Abstract
Lynch syndrome (LS) is an autosomal dominant disorder caused by mutations in mismatch repair (MMR) genes and is also known to be associated with glioblastomas. The efficacy of immunotherapy for LS-associated glioblastomas remains unknown. Herein, we report a rare case of LS-associated glioblastoma, treated with chemotherapy using immune checkpoint inhibitors (ICI). A 41-year-old female patient presented with headaches and sensory disturbances in the right upper limb for 6 weeks. She had been treated for rectal cancer and had a family history of LS. MRI revealed two ring-enhancing lesions in the left precentral gyrus. She underwent subtotal resection, leading to a pathological diagnosis of isocitrate dehydrogenase wild-type glioblastoma. She received daily administration of (temozolomide, 75 mg/m2) and concurrent radiotherapy (60 Gy) postoperatively. However, the tumor recurred 1 year after the initial treatment. A molecular genetic study showed high microsatellite instability (MSI), and she was treated with pembrolizumab therapy. Disease progression occurred despite six cycles of pembrolizumab therapy and radiotherapy at the dose of 40 Gy. She died due to glioblastoma progression 19 months after the initial treatment. The present case demonstrates that some LS-associated glioblastomas may be resistant to ICI despite high MSI, possibly because of intratumor heterogeneity related to MMR deficiency.
Lynch syndrome (LS) is an autosomal dominant disorder caused by mutations in mismatch repair (MMR) genes, including MLH1, MSH2, MSH6, and PMS2, which represent loci on 3p22.2, 2p21, 2p16.3, and 7p 22.1, respectively [12]. LS is also associated with an increased risk of colorectal, endometrial, and several other cancer types [34]. Brain tumors occur in 1%–6% of LS cases, with glioblastomas being the most common type [5]. Recent studies have elucidated the pathogenesis and treatment of MMR-deficient gliomas, including LS-associated gliomas. Immune checkpoint inhibitors (ICIs) have shown promising results in some cases of microsatellite instability (MSI)-high (H) malignancies, but their efficiency varies with the case [6]. Herein, we report a case of LS-associated MSI-H glioblastoma showing resistance to immunotherapy, and review the relevant literature.
A 41-year-old female patient presented with headaches and sensory disturbances in the right upper limb for the past 6 weeks. Her mother had been diagnosed with LS through genetic screening. The patient was treated for rectal cancer at the age of 29 years (Fig. 1A). Initial CT revealed two brain tumors in the precentral gyrus of the left frontal lobe. MRI revealed two ring-enhancing lesions with peritumoral edema at the same site (Fig. 1B, C, E, and F). A malignant brain tumor was suspected, and the patient was referred to our department for surgical treatment. Following admission, she developed right-sided facial palsy and hemiparesis. A second MRI performed at our hospital showed rapid tumor enlargement. The patient underwent subtotal surgical resection of the tumor with awake craniotomy (Fig. 1D and G).
Histopathological analysis showed focal necrosis with pseudo palisades and microvascular proliferation, along with an MIB-1 proliferation index of 70%, leading to a diagnosis of isocitrate dehydrogenase (IDH) wild-type glioblastoma (Fig. 2). Genetic testing by next-generation sequencing showed alternations in PIK3CA R88Q, TP53 R282W, TP53 R158H, NF1 R304, and ATRX T1582NX. MSI was high and the tumor showed an MSH2 variant on germline sequencing analysis.
After surgical resection, the patient was treated with daily administration of temozolomide (TMZ; 75 mg/m2) and concurrent radiotherapy (60 Gy). She was subsequently administered TMZ and bevacizumab (10 mg/kg, biweekly). However, 10 months after the initial surgery, the tumors recurred at the same location. A year after the initial surgery, the patient underwent a second surgery, involving subtotal resection. Two weeks after the second surgery, the patient received two cycles of pembrolizumab (200 mg) therapy. However, the second treatment cycle was followed by disease progression and dissemination. Disease progression occurred despite six cycles of pembrolizumab therapy and radiation therapy (40 Gy) for the cerebellar lesion. The patient died due to glioblastoma progression 19 months after the initial treatment.
