Skip to main content

Pericardial malignant infiltration as the cause of sudden death of a patient with metastatic urothelial carcinoma treated with atezolizumab

Abstract

Background

Muscle-infiltrating urothelial carcinoma of the bladder is the most common genitourinary cancer. Immunotherapeutic agents targeting protein-1 programmed death or protein-1 programmed death ligand are currently considered the standard treatment in patients with either inoperable locally advanced or metastatic urothelial carcinoma (MUC) after platinum-based chemotherapy failure.

Case presentation

Here we report the case of a Caucasian male patient with metastatic urothelial carcinoma treated with second-line atezolizumab within a trial who achieved complete response by computed tomography (CT), but suddenly died due to cardiac tamponade resulting from malignant pericardial infiltration. Histopathology confirmed this as the only site of disease progression.

Conclusions

Cardiovascular toxicity of atezolizumab was considered within differential diagnoses, however histopathological examination revealed progression of malignancy in the pericardium as the cause of the sudden death. This is the first published case report of a patient treated with second-line atezolizumab in whom the rare disease progression of pericardial infiltration was confirmed. Despite its rarity, the clinicians should always consider the possibility of pericardial metastases.

Peer Review reports

Background

Bladder carcinoma is the ninth most common malignancy with an overall incidence of 20.4 cases per 100,000 population. The most common bladder cancer is urothelial carcinoma, which accounts for 90% of all malignant bladder tumours [1]. The most common sites of metastases are extra loco-regional lymph nodes, bones, lungs, liver, and peritoneum. Pericardial metastases, while quite rare, present a risk of sudden cardiac death [2]. In this case report, we present a 66-year-old patient with metastatic urothelial carcinoma (MUC) treated with atezolizumab as second-line within the phase 3 SAUL study (NCT02928406).

Despite computed tomography (CT) showing a complete response, the patient died suddenly as a result of cardiac tamponade caused by metastases to the pericardium, the only site of disease progression that could be identified.

To the best of our knowledge this is the first case report of isolated pericardial metastases in a patient with metastatic urothelial carcinoma (MUC) treated with immunotherapy based on protein-1 programmed death (PD-1) or PD-L1 (protein-1 programmed death ligand) inhibition.

Case presentation

A 66-year-old Caucasian male underwent right-sided uretero-nephrectomy due to high-grade muscle-infiltrating urothelial carcinoma, pT2N0M0, stage II in October 2006 and radical cystectomy with Mainz pouch II urinary diversion for high-grade muscle-infiltrating urothelial bladder carcinoma, pT2N0M0, stage II in April 2010. The malignant urothelial cells from bladder were positive for cytokeratin CK7 and cytokeratin CK20, and showed negativity for p63, vimentin and calretinin. The patient was observed by a urologist.

On March 3, 2017, an enlarged lymph node in the right groin was surgically removed and relapse of urothelial carcinoma was confirmed by histopathology examination. Abdominal, pelvic and thoracic computed tomography (apt-CT) with contrast was performed on March 27, 2017. This showed lymphadenopathy (LAP) in the left supraclavicular area, in the retroperitoneum, bilaterally in the para-iliac area, and in both groins. Bajorin prognostic score (performance status > 1 and the presence of visceral involvement) was zero in this patient. Median survival of patients with no negative prognostic factors was 33.0 months in this study by Bajorin et al. [3].

From March 28, 2017 to July 18, 2017, this patient underwent six courses of gemcitabine (G, 2000 mg/m2 intravenously on days 1 and 8) with cisplatin (C, 70 mg/m2 intravenously on day 1) without any serious adverse event of grades 3 and 4. Only grade 1, anemia and fatigue were recorded. The first apt-CT scan on May 31, 2017, showed a complete response, which was confirmed by subsequent apt-CT scan after GC completion on August 10, 2017. Then, the patient was observed by an oncologist. Cardiac function was assessed by echocardiography before GC initiation. Only type 1 diastolic dysfunction of the left ventricle (LV) and a slight degenerative rearrangement of the valves were determined. LV ejection fraction was 63%, no pericardial effusion was present.

On November 10, 2017, pelvic magnetic resonance imaging (MRI) revealed localized pathological LAP close to the left external iliac vessels and in the left groin area, clinical scrotal oedema was present. Due to localized relapse, external beam radiotherapy (ExRT) to the total dose of 40 Grays (Gy) was applied to the area of LAP from November 13, 2017 to December 12, 2017 without any serious acute toxicity.

