Your new experience awaits. Try the new design now and help us make it even better

CASE REPORT article

Front. Immunol., 23 October 2025

Sec. Cancer Immunity and Immunotherapy

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1685944

This article is part of the Research TopicNew Strategies and Clinical Translation Progress in Immunotherapy for Liver CancerView all 5 articles

Effective control in MSI-high unresectable intrahepatic cholangiocarcinoma treated with gemcitabine/cisplatin/durvalumab and proton beam therapy: a case report

Takuto Nosaka*Takuto Nosaka1*Hironori NaitoHironori Naito1Yosuke MurataYosuke Murata1Yu AkazawaYu Akazawa1Tomoko TanakaTomoko Tanaka1Kazuto TakahashiKazuto Takahashi1Tatsushi NaitoTatsushi Naito1Masahiro OhtaniMasahiro Ohtani1Yoshiaki ImamuraYoshiaki Imamura2Yasunari Nakamoto*Yasunari Nakamoto1*
  • 1Second Department of Internal Medicine, Faculty of Medical Sciences, University of Fukui, Fukui, Japan
  • 2Division of Diagnostic Pathology/Surgical Pathology, University of Fukui Hospital, Fukui, Japan

Systemic chemotherapy is the standard treatment for unresectable intrahepatic cholangiocarcinoma (iCCA); however, its efficacy remains limited. We report a clinically valuable case of a 57-year-old woman with advanced unresectable iCCA characterized by high microsatellite instability (MSI-high) and major vascular invasion. The patient presented with purpura and was diagnosed with MSI-high iCCA with intravascular tumor extension reaching the right atrium and regional lymph node metastases. Combination therapy with gemcitabine, cisplatin, and durvalumab (GCD therapy) was initiated, resulting in a 56% reduction in tumor size after eight cycles. To enhance local tumor control, proton beamtherapy (3.3 Gy × 22 fractions) was added, which was completed without adverse events. The patient subsequently received durvalumab maintenance therapy, followed by pembrolizumab. Although there was minimal growth of lymph node and pulmonary metastases, no regrowth of the intrahepatic primary tumor was observed for 18 months after PBT. This case illustrates the potential clinical value of combining GCD therapy with proton beam therapy for MSI-high, unresectable iCCA with major vascular invasion. The combination achieved systemic disease control and durable local control without significant toxicity.

1 Introduction

Cholangiocarcinoma is anatomically classified into intrahepatic (iCCA) and extrahepatic subtypes (perihilar and distal). iCCA is the second most common subtype of primary liver cancer, accounting for approximately 15% of all primary liver cancers worldwide, and its incidence is increasing (1). Because the disease often progresses asymptomatically, a large proportion of patients are unresectable at diagnosis, and recurrence is common; consequently, long-term outcomes remain poor (2). For unresectable iCCA, based on the TOPAZ-1 trial, gemcitabine, cisplatin, and durvalumab (GCD) has been established as the standard first-line regimen (3); however, systemic chemotherapy alone may be insufficient to prevent local progression and liver-related events in a subset of patients (4). Accordingly, the optimal integration of systemic and local therapies remains a key clinical challenge. At the molecular level, a subset of iCCA harbors high microsatellite instability (MSI-high) (5), and such tumors have demonstrated increased sensitivity to immune checkpoint inhibitors (ICIs) (6).

Among high-precision radiotherapy modalities, proton beam therapy (PBT) provides superior dose conformality, enabling high-dose irradiation while sparing organs at risk (OARs) such as the hepatic parenchyma, biliary tract, and gastrointestinal structures; long-term survival has been reported in unresectable iCCA (79). Furthermore, because radiotherapy can modulate the tumor immune microenvironment, combining it with ICIs is expected to achieve both systemic disease control and durable local control (10). However, much of the evidence derives from nonrandomized studies and registry cohorts, and there remains scope for prospective optimization of combination strategies (3, 11).

Here, we report a case of unresectable MSI-high iCCA with major vascular invasion, in which successful local control was achieved by combining GCD therapy with subsequent PBT.