Although the association between LS and glioma is well-known, the clinical course and outcomes of LS-associated glioblastomas remain unclear. The lifetime risk of brain tumors in LS patients ranges from 1% to 6% by the age of 70 years [7]. Therkildsen et al. [8] demonstrated that 56% of LS-associated brain tumors are glioblastomas. They also reported the average survival duration of 12.1 months (1–41 months) for LS-associated glioblastomas. A literature review of the LS-associated glioblastoma cases treated with immunotherapy revealed that the survival duration is above 60 months if immunotherapy is successful. In contrast, the survival duration is 15.5–19 months in cases resistant to immunotherapy (Table 1).
LS is caused by heterozygous and homozygous germline mutations in one of the MMR genes [9]. MSH2 (40%–50%) and MLH1 (30%–37%) germline mutations are the most common, while MSH6 and PMS2 mutations are found in only 7%–13% and up to 9% of the cases, respectively [910]. Therkildsen et al. [8] demonstrated a significant increase in the risk of brain tumors with MSH2 gene mutations.
MMR-deficient IDH wild-type glioblastomas, including the present case, are associated with TP53, NF1, EP300, GATA3, NOTCH1, and PIK3CA mutations [1112]. Most LS-associated glioblastomas also have MSI-H [11]. A subset of MSI-H Lynch tumors also present with TP53 mutations, which are otherwise rare in MSI-H cancers [13]. All TP53-mutant tumors present with a non-polypous histology, suggesting that TP53 and CTNNB1 mutations may act as the drivers of nonpolyposis cancer formation in LS [14].
MMR deficiency is often associated with MSI-H and tumor mutational burden (TMB)-H, contributing to poor outcomes [1516]. Some studies have reported that MMR-deficient gliomas could be resistant to TMZ [17]. According to Kim et al. [11], the progression-free survival ranges from 1 month to 42 months for patients with recurrent malignancies. The 2-year survival rate was found to be 29.0% in MMR-deficient glioblastomas. Moreover, Kaplan-Meier survival analysis revealed that patients with MMR-deficient high-grade gliomas had shorter progression-free and overall survival rates compared to those without MMR-deficient high-grade gliomas.
ICIs have shown promising results for some MSI-H malignancies [18]. Increased expression of neoantigens promotes the effect of programmed cell death-1 (PD-1) blockade on MMR-deficient cancers [6]. However, the effect on gliomas is limited due to intratumor heterogeneity related to MMR deficiency [1920]. This heterogeneity is attributed to an acquired MMR gene mutation found in gliomas treated with TMZ [2122]. Previous reports have indicated that MSI-H is not always associated with the effect of anti PD-1 immunotherapy in MMR-deficient glioblastoma [2324]. Meanwhile, TMB is associated with favorable outcomes following anti PD-1 immunotherapy [11242526]. Indraccolo et al. [27] reported that the relationship between MMR/TMB deficiency and MSI may vary with the tumor type.
Friedman et al. [28] demonstrated that EGFR amplification is associated with poor survival rates after ICI treatment in recurrent glioblastoma patients. EGFR can serve as a biomarker for ICI resistance and may be used to distinguish between responders and non-responders.
The efficacy of ICI in the treatment of MMR-deficient glioblastomas has been disappointing, but it remains unknown whether glioblastomas with TMB-H or without EGFR amplification respond to ICI in LS patients [2428]. Further clinical trials of immunotherapy for specific MMR-deficient brain tumors are required.
In conclusion, we presented a case of LS-associated glioblastoma showing ICI resistance despite MSI-H. Our findings indicate that some LS-associated glioblastomas may be resistant to immunotherapy.
Availability of Data and Material
The datasets generated or analyzed during the study are available from the corresponding author on reasonable request.
References
1. Lynch HT, de la Chapelle A. Hereditary colorectal cancer. N Engl J Med. 2003; 348:919–932. PMID: 12621137.
2. Wimmer K, Etzler J. Constitutional mismatch repair-deficiency syndrome: have we so far seen only the tip of an iceberg? Hum Genet. 2008; 124:105–122. PMID: 18709565.
