Journal List > Ann Hepatobiliary Pancreat Surg > v.27(2) > 1516082792

Ghio and Vijay: Intrahepatic cholangiocarcinoma: Evolving role of neoadjuvant and targeted therapy

Abstract

Intrahepatic cholangiocarcinoma is an aggressive, often fatal, malignancy that arises from the bile ducts. As it often presents with metastatic disease, surgery has limited utility. However, in some cases, neoadjuvant chemotherapy has provided the necessary reduction in tumor burden to allow for adequate resection. Consequently, new advances in neoadjuvant chemoradiation and targeted therapy are of interest with numerous case reports and small series published routinely; it is challenging to present a large case series or study given the overall rare frequency with which this malignancy is seen. Herein, we aim to summarize the newest advances in both neoadjuvant chemotherapy and targeted immunotherapy.

INTRODUCTION

Cholangiocarcinoma (CCA) is an aggressive malignancy that arises from the bile duct cholangiocytes and it is classified into intrahepatic CCA (iCCA) and extrahepatic CCA (eCCA). iCCA arises from the peripheral bile ducts within the liver parenchyma, proximal to the secondary biliary radicals. iCCA represents approximately 10% to 20% of all cholangiocarcinoma cases and has a 5-year survival rate of 8% [1].

MAIN TEXT

In the early stages of iCCA, surgery has historically been thought to be the only chance for cure. However, given its oftentimes late detection and metastatic presentation, over 65% of iCCA patients present with unresectable disease or a disease stage that is not suitable for resection [2]. Consequently, the prognosis for iCCA is very poor and disease free and overall survival in patients who have undergone surgery are still dismal with a reported 5-year overall survival rate of 10% to 35% [3-5]. Historic outcomes of liver transplant in patients with iCCA are discouraging too with a reported 5-year survival rate < 25% [6,7]. The high incidence of early recurrence, even among patients with localized disease who undergo margin-negative resection, have resulted in a sustained interest towards advancement in adjuvant systemic therapy. The recently concluded Japanese JCOG1202 Trial further asserts the effectiveness of adjuvant capecitabine following resection of any biliary tract carcinoma (BTC), including iCCA [8]. The use of adjuvant gemcitabine and cisplatin following R0 or R1 resection of any BTC including iCCA is currently being investigated (ACTICCA-1 trial) [7].
The role of preoperative transarterial chemoembolization (TACE) and transarterial radioembolization with yttrium-90 (TARE) have also been investigated in patients with iCCA. A large retrospective study that used TACE on 127 patients with advanced iCCA reported partial response in 19 (15.0%) patients, stable disease in 101 (79.5%), and progressive disease (PD) in 7 patients (5.5%), 3 months after therapy, with no complete responses. Only 4% of the patients were downsized and successfully underwent resection. Studies on TARE have reported similar low conversion rates (between 4% and 11%) [9-11]. However, concomitant use of TACE/TARE with systemic therapy has shown promising results in recent studies [12,13].
Though neoadjuvant chemotherapy has proven efficacy in other resectable cancers, its indication in iCCA is limited to locally advanced/unresectable tumors. It is no surprise that most literature describing the use of neoadjuvant therapy (NT) in iCCA has stemmed from retrospective studies that have included patients with locally advanced and unresectable tumors receiving NT. Studies that have looked at gemcitabine-based NT have reported the conversion of locally advanced/unresectable tumors to resectable in approximately 22% to 53% of patients [14-16]. A systematic review that included 18 studies with 1,880 patients reported significantly longer survival in patients who underwent resection following downstaging with NT than those who did not (29 vs. 12 months) [17]. There is a growing body of literature examining the role of liver transplantation in iCCA following NT. Lunsford et al. [18] reported on 6 patients with iCCA who received either gemcitabine-cisplatin or gemcitabine-capecitabine-based therapy and subsequent liver transplantation. There was a relatively high recurrence with 3 of the liver transplantation patients developing recurrence, but overall survival was higher than usual with 100% (all 6 patients) surviving to 1 year and 83.3% (5 out of 6 patients) alive at 5 years [18]. The ongoing trials investigating the role of NT in resectable candidates will pave the way for a standard of care in iCCA in the future (Table 1).
Despite being the backbone of the current systemic therapy, platinum-based chemotherapy is often limited by its narrow therapeutic index and harsh cytotoxic side effects. Targeted molecular therapy is a growing area of interest in nearly all oncological fields, and cholangiocarcinoma is no exception. Next-generation sequencing has aided the identification of specific genetic mutations driving cholangiogenesis. Inhibiting critical molecular pathways or mutant proteins with targeted therapy can arrest tumor progression and facilitate tumor regression. A better understanding of intratumor heterogeneity has further paved the way for immunotherapeutic strategies in the management of iCCA. Adoptive cell therapy and immune checkpoint inhibitors (ICI) ascertain the role of immunotherapy to enhance natural, anti-tumor immune responses enabling the generation of anti-tumor memory and long-lived tumor destruction.
