Cancer, 2013; 49(15); 3140-48

32.. Acs M, Gerken M, Gajic I, Ten-year single-center experience with treatment of primary diffuse malignant peritoneal mesothelioma (DMPM) by cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC): Langenbecks Arch Surg, 2022; 407(7); 3057-67

33.. Helm JH, Miura JT, Glenn JA, Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy for malignant peritoneal mesothelioma: A systematic review and meta-analysis: Ann Surg Oncol, 2015; 22(5); 1686-93

34.. Santoro A, O’Brien ME, Stahel RA, Pemetrexed plus cisplatin or pemetrexed plus carboplatin for chemonaïve patients with malignant pleural mesothelioma: Results of the International Expanded Access Program: J Thorac Oncol, 2008; 3(7); 756-63

35.. Jänne PA, Wozniak AJ, Belani CP, Pemetrexed alone or in combination with cisplatin in previously treated malignant pleural mesothelioma: Outcomes from a phase IIIB expanded access program: J Thorac Oncol, 2006; 1(6); 506-12

36.. Obasaju CK, Ye Z, Wozniak AJ, Single-arm, open label study of pemetrexed plus cisplatin in chemotherapy naïve patients with malignant pleural mesothelioma: Outcomes of an expanded access program: Lung Cancer, 2007; 55(2); 187-94

37.. Yoshikawa Y, Kuribayashi K, Minami T, Epigenetic alterations and biomarkers for immune checkpoint inhibitors-current standards and future perspectives in malignant pleural mesothelioma treatment: Front Oncol, 2020; 10; 554570

38.. Votanopoulos KI, Sugarbaker P, Deraco M, Is Cytoreductive surgery with hyperthermic intraperitoneal chemotherapy justified for biphasic variants of peritoneal mesothelioma? Outcomes from the Peritoneal Surface Oncology Group International Registry: Ann Surg Oncol, 2018; 25(3); 667-73

39.. Jänne PA, Wozniak AJ, Belani CP, Open-label study of pemetrexed alone or in combination with cisplatin for the treatment of patients with peritoneal mesothelioma: Outcomes of an expanded access program: Clin Lung Cancer, 2005; 7(1); 40-46

40.. Fujimoto E, Kijima T, Kuribayashi K, First-line chemotherapy with pemetrexed plus cisplatin for malignant peritoneal mesothelioma: Expert Rev Anticancer Ther, 2017; 17(9); 865-72

41.. Nagata Y, Sawada R, Takashima A, Efficacy and safety of pemetrexed plus cisplatin as first-line chemotherapy in advanced malignant peritoneal mesothelioma: Jpn J Clin Oncol, 2019; 49(11); 1004-8

42.. Carteni G, Manegold C, Garcia GM, Malignant peritoneal mesothelioma – results from the International Expanded Access Program using pemetrexed alone or in combination with a platinum agent: Lung Cancer, 2009; 64(2); 211-18

43.. Gribkova YV, Alexander HR, Management of patients with malignant peritoneal mesothelioma: The evolving role of surgery and immunotherapy: Medical Research Archives, 2022; 10(11) 193546531

44.. Zhou M, Joshi N, Raj KP, PD-1/PD-L1 checkpoint inhibitor immunotherapy for malignant pleural mesothelioma: Case series and literature review: Clin Lung Cancer, 2021; 22(3); e329-e35

45.. Raghav K, Liu S, Overman MJ, Efficacy, safety, and biomarker analysis of combined PD-L1 (atezolizumab) and VEGF (bevacizumab) blockade in advanced malignant peritoneal mesothelioma: Cancer Discov, 2021; 11(11); 2738-47

46.. Hassan R, Thomas A, Nemunaitis JJ, Efficacy and safety of avelumab treatment in patients with advanced unresectable mesothelioma: Phase 1b results from the JAVELIN solid tumor trial: JAMA Oncol, 2019; 5(3); 351-57

47.. Raghav K, Liu S, Overman M, Clinical efficacy of immune checkpoint inhibitors in patients with advanced malignant peritoneal mesothelioma: JAMA Netw Open, 2021; 4(8); e2119934

48.. Fennell DA, Ewings S, Ottensmeier C, Nivolumab versus placebo in patients with relapsed malignant mesothelioma (CONFIRM): A multicentre, double-blind, randomised, phase 3 trial: Lancet Oncol, 2021; 22(11); 1530-40

