Germline mutations of mismatch repair (MMR) genes such as MLH1, MSH2, MSH6, and PMS2 are hallmarks of Lynch syndrome (LS), also known as hereditary nonpolyposis colorectal cancer (HNPCC).1 Due to alteration of mechanisms involved in gene editing, mutations that occur during DNA synthesis are unable to be repaired, leading to a higher rate of genomic instability. These areas of genomic instability are more likely to occur where DNA strands contain repetitive nucleotide sequences, known as microsatellites.2 The pathogenesis of LS is thought to be precipitated by high levels of microsatellite instability (MSI-H) in genes controlling cell growth, proliferation, and apoptosis. As a result of the genomic instability associated with MMR mutations, there are multiple extracolonic malignancies in addition to colorectal cancer (CRC) that are associated with LS, such as endometrial, ovarian, gastric, pancreaticobiliary, urinary tract, small bowel, brain, and sebaceous neoplasms.3 Patients with LS have an approximate 35% to 55% lifetime risk of developing CRC, although the specific germline mutation involved may affect the type of extracolonic malignancy that emerges.4,5
Immunotherapy with PD-1 receptor blockade in patients with MSI-H is effective therapy for HNPCC as well as extracolonic malignancies.6 Immune checkpoint blockade with PD-1 inhibitors is now first-line therapy for advanced MSI-H or mismatch repair–deficient (dMMR) CRC.7 Given the mechanism of action, the side effect profile of immune checkpoint inhibitors (ICIs) is associated with autoimmune reactions to visceral organs as well as squamous epithelium, and the development of such side effects is unpredictable. Immune-related adverse events (irAEs) range from mild (grade 1) to fatal (grade 5).8 The most frequent grade 3 (severe) and grade 4 (life-threatening) irAEs are gastrointestinal and dermatologic in nature.9 However, irAEs causing neurotoxicity, cardiotoxicity, or pulmonary toxicity are often the most fatal and represent a unique challenge for patients who would otherwise benefit from immunotherapy. There is a paucity of information regarding the further use of PD-1 inhibitors in patients with MSI-H who develop life-threatening systemic toxicity reactions. This case highlights a potential therapeutic strategy for patients receiving pembrolizumab with subsequent myocarditis, a well-documented adverse reaction that occurs in <1% of patients but carries a 46% chance of mortality.10
Case Description
We present a case of an 85-year-old male with Muir-Torre syndrome, a variant of Lynch syndrome (LS) associated with visceral malignancies as well as secondary sebaceous neoplasms. The patient presented with a germline mutation in the MSH2 gene. Other pertinent medical history was significant for coronary artery disease with percutaneous coronary intervention to the left anterior descending artery, resulting in mildly reduced left ventricular ejection fraction (LVEF) at 50%, as well as typical atrial flutter with ablation to the cavotricuspid isthmus. His oncologic history was significant for metachronous CRC diagnosed at age 52 years, initially treated with 5-FU–based chemotherapy, but with recurrence approximately 20 years later requiring partial colectomy and anastomosis. He also developed multiple urothelial cancers starting at age 65 years, as well as several skin cancers, including sebaceous carcinoma. As expected, his family history is also consistent with LS, including other family members with both colorectal and noncolorectal cancers.
In March 2020, a screening upper endoscopy was performed, which revealed a 3-cm polyp in the gastric fundus with pathology showing moderately differentiated, intestinal-type gastric adenocarcinoma with lymphovascular and submucosal invasion. Based on the NCCN Clinical Practice Guidelines in Oncology (NCCN Guidelines) for Gastric Cancer, the patient underwent diagnostic laparoscopy with peritoneal washings to rule out radiologically occult disease.11 A biopsy from the peritoneum was positive for metastatic adenocarcinoma, consistent with the patient’s known gastric primary. Genetic analysis confirmed MLH1 and MSH2 mutations via immunohistochemical staining.
Following the diagnosis of invasive gastric adenocarcinoma in the setting of high microsatellite instability, the patient was started on the PD-1 receptor–targeted immunotherapy pembrolizumab (200-mg infusions every 3 weeks). Shortly after receiving the second infusion of pembrolizumab, the patient experienced significant chest pain and progressive shortness of breath, requiring multiple hospital admissions. During his initial presentation, his troponin levels were noted to be elevated at 1,414 ng/L and N-terminal pro-B-type natriuretic peptide (NT-proBNP) level was 1,076 pg/mL. His 12-lead EKG showed sinus rhythm with premature ventricular contractions and nonspecific ST-T abnormalities. He underwent a transthoracic echocardiogram, which showed LVEF of 47% with regional wall motion abnormalities.