A restaging apt-CT scan was performed on January 17, 2018, revealing an enlarged aortocaval retroperitoneal lymph node sized 25 mm × 18 mm (a target lesion, Fig. 1) and another enlarged retroperitoneal lymph node sized 39 mm × 13 mm (a non-target lesion). Bellmunt prognostic score (performance status < 0, hemoglobin level less than 10 g/dL, and the presence of liver metastasis) was zero in this patient. Median survival of patients without any negative prognostic factors was 14.2 months in this Bellmunt study [4].

Fig. 1
figure 1

Computed tomography scan before atezolizumab treatment initiation (January 17, 2018): aorto-caval retroperitoneal lymph node—the target lesion (25 mm × 18 mm)—green arrow

From January 17, 2018, this patient who met the inclusion criteria was treated within the clinical trial NCT02928406 (ClinicalTrials.gov Identifier) with atezolizumab (1200 mg intravenously every 3 weeks), a monoclonal antibody against PD-L1. On the pre-planned restaging apt-CT scan on August 8, 2018, complete remission was shown. The enlarged aortocaval retroperitoneal lymph node (a target lesion) decreased in size from 25 mm × 18 mm to 6 mm × 5 mm (Fig. 2); however clinical scrotal oedema persisted. The patient was treated with immunotherapy until September 20, 2018 without any serious (grades 3 and 4) adverse event. Only grade 1 fatigue was recorded during this systemic therapy.

Fig. 2
figure 2

Computed tomography scan at the time of pre-planned restaging (August 9, 2018): Aortocaval retroperitoneal lymph node—the target lesion (6 mm × 5 mm)—green arrow

On October 4, 2018, the patient was admitted to the hospital with clinical deterioration in terms of anxiety and restlessness, weakness, rapid breathing, fainting, dizziness followed by rapid loss of consciousness. On admission, hypotension (blood pressure 60/35 mm Hg), peripheral cyanosis, and distant heartbeat sounds on auscultation were present. Increase in aminotransferases (alanine aminotransferase 6300 IU/L, aspartate aminotransferase 6300 IU/L), creatine of 3.62 mg/dL, hyponatremia, hyperkalemia, hypo-coagulation state, and metabolic acidosis were also recorded. Electrocardiogram (ECG) showed low QRS voltage and sinus tachycardia (120 per minute). Echocardiography could not be utilized because during physical examination, the patient went into pulmonary circulatory arrest, resuscitation was unsuccessful and he died due to cardiorespiratory failure.

In making differential diagnoses, we considered the toxicity of atezolizumab since complete response was present on the most recent CT re-evaluation. On pathological examination, no macroscopic visible neoplasia was found. However, in the pericardial cavity a small amount (less than 50 ml) of sanguinolent effusion was present and the pericardium was covered by irregular grey coatings. The complete pericardium was covered by gray-to-white coatings and in evaluated sections infiltrated by neoplastic cells, histologically corresponding to the metastatic dissemination of invasive urothelial carcinoma (Fig. 3). Microscopic examination (Fig. 4) showed thickening of the pericardium infiltrated by neoplastic proliferation and the formation of irregular nests of epithelioid cells with marked pleomorphism, enlarged hyperchromatic nuclei with a high n/c ratio. No significant inflammatory infiltration was present. Immunohistochemical examination confirmed pericardial infiltration by high grade urothelial carcinoma. The neoplastic cells showed positivity for cytokeratin markers CK7 and CK20 and were negative for p63, vimentin and calretinin. The activated mesothelial cells were weakly positive for CK7, negative for CK20 and p63 and showed regular positivity for vimentin and calretinin. PD-L1 expression was analyzed immunohistochemically using the rabbit monoclonal antibody against PD-L1 (28–8 clone, Merck Cell Marque, Rocklin, California, USA). PD-L1 status was negative (Fig. 5). The paraaortic lymph nodes were not significantly enlarged, with elastic consistence and gray color on cross-section. No metastatic dissemination was found in evaluated in lymph nodes.