3. Barrow E, Robinson L, Alduaij W, Shenton A, Clancy T, Lalloo F, et al. Cumulative lifetime incidence of extracolonic cancers in Lynch syndrome: a report of 121 families with proven mutations. Clin Genet. 2009; 75:141–149. PMID: 19215248.
4. Watson P, Vasen HFA, Mecklin JP, Bernstein I, Aarnio M, Järvinen HJ, et al. The risk of extra-colonic, extra-endometrial cancer in the Lynch syndrome. Int J Cancer. 2008; 123:444–449. PMID: 18398828.
5. Vasen HF, Stormorken A, Menko FH, Nagengast FM, Kleibeuker JH, Griffioen G, et al. MSH2 mutation carriers are at higher risk of cancer than MLH1 mutation carriers: a study of hereditary nonpolyposis colorectal cancer families. J Clin Oncol. 2001; 19:4074–4080. PMID: 11600610.
6. Le DT, Durham JN, Smith KN, Wang H, Bartlett BR, Aulakh LK, et al. Mismatch repair deficiency predicts response of solid tumors to PD-1 blockade. Science. 2017; 357:409–413. PMID: 28596308.
7. Barrow E, Hill J, Evans DG. Cancer risk in Lynch syndrome. Fam Cancer. 2013; 12:229–240. PMID: 23604856.
8. Therkildsen C, Ladelund S, Rambech E, Persson A, Petersen A, Nilbert M. Glioblastomas, astrocytomas and oligodendrogliomas linked to Lynch syndrome. Eur J Neurol. 2015; 22:717–724. PMID: 25648859.
9. Moreira L, Balaguer F, Lindor N, de la Chapelle A, Hampel H, Aaltonen LA, et al. Identification of Lynch syndrome among patients with colorectal cancer. JAMA. 2012; 308:1555–1565. PMID: 23073952.
10. Bhattacharya P, McHugh TW. Lynch syndrome. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing;2023. Accessed October 29, 2023. at https://www.ncbi.nlm.nih.gov/books/NBK431096/.
11. Kim H, Lim KY, Park JW, Kang J, Won JK, Lee K, et al. Sporadic and Lynch syndrome-associated mismatch repair-deficient brain tumors. Lab Invest. 2022; 102:160–171. PMID: 34848827.
12. Kawaguchi K, Otani R, Kikuchi M, Kushihara Y, Funata N, Yamada R, et al. Genetic characteristics of mismatch repair-deficient glioblastoma. NMC Case Rep J. 2021; 8:565–571. PMID: 35079518.
13. Kloth M, Ruesseler V, Engel C, Koenig K, Peifer M, Mariotti E, et al. Activating ERBB2/HER2 mutations indicate susceptibility to pan-HER inhibitors in Lynch and Lynch-like colorectal cancer. Gut. 2016; 65:1296–1305. PMID: 26001389.
14. Ahadova A, von Knebel Doeberitz M, Bläker H, Kloor M. CTNNB1-mutant colorectal carcinomas with immediate invasive growth: a model of interval cancers in Lynch syndrome. Fam Cancer. 2016; 15:579–586. PMID: 26960970.
15. Wang L, Ge J, Lan Y, Shi Y, Luo Y, Tan Y, et al. Tumor mutational burden is associated with poor outcomes in diffuse glioma. BMC Cancer. 2020; 20:213. PMID: 32164609.
16. Daniel P, Sabri S, Chaddad A, Meehan B, Jean-Claude B, Rak J, et al. Temozolomide induced hypermutation in glioma: evolutionary mechanisms and therapeutic opportunities. Front Oncol. 2019; 9:41. PMID: 30778375.
17. Goenka A, Tiek D, Song X, Huang T, Hu B, Cheng SY. The many facets of therapy resistance and tumor recurrence in glioblastoma. Cells. 2021; 10:484. PMID: 33668200.
18. Cohen R, Colle R, Pudlarz T, Heran M, Duval A, Svrcek M, et al. Immune checkpoint inhibition in metastatic colorectal cancer harboring microsatellite instability or mismatch repair deficiency. Cancers (Basel). 2021; 13:1149. PMID: 33800202.