A cardiac MRI showed delayed enhancement in a nonvascular distribution predominately involving the basilar lateral wall, with small areas of delayed enhancement scattered throughout the left ventricle. Due to findings of the cardiac MRI, invasive testing with endomyocardial biopsy was deferred. Furthermore, the patient’s last coronary angiogram had been completed 1 year prior, with placement of a drug-eluting stent to the mid left anterior descending coronary artery. Overall, this was felt to be consistent with grade 4 myocarditis related to immunotherapy, and less likely acute coronary syndrome (Figures 1 and 2). The patient also had evidence of grade 1 myositis and possible signs of grade 1 hepatitis, as noted by elevations in creatine kinase (607 ng/L) and ALT/AST (259/385 U/L) levels. Inflammatory markers, including C-reactive protein and erythrocyte sedimentation rate, at the time were not assessed. It should be noted that the elevation in liver enzymes was thought by rheumatology to be related to muscle injury rather than true liver inflammation or grade 1 hepatitis.
Pembrolizumab was discontinued, and the patient was started on immunosuppressive therapy with intravenous methylprednisolone at 1,000 mg for 3 days followed by a 60 mg of oral prednisone daily, tapered by 10 mg every 7 days. After his initial hospital discharge, he experienced recurrent chest pain on 3 separate occasions, each time presenting with elevated troponin and NT-proBNP levels, thought to be flares of ICI-related myocarditis during his steroid taper. Prior elevation in creatine kinase and liver enzymes had returned to baseline levels and there was no concern for concurrent muscle or hepatic injury. After careful consideration of the available treatment options for refractory myocarditis, the patient was treated with the tumor necrosis factor–inhibitor infliximab. A single dose of 5 mg/kg was used with concurrent pulsed methylprednisolone at 1,000 mg for 3 days and transitioned to a slow prednisone taper. He had no further episodes of chest pain or biochemical evidence of recurrent myocarditis. Troponin and NT-proBNP levels eventually stabilized to a new baseline of approximately 100 ng/L and 400 pg/mL, respectively, in addition to creatine kinase and liver enzymes remaining within normal limits. He underwent frequent monitoring by the cardiology department, with scheduled echocardiograms every 3 months and serial NT-proBNP and troponin measurements. Additionally, he was started on beta-blockade with metoprolol succinate at 50 mg daily to protect against tachyarrhythmias.
The patient had 2 repeat upper endoscopies within 7 months of the initial complications from pembrolizumab. Although he had only completed 2 doses, biopsies from the first esophagogastroduodenoscopy (EGD) showed gastric oxyntic-type mucosa and superficial fragments of neoplastic intestinal-type epithelium with no invasive carcinoma identified. However, the biopsy results from the second EGD (7 months after stopping pembrolizumab) showed recurrent focal invasive well-differentiated adenocarcinoma arising in a background of high-grade dysplasia, with morphology composed of foveolar-type epithelium with scattered goblet cells. CT imaging of the abdomen and pelvis from that time showed a 2.2-cm lesion near the proximal stomach, representing the patient's known local recurrent tumor, and no radiographic evidence of peritoneal or mesenteric disease.
Due to the patient’s disease recurrence, a risk/benefit discussion was conducted regarding his prognosis with and without ICI therapy. Systemic chemotherapy was considered; however, due to the numerous prior cycles of chemotherapy for previous malignancies that the patient had undergone, as well as their associated side effects and his advanced age, he opted not to pursue this option. Large, powered studies suggest an overall survival of roughly 3 to 6 months in unselected untreated metastatic gastric cancer.12 Following careful consideration of potential significant risks, the patient’s prior pathologic and radiographic response to anti–PD-1 therapy, and prior data suggesting a meaningful benefit in MSI-H gastric cancer treated with ICI, the decision was made to proceed with an alternative ICI therapy. Due to the significant benefit previously achieved with pembrolizumab after only 2 doses, the anti–PD-1 biosimilar nivolumab was started at a 50% dose reduction (480 mg decreased to 240 mg), with infusions administered at an interval of every 4 weeks. Dose reduction of nivolumab was chosen to mitigate the risk of recurrent myocarditis or other irAEs. Although nivolumab has not been extensively studied in MSI-H gastric cancer, results from the NCI-MATCH trial showed an objective response rate (ORR) of 36% and promising results toward progression-free and overall survival for patients with relapsed or refractory dMMR noncolorectal cancers.13
The patient was closely observed by medical oncology and cardio-oncology for potential severe adverse reactions. At the time of initiation with dose-reduced nivolumab, troponin and NT-proBNP levels were 150 ng/L and 582 pg/mL, respectively, and continued to decrease to the baseline levels mentioned previously. He tolerated nivolumab without significant side effects or irAEs. The patient received 13 cycles of dose-reduced nivolumab over 1 year. The patient remains disease-free almost 1 year after completing therapy, with no radiographic evidence of residual disease. Additionally, surveillance EGD showed no visible masses, and biopsies revealed only mild acute and chronic inflammation, negative for intestinal metaplasia or dysplasia. Images from the EGDs over the course of his diagnosis and treatment are shown in Figure 3. A timeline of the course of events is shown in Figure 4.