Fig. 3
figure 3

The complete pericardium covered by gray-to-white coatings and infiltrated by neoplastic cells, histologically corresponding to the metastatic dissemination of muscle-infiltrating urothelial carcinoma

Fig. 4
figure 4

Histological examination showed pericardial infiltration by high grade urothelial carcinoma (A). The neoplastic cells (black arrows) showed positivity for cytokeratin markers CK7 (B) and CK20 (C) and were negative for p63 (D), vimentin (E) and calretinin (F), when compared to the activated mesothelial cells (white arrows) weakly positive for CK7 (B) and negative for CK20 (C) with regular positivity for vimentin (E) and calretinin (F)

Fig. 5
figure 5

Negative PD-L1 status determined immunohistochemically using the rabbit monoclonal antibody against PD-L1 (28-8 clone, Merck Cell Marque, Rocklin, California, USA)

In this patient, progression-free survival (PFS) following first-line GC treatment was 8.3 months and 8.5 months for second-line immunotherapy. Overall survival (OS) from initiation of systemic therapy until death was 19.1 months.

Discussion and conclusions

The pericardium is the most common site of cardiac metastasis (64–69%) followed by the epicardium (25–34%) and the myocardium (29–32%) [5]. However, the possibility of pericardial metastases in patients with metastatic urothelial carcinoma is not widely recognised. In autopsies of 17 of patients with MUC, the rate of metastasis to the epicardium was approximately 11% [6]. These metastases present an increased risk of cardiac tamponade, malignant arrhythmias and subsequent death. Initial presentation of cardiac metastasis may vary, from asymptomatic to various heart rhythm abnormalities such as atrial fibrillation, atrioventricular blocks or ventricular fibrillation, or signs of cardiac tamponade [5]. Physical examination during anticancer therapies plays an important role, because suspicion of pericardial disease can be raised through findings such as decreased heart sounds, new onset cardiac murmur, development of pericardial friction rub, hypotension with narrow pulse pressure and distended jugular veins [7]. ECG evaluation can be of value but often shows nonspecific ST-T deformities [8]. On echocardiography, pericardial effusions or cardiac masses can sometimes be seen [9]. Treatment is based on urgent pericardiocentesis despite its high recurrence rates (30–60%) [10]. In patients with a longer life expectancy, surgical drainage, based on effusion location and clinical condition, offers the lowest recurrence rate [10].

PD-1 is expressed by activated T lymphocytes. It attaches with its ligands PD-L1 to restrict the activation of T lymphocytes. Monoclonal antibodies targeting PD-1 and PD-L1 can block the immune checkpoint and eliminate the inhibition of T lymphocytes activation. As a result, they enhance immune reactions to fight against malignancies. However, due to its blockade of inhibitory regulation of T-lymphocytes and therefore unregulated activation of immune reaction, it may also affect other organs and injure normal tissues, leading to immune-related adverse effects [11].

In a single arm, phase 2 study [12] with atezolizumab, side effects of any grade were present in 69% of subjects and majority of them were mild to moderate in nature. The most common toxicities of atezolizumab were fatigue, nausea, decreased appetite, and pruritus. However, in 16% of cases, the adverse events (mostly fatigue) were serious. The immune-mediated side effects of any grade were present in 7% of patients, with pneumonitis, increased aspartate aminotransferase and alanine aminotransferase being the most common. 5% of subjects had a serious immune-mediated adverse event. There were no treatment-related deaths reported in this study.

In IMvigor-211 trial [13], in both the expression on ≥ 5% of tumor-infiltrating immune cells (IC2/3) and intention-to-treat (ITT) populations, grade 3 or 4 treatment-related adverse events were less common with atezolizumab than chemotherapy. Among the 467 patients treated with atezolizumab (ITT population), treatment-related adverse events were reported as follow: fatigue (2%), anaemia (2%), neutropenia (< 1%), peripheral neuropathy (< 1%), asthenia (2%), and febrile neutropenia (< 1%). In both trials with atezolizumab the rate of pericarditis and pericardial effusion were not reported. So far there are limited data about the rate of occurrence of pericardial disease in patients treated with ICIs.

In our patient treated with atezolizumab, histopathological examination confirmed the urothelial immunophenotype (CK7 +, CK20 +) of the neoplastic cells within the pericardium. Impaired p63 expression, observed in this case, was found in other studies to be associated with higher biological aggression of high-grade urothelial carcinoma [14,15,16]. When we searched through PubMed using the key words “pericardial infiltration”, “urothelial carcinoma”, “high grade”, and “atezolizumab” no articles were found. Therefore, we believe that this is the first case report of a patient with metastatic urothelial carcinoma who achieved complete CT response with atezolizumab after the failure of cisplatin-based chemotherapy, but who suddenly died due to malignant tumour infiltration of the pericardium as the only site of disease progression.