2 Case description

A 57-year-old woman noticed purpura on her left buttock and limbs in Month Y–1, Year X, and visited a nearby clinic. She had no significant past medical history. Laboratory tests revealed anemia and decreased fibrinogen levels, raising suspicion of fibrinolytic-dominant disseminated intravascular coagulation (DIC) (Table 1). The calculated DIC score according to the International Society on Thrombosis and Haemostasis (ISTH) criteria was 5, confirming the diagnosis of DIC. Gadoxetic acid-enhanced magnetic resonance imaging (EOB-MRI) and transabdominal ultrasound revealed a 77-mm mass in the right hepatic lobe (Figure 1A), with tumor invasion extending from the inferior vena cava into the right atrium (Figure 1B). She was referred and admitted to our department for further evaluation. Fluorodeoxyglucose positron emission tomography/CT (FDG PET/CT) demonstrated intense uptake in the hepatic tumor, with a maximum standardized uptake value (SUVmax) of 13.3, as well as in the hilar lymph nodes (Figure 1C). A percutaneous biopsy of the liver tumor demonstrated adenocarcinoma with CK7-positive and Glypican-3-negative immunostaining (Figure 1D), consistent with intrahepatic cholangiocarcinoma. Given the patient’s advanced malignancy and laboratory findings, a tumor-related DIC was suspected. Comprehensive genomic profiling using the FoundationOne CDx panel revealed high microsatellite instability (MSI-high). Combination chemotherapy with gemcitabine, cisplatin, and durvalumab (GCD therapy) was initiated in Month Y, Year X, resulting in a 56% reduction in tumor size after eight cycles, according to the Response Evaluation Criteria in Solid Tumors (RECIST 1.1) (Figure 2). To achieve further local control of the intrahepatic lesion, proton beam therapy was initiated at 5.8 months following treatment initiation. The patient was immobilized individually in the supine position, and respiratory motion of the tumor was assessed using respiratory-gated four-dimensional computed tomography (4D-CT). Treatment planning ensured dose coverage of the planning target volume (PTV; V95%) while adhering to dose constraints for OARs. The prescribed regimen consisted of 3.3 Gy (RBE) per fraction, once daily, five days per week, for a total of 22 fractions (72.6 Gy [RBE]). Irradiation was delivered under respiratory gating, with image-guided position verification performed before each session. The patient was subsequently transitioned to maintenance therapy with durvalumab. At 12 months after treatment initiation, new pulmonary metastases were detected and subsequently showed progression, prompting a switch to pembrolizumab. Following ICI-based chemotherapy combined with PBT, the ISTH score improved to 0. No treatment-related toxicities were observed (Common Terminology Criteria for Adverse Events [CTCAE], version 5.0). At 18 months, the intrahepatic tumor remained stable without evidence of progression, and local control was maintained. At the latest follow-up, 26 months after treatment initiation, the patient remains alive.

Table 1
www.frontiersin.org

Table 1. Laboratory examinations.

Figure 1
A set of medical images showing diagnostic imaging and histological findings: A) MRI scans display a liver lesion indicated by arrows. B) An ultrasound image illustrates the inferior vena cava (IVC) and highlighted areas. C) PET scans show intense uptake at specified regions, marked with arrows and arrowheads. D) Histological sections using different stains: H&E stain shows a pink-stained tissue with a highlighted section; CK7 stain shows brown-stained cellular areas; Glypican-3 stain reveals a blue-stained tissue.

Figure 1. Imaging findings and liver tumor biopsy of unresectable intrahepatic cholangiocarcinoma (iCCA). (A) Gadoxetic acid-enhanced magnetic resonance imaging (EOB-MRI) images. Left: arterial phase; Right: hepatobiliary phase. The intrahepatic tumor is indicated by white arrows. (B) Transabdominal ultrasound image. Tumor invasion into the inferior vena cava (IVC) is observed (yellow arrowhead), with partial extension into the right atrium (white asterisk). (C) Fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) findings. Left: High uptake in the intrahepatic tumor (arrow). Right: High uptake in the metastatic hilar lymph node (arrowhead). (D) Histopathological findings of the liver tumor obtained by percutaneous needle biopsy. From left to right: Hematoxylin and eosin (H&E) staining (low magnification), H&E staining (high magnification), immunohistochemistry for cytokeratin 7 (CK7), and Glypican-3. Bar, 100 µm.