Discussion
This case represents an approach that includes a shared decision to rechallenge patients with ICI following immune-related life-threatening complications in highly selective cases in which considerable benefit may be expected. Multiple clinical practice guidelines report on management of ICI-related adverse events.14–17 The specific management of irAEs in the acute setting ultimately depends on the type and extent of organ involvement and should be carefully considered on a case-by-case basis. However, there is no specific guidance regarding secondary considerations for rechallenge in patients with MSI-H malignancies who develop life-threatening irAEs from PD-1 receptor inhibitors.
The general recommendations from the Society for Immunotherapy of Cancer (SITC) for patients who encounter grade 3 or 4 irAEs are to risk adjust the possibility of re-treatment based on the expected benefit from immunotherapy against the risk of potential toxicity.14,18,19 However, the panel makes very specific recommendations for patients who develop myocarditis due to the significant overlap with myositis and potentially fatal neurotoxicity. Myocarditis can develop as a spectrum of multisystem irAEs, with concurrent irAEs present in 9.8% of patients.20 An individual who develops any grade of muscle, cardiac, or neurologic involvement from immunotherapy should be tested for concurrent toxicity with a shared set of diagnostics.21 Due to the high mortality rates associated with myocarditis, the panel states that permanent discontinuation of immunotherapy should be strongly considered.14 Of note, SITC does not comment on the utility of dose reduction as a potential alternative strategy for those who develop irAEs, irrespective of the underlying severity.
The Multinational Association of Supportive Care in Cancer (MASCC) is another international society that provides general recommendations for patients who experience irAEs from ICIs. The consensus from MASCC regarding myocarditis is permanent discontinuation of any ICI due to the high risk of recurrence.15 ASCO has similar recommendations for the diagnosis and management of toxicity reactions, with guidelines regarding grade 4 toxicities (except for endocrinopathies controlled by hormone replacement) recommending permanent discontinuation of ICIs.16
The importance of a thorough risk assessment for secondary challenge is especially critical for cancers harboring a dismal prognosis. Cancers with MSI-H are recalcitrant to adjuvant chemotherapy and may in fact worsen outcomes in earlier stages of disease (stages II and III) as shown in post hoc analysis of the MAGIC and CLASSIC randomized controlled trials.22,23 A meta-analysis from 4 randomized controlled trials showed improved OS in resected MSI-H resectable gastric cancer, but no observable increase in disease-free survival (DFS) for those treated with additional adjuvant chemotherapy.24 MSI-H status also confers prognostic benefits compared with MSI-L/MSS resectable gastric cancer (5-year DFS, 71.8% vs 52.3%; 5-year OS, 77.9% vs 52.3%), as shown in the pooled meta-analysis from the MAGIC, CLASSIC, ARTIST, and ITACA-S trials.25 The utility of traditional chemotherapy in patients with advanced or metastatic disease should be explored and MSI status should not preclude their use, particularly in those who have contraindications to or side effects from immunotherapy. Although our patient declined systemic chemotherapy for reasons stated previously, 5-FU–based or platinum-based chemotherapy should also be considered due to the improved outcomes compared with untreated patients with metastatic disease.12
Evidence for continued immunotherapy for MSI-H–detected cancer comes from results the KEYNOTE-158 and KEYNOTE-177 trials.7,26 Specifically, the KEYNOTE-177 trial showed significant improvements for MSI-H CRC toward median progression-free survival (PFS) (16.5 vs 8.2 months) with pembrolizumab compared with 5-FU–based chemotherapy.27 Further evidence for the use of immunotherapy in advanced MSI-H or dMMR tumors has been demonstrated from a subanalysis of the CheckMate 649 and ATTRACTION-4 trials.28,29 Patients with advanced, resistant, or recurrent MMR-deficient gastric or gastroesophageal cancer who received nivolumab + oxaliplatin-based chemotherapy experienced a median survival of 44.8 months compared with 8.8 months with chemotherapy alone.28 Results of these studies suggest a survival benefit for immunotherapy with or without traditional chemotherapy and the risks and benefits should considered carefully on an individual basis.