In this patient, OS calculated from atezolizumab initiation was shorter (8.5 months) than expected survival (14.2 months) considering the Bellmunt prognostic score. However, the Bellmunt study [4] identified pretreatment prognostic factors for OS only in MUC patients who experienced treatment failure with the first-line, platinum-based regimen included in the phase 3 vinflunine trial. In the phase 2 and phase 3 studies [12, 13] with atezolizumab, median OS was 11.4 months and 11.1 months, respectively.

In conclusion, immunotherapy based on check-point inhibition, regardless of PD-L1 expression, is the current standard treatment for patients with advanced urothelial carcinoma after failure of platinum-based combined chemotherapy. In comparison to cytostatic treatment, immunotherapy improves both survival and the percentage of objective responses including complete responses while having fewer adverse effects in general.

Any new treatment carries the risk of new side effects, which may be causally related to the systemic therapy. However, the sudden death of our patient was not related to atezolizumab, but was due to malignant pericardial infiltration confirmed by histopathological examination, despite the complete response to immunotherapy on CT. It is therefore important that the possibility of pericardial metastases should not be discounted, especially where any cardiopulmonary symptoms are evident.

Availability of data and materials

The data and materials are available from the corresponding author.

Abbreviations

apt-CT:

Abdominal, pelvic, and thoracic–computed tomography

ECG:

Electrocardiogram

ExRT:

External radiotherapy

GC:

Gemcitabine plus cisplatin

ITT:

Intention to treat

LAP:

Lymphadenopathy

MUC:

Metastatic urothelial carcinoma

MRI:

Magnetic resonance imaging

OS:

Overall survival

PFS:

Progression-free survival

PD-1:

Programmed cell death protein–1

PD-L1:

Programmed death protein–1 ligand

References

  1. Cowan NC, De Santis M, Bruins HM, Hernández V, Espinós EL, Dunn J, Rouanne M, Neuzillet Y, Veskimäe E, van der Heijden AG, Gakis G, Ribal MJ. Updated 2016 EAU guidelines on muscle-invasive and metastatic bladder cancer. Eur Urol. 2017;71:462–75.

    Article  Google Scholar 

  2. MacGee W. Metastatic and invasive tumours involving the heart in a geriatric population: a necropsy study. Virchows Arch A Pathol Anat Histopathol. 1991;419:183–9.

    Article  CAS  Google Scholar 

  3. Bajorin DF, Dodd PM, Mazumdar M, Fazzari M, McCaffrey JA, Scher HI, Herr H, Higgins G, Boyle MG. Long-term survival in metastatic transitional-cell carcinoma and prognostic factors predicting outcome of therapy. J Clin Oncol. 1999;17:3173–81.

    Article  CAS  Google Scholar 

  4. Bellmunt J, Choueiri TK, Fougeray R, Schutz FAB, Salhi Y, Winquist E, Culine S, von der Maase H, Vaughn DJ, Rosenberg JE. Prognostic factors in patients with advanced transitional cell carcinoma of the urothelial tract experiencing treatment failure with platinum-containing regimens. J Clin Oncol. 2010;28(11):1850–5.

    Article  Google Scholar 

  5. Bussani R, De-Giorgio F, Abbate A, Silvestri F. Cardiac metastases. J Clin Pathol. 2007;60:27–34.

    Article  CAS  Google Scholar 

  6. Klatt EC, Heitz DR. Cardiac metastases. Cancer. 1990;65:1456–9.

    Article  CAS  Google Scholar 

  7. Lewis MA, Hendrickson AW, Moynihan TJ. Oncologic emergencies: pathophysiology, presentation, diagnosis, and treatment. CA Cancer J Clin. 2011;61:287–314.

    PubMed  Google Scholar 

  8. Cates CU, Virmani R, Vaughn WK, Robertson RM. Electrocardiographic markers of cardiac metastasis. Am Heart J. 1986;112:1297–303.

    Article  CAS  Google Scholar 

  9. Auger D, Pressacco J, Marcotte F, Tremblay A, Dore A, Ducharme A. Cardiac masses: an integrative approach using echocardiography and other imaging modalities. Heart. 2011;97:1101–9.