Figure 2
Line graph and CT scan images depict a hepatic tumor's response to treatment. The graph shows tumor size reduction over 20 months with treatments including Gemcitabine, Cisplatin, Durvalumab, PBT, and Pembrolizumab. CT scans at 0.0, 4.4, 5.8, and 18.2 months visually represent tumor shrinking. Treatment timelines are marked with arrows above the graph.

Figure 2. Clinical course of the patient. Treatment was initiated in Month Y, Year X (Day 0). Contrast-enhanced computed tomography (CT) images show the intrahepatic lesion (white arrows, white dot lines), intravascular invasion extending into the right atrium (yellow arrows), and the dose distribution of proton beam therapy (PBT).

3 Discussion

We experienced a case of unresectable iCCA that demonstrated significant tumor shrinkage and systemic disease control following GCD therapy, with additional effective local control achieved through PBT. Because iCCA is an aggressive malignancy, few patients are eligible for surgical resection at the time of diagnosis; thus, systemic chemotherapy and radiotherapy are commonly employed (2). In the TOPAZ-1 trial (12, 13), the addition of an ICI to gemcitabine and cisplatin improved overall survival, establishing GCD therapy as the new standard first-line treatment (3). In this case, comprehensive genomic profiling of the tumor biopsy confirmed MSI-high status. The prevalence of MSI-high is reported to be only 0–2.1% in biliary tract cancers (14) and 4.7% in iCCA (5). MSI-high tumors are known to be highly sensitive to ICIs (6), which may have contributed to the favorable response to ICI-based therapy observed in this case.

Although chemotherapy remains the standard first-line treatment for unresectable iCCA, its efficacy is limited, underscoring the urgent need for combination strategies with effective local therapies (4). In patients with iCCA and extrahepatic metastases, radiotherapy to the primary liver tumor has been reported to significantly delay the progression of local disease, the onset of tumor-related liver failure, and death (15). Particle beam therapy offers excellent dose concentration and has the potential to achieve high local control rates (11). In unresectable iCCA, PBT has demonstrated 1-year local control rates ranging from 91.2% to 100%, along with favorable overall survival outcomes (11, 16). Several studies have demonstrated the efficacy of combining chemotherapy and radiotherapy in biliary tract cancers (17, 18). Additionally, a case report has shown a favorable therapeutic response to the combination of a PD-1 inhibitor and radiotherapy (19). In this case, GCD therapy was selected as the initial treatment, resulting in shrinkage of the primary tumor and a tendency toward reduction of lymph node metastases, although complete response was not achieved. To enhance local control, PBT was administered, which led to further improvement in local control without any adverse events. To our knowledge, this is the first reported case of unresectable iCCA treated with PBT following GCD therapy, highlighting its clinical significance.

In resectable iCCA, surgical resection is generally the first-line option (3); however, this patient was unresectable owing to major vascular invasion with tumor thrombus extending from the inferior vena cava to the right atrium and concomitant metastases. Although radiofrequency ablation (RFA) can yield favorable outcomes in small, localized tumors (20), its indications are limited for a 77-mm, vessel-adjacent lesion with tumor thrombus because the heat-sink effect and procedural complications increase the risk of incomplete ablation. By contrast, PBT offers superior dose conformality, allowing delivery of adequate tumoricidal doses while sparing the normal liver, biliary tract, and gastrointestinal organs (11). In this case, consolidation PBT after GCD achieved durable local control of the primary lesion without treatment-related adverse events. Accordingly, for advanced cases ineligible for resection or RFA, a sequential strategy of “systemic therapy first, followed by high-precision radiotherapy for local consolidation” may represent a practical option. As this is a single case, modality superiority cannot be concluded, and treatment selection should be individualized.