In 2019, a JAMA Oncology study of case safety reports from the WHO database VigiBase analyzed the rate of irAE recurrence after ICI rechallenge.17 Results of the study showed 0 of 3 patients with prior ICI-related myocarditis experienced recurrence after secondary rechallenge. Moreover, a patient with recurrent endometrial cancer who developed myocarditis from the PD-1 inhibitor sintilimab was successfully rechallenged with an alternative agent, zimberelimab.30 However, based on our review of the literature, there have been no published reports of patients who developed grade 4 myocarditis from ICI therapy who were subsequently rechallenged with dose reduction of same class agents.
Conclusions
We present a case of a patient with metastatic gastric adenocarcinoma and Muir-Torre/LS secondary to a confirmed mutation in the MSH2 gene. Following the early administration of pembrolizumab, the patient developed several irAEs, including grade 4 myocarditis as defined by ASCO as symptomatic life-threatening disease with radiographic and biologic evidence of myocarditis, requiring treatment with corticosteroids and infliximab.31 He was later safely rechallenged with nivolumab at 50% dose reduction and achieved a complete response after treatment for 1 year. This case highlights a therapeutic strategy for cautious rechallenge of select patients who develop severe irAE from ICIs, through careful consideration and a shared decision-making process that highlights the risk/benefit ratio.
References
- 1.↑
Win AK, Jenkins MA, Dowty JG, et al. Prevalence and penetrance of major genes and polygenes for colorectal cancer. Cancer Epidemiol Biomarkers Prev 2017;26:404–412.
- 2.↑
Evrard C, Tachon G, Randrian V, et al. Microsatellite instability: diagnosis, heterogeneity, discordance, and clinical impact in colorectal cancer. Cancers (Basel) 2019;11:1567.
- 3.↑
Moreira L, Balaguer F, Lindor N, et al. Identification of Lynch syndrome among patients with colorectal cancer. JAMA 2012;308:1555–1565.
- 5.↑
Bonadona V, Bonaïti B, Olschwang S, et al. Cancer risks associated with germline mutations in MLH1, MSH2, and MSH6 genes in Lynch syndrome. JAMA 2011;305:2304–2310.
- 6.↑
Le DT, Uram JN, Wang H, et al. PD-1 blockade in tumors with mismatch-repair deficiency. N Engl J Med 2015;372:2509–2520.
- 7.↑
Shiu KK, Andre T, Kim TW, et al. KEYNOTE-177: phase III randomized study of pembrolizumab versus chemotherapy for microsatellite instability-high advanced colorectal cancer. J Clin Oncol 2021;39(Suppl):Abstract 6.
- 8.↑
Choi J, Lee SY. Clinical characteristics and treatment of immune-related adverse events of immune checkpoint inhibitors. Immune Netw 2020;20:e9.
- 9.↑
Das S, Johnson DB. Immune-related adverse events and anti-tumor efficacy of immune checkpoint inhibitors. J Immunother Cancer 2019;7:306.
- 10.↑
Moslehi JJ, Salem JE, Sosman JA, et al. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 2018;391:933.
- 11.↑
Ajani JA, D’Amico TA, Bentrem DJ, et al. NCCN Clinical Practice Guidelines in Oncology: Gastric Cancer. Version 1.2023. Accessed March 20, 2023. To view the most recent version, visit https://www.nccn.org
- 12.↑
Yang D, Hendifar A, Lenz C, et al. Survival of metastatic gastric cancer: significance of age, sex and race/ethnicity. J Gastrointest Oncol 2011;2:77–84.
- 13.↑
Azad NS, Gray RJ, Overman MJ, et al. Nivolumab is effective in mismatch repair-deficient noncolorectal cancers: results from arm Z1D-A subprotocol of the NCI-MATCH (EAY131) study. J Clin Oncol 2020;38:214–222.