ICI targeting programmed death 1 (PD-1), programmed death-ligand 1 (PD-L1), and cytotoxic T-lymphocyte antigen-4 (CTLA-4) seem to play a pivotal role in countering the tumor-tolerant microenvironment in iCCA [19]. Immunotherapies targeting PD-1 and its associated ligand are increasingly gaining interest. Lack of mismatch repair proteins or microsatellite instability occurs in approximately 10% of iCCA patients and hence these patients are good targets for immunotherapy [20]. Based on encouraging results from PD-L1 blockade, the United States Food and Drug Administration (FDA) approved pembrolizumab for microsatellite instability high/mismatch repair deficient tumors in 2017 [20]. In a retrospective review of patients with advanced biliary tract cancers, in a propensity score matching analysis, Gou et al. [21] found that anti-PD-1 therapy, in addition to chemotherapy, resulted in prolonged progression-free survival compared to patients who received chemotherapy alone. A case report by Zhang et al. [22] demonstrated that neoadjuvant PD-1 blockade and tyrosine kinase inhibitor therapy in a 38-year-old female with iCCA resulted in an R0 resection with prolonged survival. The DEBATE Trial (Neoadjuvant Gemcitabine Plus Cisplatin With or Without Durvalumab in Resectable Biliary Tract Cancer) is currently recruiting patients and the trial results will hopefully guide future PD-L1 treatment of cholangiocarcinoma [23]. Another study examining the role of anti-PD-L1, titled “PD1 Antibody (Toripalimab), GEMOX, and Lenvatinib Neoadjuvant Treatment for Resectable Intrahepatic Cholangiocarcinoma with High-Risk Recurrence Factors” is also recruiting patients. There is early, emerging, data from Moffitt Cancer Center, AstraZeneca, and Michigan Cancer Center trials demonstrating that PD-1 antibodies, Nivolumab and Durvalumab, have improved progression-free survival and overall survival (NCT02829918, NCT03875235, NCT03101566). This data, while just preliminary, has shown that PD-1 therapy is more effective in monotherapy than when combined with gemcitabine-cisplatin. There is also data from the National Cancer Institute study examining the combination of PD-1 antibody (Durvalumab) with CTLA-4 antibody (Tremelimumab) and TACE/RFA versus Cryoablation, but at this time that study has too few patients (cryoablation arm only has 1 patient currently reported in it) to draw conclusions from.
Isocitrate dehydrogenase (IDH) mutations are found in approximately 13% to 14% of iCCA patients [24,25]. In a multi-center, randomized, double-blinded study involving 124 patients with chemotherapy-refractory disease, Ivosidenib (IDH-1 inhibitor) provided overall improved progression-free survival compared to those receiving the placebo [26]. IDH2 inhibitors such as Enasidenib are currently being tested in clinical trials.
The rate of fibroblast growth factor receptor (FGFR) 2 mutations in iCCA is approximately 13% to 15% [27]. Abou-Alfa et al. [28] in 2020, described the use of Pemigatinib (a selective, oral FGFR1-3 inhibitor and the first FDA-approved targeted agent for the second-line treatment of iCCA) in 107 patients with confirmed FGFR2 mutations. Complete response was reported in 3 patients (2.8%), a partial response in 35 (32.7%), and stable disease in 50 (46.7%) [28]. Overall survival was 21.1 months and progress-free survival was approximately 6.9 months [28]. A Phase II trial examining the role of BGJ398 (FGFR kinase inhibitor) demonstrated an overall response rate of 14.8%, progression-free survival of 5.8 months, and disease control rate of 75.4% [29].
While BRAF and MEK mutations are believed to be relatively rare mutations in biliary tract malignancies, a combination of BRAF inhibition (using Dabrafenib) and MEK inhibition (using Trametinib/Selumetinib) seems to have a synergistic impact with improved results in several phase II trials including the ROAR Basket Trial [30-33]. Mutations in Human Epidermal Growth Factor Receptors have been noted in 8% of iCCA patients [34,35]. The combination of erlotinib (EGFR tyrosine kinase inhibitor) and bevacizumab (vascular endothelial growth factor inhibitor) studied in a phase II trial on 49 patients with advanced BTC described partial response in 6 patients (12.2%) and stable disease in 25 patients (51.0%) [34]. Early investigations into inhibiting EGFR with Pantumumab for unresectable tumors have not shown significant improvement in disease free survival [36]. Mutations in BRCA1 and BRCA2 carry an increased risk of developing cholangiocarcinoma given DNA repair pathway mutations [34]. In a retrospective cohort study by Golan et al. [37], which included 7 patients with iCCA, treatment with poly-ADP ribose polymerase inhibitors resulted in a favorable response, with one patient’s overall survival censored at 64.76 months and progression-free survival of 42.6 months.
There is also an increased emphasis by the scientific community on examining Wnt/[Symbol - b]-catenin signaling, Hedgehog signaling, and JAK/STAT pathways which are all involved in cell growth, cell death, and proliferation [38]. The increased activation in these pathways is thought to be secondary to increased IL-6 secretion by activated Kupffer cells, and other cells activated by cancer pathogenesis, including tumor-associated macrophages and fibroblasts [38]. Increased IL-6 can also result in STAT3 overexpression and loss of negative feedback of JAKs [39]. Early cellular studies investigating these pathways are underway, with the hope of inhibiting the Wnt pathway in the progression to uncontrolled metastatic growth of cholangiocarcinoma cells [40].