19. McCord M, Steffens A, Javier R, Kam KL, McCortney K, Horbinski C. The efficacy of DNA mismatch repair enzyme immunohistochemistry as a screening test for hypermutated gliomas. Acta Neuropathol Commun. 2020; 8:15. PMID: 32051040.
20. Mukasa A. Genome medicine for brain tumors: current status and future perspectives. Neurol Med Chir (Tokyo). 2020; 60:531–542. PMID: 33071276.
21. Cahill DP, Levine KK, Betensky RA, Codd PJ, Romany CA, Reavie LB, et al. Loss of the mismatch repair protein MSH6 in human glioblastomas is associated with tumor progression during temozolomide treatment. Clin Cancer Res. 2007; 13:2038–2045. PMID: 17404084.
22. Touat M, Li YY, Boynton AN, Spurr LF, Iorgulescu JB, Bohrson CL, et al. Mechanisms and therapeutic implications of hypermutation in gliomas. Nature. 2020; 580:517–523. PMID: 32322066.
23. Yao ZG, Hua F, Yin ZH, Xue YJ, Hou YH, Nie YC, et al. Characteristics of glioblastomas and immune microenvironment in a Chinese family with Lynch syndrome and concurrent porokeratosis. Front Oncol. 2023; 13:1194232. PMID: 37529690.
24. Anghileri E, Di Ianni N, Paterra R, Langella T, Zhao J, Eoli M, et al. High tumor mutational burden and T-cell activation are associated with long-term response to anti-PD1 therapy in Lynch syndrome recurrent glioblastoma patient. Cancer Immunol Immunother. 2021; 70:831–842. PMID: 33140187.
25. Cho YA, Kim D, Lee B, Shim JH, Suh YL. Incidence, clinicopathologic, and genetic characteristics of mismatch repair gene-mutated glioblastomas. J Neurooncol. 2021; 153:43–53. PMID: 33864561.
26. Hodges TR, Ott M, Xiu J, Gatalica Z, Swensen J, Zhou S, et al. Mutational burden, immune checkpoint expression, and mismatch repair in glioma: implications for immune checkpoint immunotherapy. Neuro Oncol. 2017; 19:1047–1057. PMID: 28371827.
27. Indraccolo S, Lombardi G, Fassan M, Pasqualini L, Giunco S, Marcato R, et al. Genetic, epigenetic, and immunologic profiling of MMR-deficient relapsed glioblastoma. Clin Cancer Res. 2019; 25:1828–1837. PMID: 30514778.
28. Friedman JS, Jun T, Rashidipour O, Huang KL, Ellis E, Kadaba P, et al. Using EGFR amplification to stratify recurrent glioblastoma treated with immune checkpoint inhibitors. Cancer Immunol Immunother. 2023; 72:1893–1901. PMID: 36707424.
Table 1
Study | Age/Sex | Location | Other associated tumors | Duration between the diagnosis of high-grade glioma and cancer | Surgery | Treatment | MMR mutations | MSI | TMB | Ki-67 | Survival after the diagnosis of high-grade glioma |
---|---|---|---|---|---|---|---|---|---|---|---|
Anghileri et al. [24] | 33/F | Left frontal lobe | Adenoma with high-grade dysplasia in the transverse colon | 4 years | GTR | RT/TMZ → nivolumab | MSH2, MSH6 | N/A | High | N/A | Alive at 84 months |
Sherman and Vitaz [29] | 57/F | Left occipital lobe | Colorectal cancer | 10 years | GTR | RT 60 Gy/TMZ → RT 45 Gy/nivolumab 240 mg | N/A | MSI-H | N/A | N/A | Alive at 5 years |
Kawaguchi et al. [12] | 79/F | Left parietal lobe | Colorectal cancer | 12 years | GTR | RT 34 Gy/TMZ → pembrolizumab | MSH2 | MSI-H | 101.6/Mb | 47% | Died after 15.5 months |
Present case | 41/F | Left frontal lobe | Colorectal cancer | 12 years | STR | RT 60 Gy/TMZ → pembrolizumab 200 mg | MSH2 | MSI-H | High | 70% | Died after 19 months |