- 14.↑
Brahmer JR, Abu-Sbeih H, Ascierto PA, et al. Society for Immunotherapy of Cancer (SITC) clinical practice guideline on immune checkpoint inhibitor-related adverse events. J Immunother Cancer 2021;9:e002435.
- 15.↑
Suarez-Almazor ME, Pundole X, Abdel-Wahab N, et al. Multinational Association of Supportive Care in Cancer (MASCC) 2020 clinical practice recommendations for the management of immune-mediated cardiovascular, rheumatic, and renal toxicities from checkpoint inhibitors. Support Care Cancer 2020;28:6159–6173.
- 16.↑
Schneider BJ, Naidoo J, Santomasso BD, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO guideline update. J Clin Oncol 2021;39:4073–4126.
- 17.↑
Dolladille C, Ederhy S, Sassier M, et al. Immune checkpoint inhibitor rechallenge after immune-related adverse events in patients with cancer. JAMA Oncol 2020;6:865–871.
- 18.↑
Santini FC, Rizvi H, Plodkowski AJ, et al. Safety and efficacy of re-treating with immunotherapy after immune-related adverse events in patients with NSCLC. Cancer Immunol Res 2018;6:1093–1099.
- 19.↑
Simonaggio A, Michot JM, Voisin AL, et al. Evaluation of readministration of immune checkpoint inhibitors after immune-related adverse events in patients with cancer. JAMA Oncol 2019;5:1310–1317.
- 20.↑
Shankar B, Zhang J, Naqash AR, et al. Multisystem immune-related adverse events associated with immune checkpoint inhibitors for treatment of non-small cell lung cancer. JAMA Oncol 2020;6:1952–1956.
- 21.↑
Johnson DB, Manouchehri A, Haugh AM, et al. Neurologic toxicity associated with immune checkpoint inhibitors: a pharmacovigilance study. J Immunother Cancer 2019;7:134.
- 22.↑
Choi YY, Kim H, Shin SJ, et al. Microsatellite instability and programmed cell death-ligand 1 expression in stage II/III gastric cancer: post hoc analysis of the CLASSIC randomized controlled study. Ann Surg 2019;270:309–316.
- 23.↑
Smyth EC, Wotherspoon A, Peckitt C, et al. Mismatch repair deficiency, microsatellite instability, and survival: an exploratory analysis of the Medical Research Council Adjuvant Gastric Infusional Chemotherapy (MAGIC) trial. JAMA Oncol 2017;3:1197–1203.
- 24.↑
Joshi SS, Badgwell BD. Current treatment and recent progress in gastric cancer. CA Cancer J Clin 2021;71:264–279.
- 25.↑
Pietrantonio F, Raimondi A, Choi YY, et al. MSI-GC-01: individual patient data (IPD) meta-analysis of microsatellite instability (MSI) and gastric cancer (GC) from four randomized clinical trials (RCTs). J Clin Oncol 2019;37(Suppl):Abstract 66.
- 26.↑
Marabelle A, Le DT, Ascierto PA, et al. Efficacy of pembrolizumab in patients with noncolorectal high microsatellite instability/mismatch repair-deficient cancer: results from the phase II KEYNOTE-158 study. J Clin Oncol 2020;38:1–10.
- 27.↑
Diaz LA Jr, Shiu KK, Kim TW, et al. Pembrolizumab versus chemotherapy for microsatellite instability-high or mismatch repair-deficient metastatic colorectal cancer (KEYNOTE-177): final analysis of a randomised, open-label, phase 3 study. Lancet Oncol 2022;23:659–670.
- 28.↑
Shitara K, Ajani JA, Moehler M, et al. Nivolumab plus chemotherapy or ipilimumab in gastro-oesophageal cancer. Nature 2022;603:942–948.
- 29.↑
Kang YK, Chen LT, Ryu MH, et al. Nivolumab plus chemotherapy versus placebo plus chemotherapy in patients with HER2-negative, untreated, unresectable advanced or recurrent gastric or gastro-oesophageal junction cancer (ATTRACTION-4): a randomised, multicentre, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2022;23:234–247.
- 30.↑
Chen Y, Huang A, Yang Q, et al. Case report: a successful re-challenge report of GLS-010 (zimberelimab), a novel fully humanized mAb to PD-1, in a case of recurrent endometrial cancer. Front Immunol 2022;13:987345.
- 31.↑
Palaskas N, Lopez-Mattei J, Durand JB, et al. Immune checkpoint inhibitor myocarditis: pathophysiological characteristics, diagnosis, and treatment. J Am Heart Assoc 2020;9:e013757.