CONCLUSIONS

Though the use of immune/targeted therapy in iCCA is at present under investigation, the surfeit of ongoing clinical trials is exciting; the results of which are eagerly anticipated. While most of these targeted therapies are currently being investigated in patients with advanced disease and early phase trials, these therapies hold great potential for use in the neoadjuvant and perioperative settings. Limited success with immune checkpoint blockade mono therapy has led to more trials that are studying combination strategies for enhanced efficacy, which include dual immune checkpoint blockade (ICB) or constituting of ICB along with chemotherapy and/or targeted therapy (Table 2). Acknowledging the tumor microenvironment and genetic heterogeneity displayed by iCCA is essential to furthering the therapeutic potential of ICI and targeted therapy. The next frontier in the treatment of iCCA awaits the development of predictive biomarkers that can both guide iCCA treatment decisions and predict response to immune/targeted therapy. A better understanding of the synergy associated with combinational therapeutic approaches and the advent of precise biomarkers may well represent the future direction of NT in iCCA.

Notes

CONFLICT OF INTEREST

No potential conflict of interest relevant to this article was reported.

AUTHOR CONTRIBUTIONS

Conceptualization: All authors. Data curation: All authors. Methodology: All authors. Visualization: All authors. Writing - original draft: All authors. Writing - review & editing: All authors.