    Article  Google Scholar 

  10. Schusler R, Meyerson SL. Pericardial disease associated with malignancy. Curr Cardiol Rep. 2018;20:92.

    Article  Google Scholar 

  11. Weber JS, Yang JC, Atkins MB, Disis ML. Toxicities of immunotherapy for the practitioner. J Clin Oncol. 2015;33:2092–9.

    Article  CAS  Google Scholar 

  12. Rosenberg JE, Hoffman-Censits J, Powles T, van der Heijden MS, Balar AV, Necchi A, Dawson N, O’Donnell PH, Balmanoukian A, Loriot Y, Srinivas S, Retz MM, Grivas P, Joseph RW, Galsky MD, Fleming MT, Petrylak DP, Perez-Gracia JL, Burris HA, Castellano D, Canil C, Bellmunt J, Bajorin D, Nickles D, Bourgon R, Frampton GM, Cui N, Mariathasan S, Abidoye O, Fine GD, Dreicer R. Atezolizumab in patients with locally advanced and metastatic urothelial carcinoma who have progressed following treatment with platinum-based chemotherapy: a single-arm, multicentre, phase 2 trial. Lancet. 2016;387:1909–20.

    Article  CAS  Google Scholar 

  13. Powles T, Durán I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U, Oudard S, Retz MM, Castellano D, Bamias A, Fléchon A, Gravis G, Hussain S, Takano T, Leng N, Kadel EE 3rd, Banchereau R, Hegde PS, Mariathasan S, Cui N, Shen X, Derleth CL, Green MC, Ravaud A. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet. 2017;391:748–57.

    Article  Google Scholar 

  14. Koga F, Kawakami S, Fujii Y, Saito K, Ohtsuka Y, Iwai A, Ando N, Takizawa T, Kageyama Y, Kihara K. Impaired p63 expression associates with poor prognosis and uroplakin III expression in invasive urothelial carcinoma of the bladder. Clin Cancer Res. 2003;9:5501–7.

    CAS  PubMed  Google Scholar 

  15. Park BJ, Lee SJ, Kim JI, Lee SU, Lee CH, Chang SG, Park JH, Chi SG. Frequent alteration of p63 expression in human primary bladder carcinomas. Cancer Res. 2000;60:3370.

    CAS  PubMed  Google Scholar 

  16. Urist MJ, Di CCJ, Lu ML, Charytonowicz E, Verbel D, Crum CP, Ince TA, McKeon FD, Cordon-Cardo C. Loss of p63 expression is associated with tumor progression in bladder cancer. Am J Pathol. 2002;161:1199–206.

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Our thanks go to Michael K. Hill, an experienced medical writer and Partner, Hill Long Associates (Teddington, UK) for assistance in preparing this manuscript.

Funding

Article processing charge (APC) was funded by OncoReSearch, Rovinka (Slovakia). However, the funding body did not have a role in the writing of this manuscript.

Author information

Authors and Affiliations

Authors

Contributions

PP designed the study, performed the treatment, and wrote and approved the final manuscript. JS performed the literature search, acquisition of data, manuscript review and revision. PJ performed histopathological diagnosis and prepared Figs. 3, 4 and 5. HP performed CT diagnosis and prepared Figs. 1 and 2. VA performed the treatment with PP. MM also revised the manuscript. All authors read and approved the final manuscript.

Author's information

PP: Assistant Professor at the 2nd Department of Oncology, Faculty of Medicine, Comenius University in Bratislava (Slovakia) and the Head of Outpatient Chemotherapy Department at the National Cancer Institute, Bratislava (Slovakia).

Corresponding author

Correspondence to Patrik Palacka.

Ethics declarations

Ethics approval and consent to participate

The patient was treated with atezolizumab within the SAUL study (clinicaltrials.gov: NCT02928406). Informed consent was signed before enrollment.

Consent for publication

Written informed consent for publication of clinical details and clinical images were obtained from the patient prior to his death.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Palacka, P., Janega, P., Polakova, H. et al. Pericardial malignant infiltration as the cause of sudden death of a patient with metastatic urothelial carcinoma treated with atezolizumab. BMC Urol 22, 108 (2022). https://doi.org/10.1186/s12894-022-01064-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12894-022-01064-x

Keywords