In this case, after treatment with durvalumab (an anti–PD-L1 antibody), pulmonary metastases were detected, and therapy was switched to pembrolizumab (an anti–PD-1 antibody). Recently, cases have been reported in which switching from an anti–PD-L1 antibody to an anti–PD-1 antibody after the development of resistance to the former has resulted in either tumor response or disease stabilization. In a case series of non–small cell lung cancer (NSCLC) by Kitagawa et al., partial responses or stable disease were observed in some patients after rechallenge with an anti–PD-1 antibody following anti–PD-L1 antibody therapy, suggesting that this approach may be a viable treatment option in selected cases (21).

The patient remains alive 26 months after treatment initiation, with no regrowth of the intrahepatic primary tumor up to 18 months after PBT. By contrast, pulmonary and nodal lesions exhibited mild progression, prompting a switch from anti–PD-L1 to anti–PD-1 therapy. Going forward, we will undertake extended follow-up with imaging every 3–4 months, together with clinical and laboratory assessments, and longitudinal evaluation using validated quality-of-life instruments, to comprehensively assess the long-term effectiveness of this treatment plan, in which GCD is administered first, followed by PBT.

Although no treatment-related adverse events were observed in this case, GCD therapy generally carries risks of myelosuppression, nephrotoxicity, and immune-related adverse events (irAEs) (13), whereas PBT may cause radiation-induced liver injury and gastrointestinal/biliary toxicities (7). These risks can be mitigated through baseline and periodic monitoring, adequate hydration with guideline-concordant antiemetic prophylaxis, dose reductions or delays when indicated, early recognition of irAEs with CTCAE-based management, weekly liver-function assessment, and prophylactic acid suppression when high gastrointestinal doses are anticipated (8, 22). Accordingly, a sequential strategy in which GCD therapy is administered first, followed by consolidative PBT, may help balance toxicity reduction with durable local control.

Within the GCD regimen, gemcitabine and cisplatin (GEM/CDDP) have been reported to induce immunogenic cell death (ICD) through tumor cell death and antigen release (23). Meanwhile, chemotherapy and radiation are known to increase tumor PD-L1 expression, and the addition of durvalumab provides a rational strategy to block this therapy-induced immune-suppressive loop (24). Radiotherapy, particularly PBT, can activate antitumor immunity through dendritic cell–mediated cross-priming, forming the basis for both local control and potential abscopal effects (25). Furthermore, PBT concentrates the radiation dose within the tumor while minimizing low-dose exposure to surrounding normal tissues, thereby reducing radiation-induced lymphopenia and preserving systemic immune function (26). These characteristics make PBT highly compatible with immune checkpoint inhibitors. In the present case, ICD induction by GEM/CDDP, enhanced antigen presentation and cross-priming by PBT, and PD-L1 pathway inhibition by durvalumab may have acted synergistically, and together with the MSI-high immune-responsive phenotype, contributed to durable local control and improvement of DIC.

Diverse mechanisms contributing to tumor progression and therapeutic resistance have been reported (27, 28). The assessment of anticancer treatment efficacy has progressed through the use of patient-derived xenograft models in preclinical studies (29). Nevertheless, clinical validation requires real-world evidence derived from larger patient cohorts. As noted in the limitations, this study represents a single case with a limited level of evidence, and therefore caution should be exercised when interpreting and generalizing the findings. This single-case report limits statistical inference and generalizability. To validate the efficacy and universality of the proposed therapy and strengthen the reliability of the conclusions, future studies should secure larger sample sizes to enable robust statistical analyses. When feasible, multicenter prospective registries or comparative trials are recommended.

4 Conclusion

This is a clinically valuable case of unresectable iCCA treated with GCD therapy and PBT, resulting in tumor shrinkage, systemic disease control, and durable local control. These findings support the potential of ICI-based chemotherapy combined with PBT as an effective strategy for managing MSI-high iCCA with major vascular invasion.

Data availability statement

The original contributions presented in the study are included in the article/supplementary material. Further inquiries can be directed to the corresponding authors.

Ethics statement

Ethical approval was not required for the studies involving humans because Ethical approval was not required for the study involving humans in accordance with the local legislation and institutional requirements. The studies were conducted in accordance with the local legislation and institutional requirements. The participants provided their written informed consent to participate in this study. Written informed consent was obtained from the individual(s) for the publication of any potentially identifiable images or data included in this article.