REFERENCES

1. Rizzo A, Brandi G. 2021; Neoadjuvant therapy for cholangiocarcinoma: a comprehensive literature review. Cancer Treat Res Commun. 27:100354. DOI: 10.1016/j.ctarc.2021.100354. PMID: 33756174.
crossref
2. Valle JW, Lamarca A, Goyal L, Barriuso J, Zhu AX. 2017; New horizons for precision medicine in biliary tract cancers. Cancer Discov. 7:943–962. DOI: 10.1158/2159-8290.CD-17-0245. PMID: 28818953. PMCID: PMC5586506.
crossref
3. Raufi AG, Manji GA, Chabot JA, Bates SE. 2019; Neoadjuvant treatment for pancreatic cancer. Semin Oncol. 46:19–27. DOI: 10.1053/j.seminoncol.2018.12.002. PMID: 30630600.
crossref
4. Jung JH, Lee HJ, Lee HS, Jo JH, Cho IR, Chung MJ, et al. 2017; Benefit of neoadjuvant concurrent chemoradiotherapy for locally advanced perihilar cholangiocarcinoma. World J Gastroenterol. 23:3301–3308. DOI: 10.3748/wjg.v23.i18.3301. PMID: 28566890. PMCID: PMC5434436.
crossref
5. Sumiyoshi T, Shima Y, Okabayashi T, Negoro Y, Shimada Y, Iwata J, et al. 2018; Chemoradiotherapy for initially unresectable locally advanced cholangiocarcinoma. World J Surg. 42:2910–2918. DOI: 10.1007/s00268-018-4558-1. PMID: 29511872.
crossref
6. Kato A, Shimizu H, Ohtsuka M, Yoshidome H, Yoshitomi H, Furukawa K, et al. 2013; Surgical resection after downsizing chemotherapy for initially unresectable locally advanced biliary tract cancer: a retrospective single-center study. Ann Surg Oncol. 20:318–324. DOI: 10.1245/s10434-012-2312-8. PMID: 23149849.
crossref
7. Stein A, Arnold D, Bridgewater J, Goldstein D, Jensen LH, Klümpen HJ, et al. 2015; Adjuvant chemotherapy with gemcitabine and cisplatin compared to observation after curative intent resection of cholangiocarcinoma and muscle invasive gallbladder carcinoma (ACTICCA-1 trial) - a randomized, multidisciplinary, multinational phase III trial. BMC Cancer. 15:564. DOI: 10.1186/s12885-015-1498-0. PMID: 26228433. PMCID: PMC4520064.
crossref
8. Nakachi K, Konishi M, Ikeda M, Mizusawa J, Eba J, Okusaka T, et al. 2018; A randomized Phase III trial of adjuvant S-1 therapy vs. observation alone in resected biliary tract cancer: Japan Clinical Oncology Group Study (JCOG1202, ASCOT). Jpn J Clin Oncol. 48:392–395. DOI: 10.1093/jjco/hyy004. PMID: 29462482.
crossref
9. Ibrahim SM, Mulcahy MF, Lewandowski RJ, Sato KT, Ryu RK, Masterson EJ, et al. 2008; Treatment of unresectable cholangiocarcinoma using yttrium-90 microspheres: results from a pilot study. Cancer. 113:2119–2128. DOI: 10.1002/cncr.23818. PMID: 18759346.
crossref
10. Mouli S, Memon K, Baker T, Benson AB 3rd, Mulcahy MF, Gupta R, et al. 2013; Yttrium-90 radioembolization for intrahepatic cholangiocarcinoma: safety, response, and survival analysis. J Vasc Interv Radiol. 24:1227–1234. DOI: 10.1016/j.jvir.2013.02.031. PMID: 23602420. PMCID: PMC3800023.
crossref
11. Saxena A, Bester L, Chua TC, Chu FC, Morris DL. 2010; Yttrium-90 radiotherapy for unresectable intrahepatic cholangiocarcinoma: a preliminary assessment of this novel treatment option. Ann Surg Oncol. 17:484–491. DOI: 10.1245/s10434-009-0777-x. PMID: 19876691.
crossref
12. Edeline J, Touchefeu Y, Guiu B, Farge O, Tougeron D, Baumgaertner I, et al. 2020; Radioembolization plus chemotherapy for first-line treatment of locally advanced intrahepatic cholangiocarcinoma: a phase 2 clinical trial. JAMA Oncol. 6:51–59. DOI: 10.1001/jamaoncol.2019.3702. PMID: 31670746. PMCID: PMC6824230.
crossref
13. Riby D, Mazzotta AD, Bergeat D, Verdure L, Sulpice L, Bourien H, et al. 2020; Downstaging with radioembolization or chemotherapy for initially unresectable intrahepatic cholangiocarcinoma. Ann Surg Oncol. 27:3729–3737. DOI: 10.1245/s10434-020-08486-7. PMID: 32472411.