49.. Conroy M, Naidoo J, Immune-related adverse events and the balancing act of immunotherapy: Nat Commun, 2022; 13(1); 392

50.. Choi J, Lee SY, Clinical characteristics and treatment of immune-related adverse events of immune checkpoint inhibitors: Immune Netw, 2020; 20(1); e9

51.. Villadolid J, Amin A, Immune checkpoint inhibitors in clinical practice: Update on management of immune-related toxicities: Transl Lung Cancer Res, 2015; 4(5); 560-75

52.. Hodi FS, O’Day SJ, McDermott DF, Improved survival with ipilimumab in patients with metastatic melanoma: N Engl J Med, 2010; 363(8); 711-23

53.. Robert C, Thomas L, Bondarenko I, Ipilimumab plus dacarbazine for previously untreated metastatic melanoma: N Engl J Med, 2011; 364(26); 2517-26

54.. Robert C, Schachter J, Long GV, Pembrolizumab versus ipilimumab in advanced melanoma: N Engl J Med, 2015; 372(26); 2521-32

55.. Cukier P, Santini FC, Scaranti M, Hoff AO, Endocrine side effects of cancer immunotherapy: Endocr Relat Cancer, 2017; 24(12); T331-47

56.. de Filette J, Jansen Y, Schreuer M, Incidence of thyroid-related adverse events in melanoma patients treated with pembrolizumab: J Clin Endocrinol Metab, 2016; 101(11); 4431-39

57.. Mahmood SS, Fradley MG, Cohen JV, Myocarditis in patients treated with immune checkpoint inhibitors: J Am Coll Cardiol, 2018; 71(16); 1755-64

58.. Ganatra S, Neilan TG, Immune checkpoint inhibitor-associated myocarditis: Oncologist, 2018; 23(8); 879-86

59.. Cortazar FB, Marrone KA, Troxell ML, Clinicopathological features of acute kidney injury associated with immune checkpoint inhibitors: Kidney Int, 2016; 90(3); 638-47

60.. Naidoo J, Wang X, Woo KM, Pneumonitis in patients treated with anti-programmed death-1/programmed death ligand 1 therapy: J Clin Oncol, 2017; 35(7); 709-17

61.. Nishino M, Sholl LM, Hodi FS, Anti-PD-1-related pneumonitis during cancer immunotherapy: N Engl J Med, 2015; 373(3); 288-90

62.. Albarel F, Gaudy C, Castinetti F, Long-term follow-up of ipilimumab-induced hypophysitis, a common adverse event of the anti-CTLA-4 antibody in melanoma: Eur J Endocrinol, 2015; 172(2); 195-204

63.. Lee CH, Motzer RJ, Immune checkpoint therapy in renal cell carcinoma: Cancer J, 2016; 22(2); 92-95

64.. Remon J, Planchard D, AZD9291 in EGFR-mutant advanced non-small-cell lung cancer patients: Future Oncol, 2015; 11(22); 3069-81

65.. Meirow D, Nugent D, The effects of radiotherapy and chemotherapy on female reproduction: Hum Reprod Update, 2001; 7(6); 535-43

66.. Gougis P, Hamy A-S, Jochum F, Immune checkpoint inhibitor use during pregnancy and outcomes in pregnant individuals and newborns: JAMA Network Open, 2024; 7(4); e245625

67.. Tripathy D, Im SA, Colleoni M, Ribociclib plus endocrine therapy for premenopausal women with hormone-receptor-positive, advanced breast cancer (MONALEESA-7): A randomised phase 3 trial: Lancet Oncol, 2018; 19(7); 904-15