Author contributions

TNo: Visualization, Writing – original draft, Funding acquisition, Writing – review & editing. HN: Writing – original draft, Writing – review & editing. YM: Writing – review & editing, Writing – original draft. YA: Writing – review & editing, Writing – original draft. TT: Writing – review & editing, Writing – original draft. KT: Writing – review & editing, Writing – original draft. TNa: Writing – review & editing, Writing – original draft. MO: Writing – review & editing, Writing – original draft. YI: Writing – review & editing, Writing – original draft. YN: Writing – review & editing, Conceptualization, Funding acquisition, Writing – original draft, Supervision.

Funding

The author(s) declare financial support was received for the research and/or publication of this article. This research was partially supported by Japan Agency for Medical Research and Development (AMED) under Grant Numbers JP25fk0310529 and JP25fk0210174 and Japan Society for the Promotion of Science (JSPS) KAKENHI Grant-in-Aid for Scientific Research Number JP25K11170.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declare that no Generative AI was used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I, et al. Global cancer statistics 2022: globocan estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. (2024) 74:229–63. doi: 10.3322/caac.21834

PubMed Abstract | Crossref Full Text | Google Scholar

2. Ilyas SI, Khan SA, Hallemeier CL, Kelley RK, and Gores GJ. Cholangiocarcinoma - evolving concepts and therapeutic strategies. Nat Rev Clin Oncol. (2018) 15:95–111. doi: 10.1038/nrclinonc.2017.157

PubMed Abstract | Crossref Full Text | Google Scholar

3. Vogel A, Bridgewater J, Edeline J, Kelley RK, Klümpen HJ, Malka D, et al. Biliary tract cancer: esmo clinical practice guideline for diagnosis, treatment and follow-up. Ann Oncol. (2023) 34:127–40. doi: 10.1016/j.annonc.2022.10.506

PubMed Abstract | Crossref Full Text | Google Scholar

4. Kubo S, Shinkawa H, Asaoka Y, Ioka T, Igaki H, Izumi N, et al. Liver cancer study group of Japan clinical practice guidelines for intrahepatic cholangiocarcinoma. Liver Cancer. (2022) 11:290–314. doi: 10.1159/000522403

PubMed Abstract | Crossref Full Text | Google Scholar

5. Isa T, Tomita S, Nakachi A, Miyazato H, Shimoji H, Kusano T, et al. Analysis of microsatellite instability, K-ras gene mutation and P53 protein overexpression in intrahepatic cholangiocarcinoma. Hepatogastroenterology. (2002) 49:604–8.

PubMed Abstract | Google Scholar

6. Marabelle A, Le DT, Ascierto PA, Di Giacomo AM, De Jesus-Acosta A, Delord JP, 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. doi: 10.1200/jco.19.02105

PubMed Abstract | Crossref Full Text | Google Scholar

7. Hong TS, Wo JY, Yeap BY, Ben-Josef E, McDonnell EI, Blaszkowsky LS, et al. Multi-institutional phase ii study of high-dose hypofractionated proton beam therapy in patients with localized, unresectable hepatocellular carcinoma and intrahepatic cholangiocarcinoma. J Clin Oncol. (2016) 34:460–8. doi: 10.1200/jco.2015.64.2710

PubMed Abstract | Crossref Full Text | Google Scholar

8. Makita C, Nakamura T, Takada A, Takayama K, Suzuki M, Ishikawa Y, et al. Clinical outcomes and toxicity of proton beam therapy for advanced cholangiocarcinoma. Radiat Oncol. (2014) 9:26. doi: 10.1186/1748-717x-9-26

PubMed Abstract | Crossref Full Text | Google Scholar

9. Ohkawa A, Mizumoto M, Ishikawa H, Abei M, Fukuda K, Hashimoto T, et al. Proton beam therapy for unresectable intrahepatic cholangiocarcinoma. J Gastroenterol Hepatol. (2015) 30:957–63. doi: 10.1111/jgh.12843

PubMed Abstract | Crossref Full Text | Google Scholar

10. Lynch C, Pitroda SP, and Weichselbaum RR. Radiotherapy, immunity, and immune checkpoint inhibitors. Lancet Oncol. (2024) 25:e352–e62. doi: 10.1016/s1470-2045(24)00075-5