Figures

Figure 1.. Schematic diagram of the tumor invasion site. The stage IIIb tumors have invaded deeper tissues in nearby organs and distant lymph nodes. Sites involved by peritoneal mesothelioma: peritoneum, abdominal wall, omentum, lymph nodes, large intestine, mesentery and bladder.Figure 2.. Postoperative pelvic CT scan (February 26th, 2020). (A) The scan reveals moderate ascites (white arrow) and nonspecific peritoneal thickening. (B) The scan shows small enhancing nodules on the peritoneal membrane, with notable soft-tissue thickening in the right adnexa compared to the left.Figure 3.. Pathology report of pelvic tissue (February 17th, 2020). (A–C). Histology showed bland mesothelial cells and no invasions. The diagnosis of well-differentiated papillary mesothelioma was highly suspected. (D, E) Immunohistochemical analysis demonstrated diffuse positivity for (D) calretinin and (E) BAP1. Notably, the BAP1 stain indicated a preserved nuclear staining pattern.Figure 4.. Pathology report (July 16th, 2020). (A) Histopathological examination of the peritoneal specimen revealed mild nuclear atypia with a finely granular chromatin pattern. (B, C) Immunohistochemical (IHC) staining demonstrated both nuclear and cytoplasmic positivity for calretinin, with preserved nuclear staining observed for BAP1. (D, E) The specimen from the large intestine exhibited papillary growth and glandular differentiation. Notably, there was evidence of stromal invasion extending into the subserosa and muscularis propria, indicative of malignant transformation.Figure 5.. FDG-PET CT interpretation (October 15th, 2020). (A–C) The PET CT shows metabolic activity in the pelvis and a 1.2-cm low-FDG nodule in the umbilical area, consistent with post-treatment response in peritoneal mesothelioma.Figure 6.. Laparoscopic fertility-sparing surgery with HIPEC (November 5th, 2020): Intraoperative findings and surgical notes. (A) The PCI score was 14, indicating extensive peritoneal involvement, with tumors mainly in the pelvis and lower abdomen. The diagram shows numerous nodules over 1 cm. (B) Surgical exploration revealed multiple miliary tumors (0.1–1 cm) scattered across the peritoneum, mesentery, omentum, and abdominal wall, indicating widespread tumor dissemination. (C) The images show a hydatidiform lesion in the upper quadrant and tumor spread into the pelvic region, highlighting extensive abdominal disease (ovaries).Figure 7.. Pathological findings (November 6th, 2020). (A) The pelvic specimen shows marked adhesion and stromal invasion with a papillary growth pattern, consistent with epithelioid mesothelioma. (B) Immunohistochemical staining confirmed the diagnosis, showing strong, diffuse positivity for calretinin, indicating a mesothelial origin of the tumor. (C) WT-1 and (D) BAP1 were positive. WT-1 is linked to mesotheliomas, while BAP1 positivity, uncommon in aggressive cases, indicates a less aggressive tumor subtype.Figure 8.. Treatment timeline of the patient.

In Press

Case report  China

Telitacicept and Tacrolimus Synergy in Managing Refractory Primary Membranous Nephropathy: Case Insights

Am J Case Rep In Press; DOI: 10.12659/AJCR.946727  

0:00

Case report  Denmark

Neonatal Congenital Myasthenic Syndrome Linked to CHAT Gene Variants: A Case Report and Treatment Insights

Am J Case Rep In Press; DOI: 10.12659/AJCR.946839  

0:00

Case report  Japan

Arm-Behind-the-Back Position for Breast Cancer Radiotherapy in Patients with Lupus Erythematosus and Should...

Am J Case Rep In Press; DOI: 10.12659/AJCR.946674  

0:00

Case report  Italy

Multiple Myeloma Presenting as Breast Metastasis: Diagnostic Challenges and Case Analysis

Am J Case Rep In Press; DOI: 10.12659/AJCR.945853  

0:00

Most Viewed Current Articles

21 Jun 2024 : Case report  China (mainland) 96,182

Intracranial Parasitic Fetus in a Living Infant: A Case Study with Surgical Intervention and Prognosis Anal...

DOI :10.12659/AJCR.944371

Am J Case Rep 2024; 25:e944371

0:00

07 Mar 2024 : Case report  USA 52,159

Neurocysticercosis Presenting as Migraine in the United States

DOI :10.12659/AJCR.943133

Am J Case Rep 2024; 25:e943133

0:00

20 Nov 2023 : Case report  Saudi Arabia 31,026

Azithromycin Treatment for Acne Vulgaris: A Case Report on the Risk of Clostridioides difficile Infection

DOI :10.12659/AJCR.941424

Am J Case Rep 2023; 24:e941424

0:00

18 Feb 2024 : Case report  Japan 23,365

A Case of Thoracic Empyema Caused by Actinomyces naeslundii

DOI :10.12659/AJCR.943030

Am J Case Rep 2024; 25:e943030

0:00

Your Privacy

We use cookies to ensure the functionality of our website, to personalize content and advertising, to provide social media features, and to analyze our traffic. If you allow us to do so, we also inform our social media, advertising and analysis partners about your use of our website, You can decise for yourself which categories you you want to deny or allow. Please note that based on your settings not all functionalities of the site are available. View our privacy policy.

American Journal of Case Reports eISSN: 1941-5923
American Journal of Case Reports eISSN: 1941-5923