PubMed Abstract | Crossref Full Text | Google Scholar

11. Mizumoto M, Terashima K, Makishima H, Suzuki M, Ogino T, Waki T, et al. Proton beam therapy for intrahepatic cholangiocarcinoma: A multicenter prospective registry study in Japan. Liver Cancer. (2024) 13:161–8. doi: 10.1159/000531376

PubMed Abstract | Crossref Full Text | Google Scholar

12. Oh DY, He AR, Bouattour M, Okusaka T, Qin S, Chen LT, et al. Durvalumab or placebo plus gemcitabine and cisplatin in participants with advanced biliary tract cancer (Topaz-1): updated overall survival from a randomised phase 3 study. Lancet Gastroenterol Hepatol. (2024) 9:694–704. doi: 10.1016/s2468-1253(24)00095-5

PubMed Abstract | Crossref Full Text | Google Scholar

13. Oh DY, Ruth He A, Qin S, Chen LT, Okusaka T, Vogel A, et al. Durvalumab plus gemcitabine and cisplatin in advanced biliary tract cancer. NEJM Evid. (2022) 1:EVIDoa2200015. doi: 10.1056/EVIDoa2200015

PubMed Abstract | Crossref Full Text | Google Scholar

14. Naganuma A, Sakuda T, Murakami T, Aihara K, Watanuki Y, Suzuki Y, et al. Microsatellite instability-high intrahepatic cholangiocarcinoma with portal vein tumor thrombosis successfully treated with pembrolizumab. Intern Med. (2020) 59:2261–7. doi: 10.2169/internalmedicine.4588-20

PubMed Abstract | Crossref Full Text | Google Scholar

15. De B, Upadhyay R, Liao K, Kumala T, Shi C, Dodoo G, et al. Definitive liver radiotherapy for intrahepatic cholangiocarcinoma with extrahepatic metastases. Liver Cancer. (2023) 12:198–208. doi: 10.1159/000530134

PubMed Abstract | Crossref Full Text | Google Scholar

16. Shimizu S, Okumura T, Oshiro Y, Fukumitsu N, Fukuda K, Ishige K, et al. Clinical outcomes of previously untreated patients with unresectable intrahepatic cholangiocarcinoma following proton beam therapy. Radiat Oncol. (2019) 14:241. doi: 10.1186/s13014-019-1451-5

PubMed Abstract | Crossref Full Text | Google Scholar

17. Kim YI, Park JW, Kim BH, Woo SM, Kim TH, Koh YH, et al. Outcomes of concurrent chemoradiotherapy versus chemotherapy alone for advanced-stage unresectable intrahepatic cholangiocarcinoma. Radiat Oncol. (2013) 8:292. doi: 10.1186/1748-717x-8-292

PubMed Abstract | Crossref Full Text | Google Scholar

18. Smart AC, Goyal L, Horick N, Petkovska N, Zhu AX, Ferrone CR, et al. Hypofractionated radiation therapy for unresectable/locally recurrent intrahepatic cholangiocarcinoma. Ann Surg Oncol. (2020) 27:1122–9. doi: 10.1245/s10434-019-08142-9

PubMed Abstract | Crossref Full Text | Google Scholar

19. Wang N, Wang L, Huang A, Han J, Cao T, Mei X, et al. Case report: A novel mixture of dose-fractioned radiation and immunotherapy for treatment of cholangiocarcinoma. Front Immunol. (2023) 14:1273962. doi: 10.3389/fimmu.2023.1273962

PubMed Abstract | Crossref Full Text | Google Scholar

20. Liver EAftSot. Easl-ilca clinical practice guidelines on the management of intrahepatic cholangiocarcinoma. J Hepatol. (2023) 79:181–208. doi: 10.1016/j.jhep.2023.03.010

PubMed Abstract | Crossref Full Text | Google Scholar

21. Kitagawa S, Hakozaki T, Kitadai R, and Hosomi Y. Switching administration of anti-pd-1 and anti-pd-L1 antibodies as immune checkpoint inhibitor rechallenge in individuals with advanced non-small cell lung cancer: case series and literature review. Thorac Cancer. (2020) 11:1927–33. doi: 10.1111/1759-7714.13483

PubMed Abstract | Crossref Full Text | Google Scholar

22. Schneider BJ, Naidoo J, Santomasso BD, Lacchetti C, Adkins S, Anadkat M, et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: asco guideline update. J Clin Oncol. (2021) 39:4073–126. doi: 10.1200/jco.21.01440

PubMed Abstract | Crossref Full Text | Google Scholar

23. Galluzzi L, Buqué A, Kepp O, Zitvogel L, and Kroemer G. Immunogenic cell death in cancer and infectious disease. Nat Rev Immunol. (2017) 17:97–111. doi: 10.1038/nri.2016.107

PubMed Abstract | Crossref Full Text | Google Scholar

24. Peng J, Hamanishi J, Matsumura N, Abiko K, Murat K, Baba T, et al. Chemotherapy induces programmed cell death-ligand 1 overexpression via the nuclear factor-Kb to foster an immunosuppressive tumor microenvironment in ovarian cancer. Cancer Res. (2015) 75:5034–45. doi: 10.1158/0008-5472.Can-14-3098

PubMed Abstract | Crossref Full Text | Google Scholar

25. Demaria S and Formenti SC. Radiation as an immunological adjuvant: current evidence on dose and fractionation. Front Oncol. (2012) 2:153. doi: 10.3389/fonc.2012.00153

PubMed Abstract | Crossref Full Text | Google Scholar

26. Kim N, Myoung Noh J, Lee W, Park B, Park H, Young Park J, et al. Proton beam therapy reduces the risk of severe radiation-induced lymphopenia during chemoradiotherapy for locally advanced non-small cell lung cancer: A comparative analysis of proton versus photon therapy. Radiother Oncol. (2021) 156:166–73. doi: 10.1016/j.radonc.2020.12.019

PubMed Abstract | Crossref Full Text | Google Scholar

27. Huang M, Gong G, Deng Y, Long X, Long W, Liu Q, et al. Crosstalk between cancer cells and the nervous system. Med Adv. (2023) 1:173–89. doi: 10.1002/med4.27

Crossref Full Text | Google Scholar

28. Pan W, Miao Q, Yin W, Li X, Ye W, Zhang D, et al. The role and clinical applications of exosomes in cancer drug resistance. Cancer Drug Resistance. (2024) 7:43. doi: 10.20517/cdr.2024.97

PubMed Abstract | Crossref Full Text | Google Scholar

29. Li J, Gu A, Tang N, Zengin G, Li M-Y, and Liu Y. Patient-derived xenograft models in pan-cancer: from bench to clinic. Interdiscip Med. (2025) 3:e20250016. doi: 10.1002/inmd.20250016

Crossref Full Text | Google Scholar

Keywords: intrahepatic cholangiocarcinoma, immune checkpoint inhibitor, proton beam therapy, vascular invasion, microsatellite instability

Citation: Nosaka T, Naito H, Murata Y, Akazawa Y, Tanaka T, Takahashi K, Naito T, Ohtani M, Imamura Y and Nakamoto Y (2025) Effective control in MSI-high unresectable intrahepatic cholangiocarcinoma treated with gemcitabine/cisplatin/durvalumab and proton beam therapy: a case report. Front. Immunol. 16:1685944. doi: 10.3389/fimmu.2025.1685944

Received: 14 August 2025; Accepted: 14 October 2025;
Published: 23 October 2025.

Edited by:

Shui Liu, The Second Hospital of Jilin University Department of Hepatobiliary and Pancreatic Surgery, China

Reviewed by:

Meng-Yao Li, Shanghai Jiao Tong University, China
Wenya Tian, IQVIA Laboratories, United States

Copyright © 2025 Nosaka, Naito, Murata, Akazawa, Tanaka, Takahashi, Naito, Ohtani, Imamura and Nakamoto. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Takuto Nosaka, bm9zYXRAdS1mdWt1aS5hYy5qcA==; Yasunari Nakamoto, bmFrYW1vdG8tbWVkMkBtZWQudS1mdWt1aS5hYy5qcA==

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.