crossref
14. Kato A, Shimizu H, Ohtsuka M, Yoshitomi H, Furukawa K, Takayashiki T, et al. 2015; Downsizing chemotherapy for initially unresectable locally advanced biliary tract cancer patients treated with gemcitabine plus cisplatin combination therapy followed by radical surgery. Ann Surg Oncol. 22 Suppl 3:S1093–S1099. DOI: 10.1245/s10434-015-4768-9. PMID: 26240009.
crossref
15. Rayar M, Sulpice L, Edeline J, Garin E, Levi Sandri GB, Meunier B, et al. 2015; Intra-arterial yttrium-90 radioembolization combined with systemic chemotherapy is a promising method for downstaging unresectable huge intrahepatic cholangiocarcinoma to surgical treatment. Ann Surg Oncol. 22:3102–3108. DOI: 10.1245/s10434-014-4365-3. PMID: 25623598.
crossref
16. Le Roy B, Gelli M, Pittau G, Allard MA, Pereira B, Serji B, et al. 2018; Neoadjuvant chemotherapy for initially unresectable intrahepatic cholangiocarcinoma. Br J Surg. 105:839–847. DOI: 10.1002/bjs.10641. PMID: 28858392.
crossref
17. Kamarajah S, Giovinazzo F, Roberts KJ, Punia P, Sutcliffe RP, Marudanayagam R, et al. 2020; The role of down staging treatment in the management of locally advanced intrahepatic cholangiocarcinoma: review of literature and pooled analysis. Ann Hepatobiliary Pancreat Surg. 24:6–16. DOI: 10.14701/ahbps.2020.24.1.6. PMID: 32181423. PMCID: PMC7061034.
crossref
18. Lunsford KE, Javle M, Heyne K, Shroff RT, Abdel-Wahab R, Gupta N, et al. 2018; Liver transplantation for locally advanced intrahepatic cholangiocarcinoma treated with neoadjuvant therapy: a prospective case-series. Lancet Gastroenterol Hepatol. 3:337–348. Erratum in: Lancet Gastroenterol Hepatol 2018;3:e3. DOI: 10.1016/S2468-1253(18)30045-1. PMID: 29548617.
crossref
19. Moris D, Rahnemai-Azar AA, Zhang X, Ntanasis-Stathopoulos I, Tsilimigras DI, Chakedis J, et al. 2017; Program death-1 immune checkpoint and tumor microenvironment in malignant liver tumors. Surg Oncol. 26:423–430. DOI: 10.1016/j.suronc.2017.08.005. PMID: 29113661.
crossref
20. Ju JY, Dibbern ME, Mahadevan MS, Fan J, Kunk PR, Stelow EB. 2020; Mismatch repair protein deficiency/microsatellite instability is rare in cholangiocarcinomas and associated with distinctive morphologies. Am J Clin Pathol. 153:598–604. DOI: 10.1093/ajcp/aqz199. PMID: 31844887.
crossref
21. Gou M, Zhang Y, Liu T, Si H, Wang Z, Yan H, et al. 2021; PD-1 inhibitors could improve the efficacy of chemotherapy as first-line treatment in biliary tract cancers: a propensity score matching based analysis. Front Oncol. 11:648068. DOI: 10.3389/fonc.2021.648068. PMID: 34221968. PMCID: PMC8248534.
crossref
22. Zhang Z, Zhang W, Wang H, Hu B, Wang Z, Lu S. 2021; Successful treatment of advanced intrahepatic cholangiocarcinoma with a high tumor mutational burden and PD-L1 expression by PD-1 blockade combined with tyrosine kinase inhibitors: a case report. Front Immunol. 12:744571. DOI: 10.3389/fimmu.2021.744571. PMID: 34603331. PMCID: PMC8484748.
crossref
23. Rizzo A, Ricci AD, Brandi G. 2021; Durvalumab: an investigational anti-PD-L1 antibody for the treatment of biliary tract cancer. Expert Opin Investig Drugs. 30:343–350. DOI: 10.1080/13543784.2021.1897102. PMID: 33645367.
crossref
24. Borger DR, Tanabe KK, Fan KC, Lopez HU, Fantin VR, Straley KS, et al. 2012; Frequent mutation of isocitrate dehydrogenase (IDH)1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. Oncologist. 17:72–79. DOI: 10.1634/theoncologist.2011-0386. PMID: 22180306. PMCID: PMC3267826.
crossref
25. Boscoe AN, Rolland C, Kelley RK. 2019; Frequency and prognostic significance of isocitrate dehydrogenase 1 mutations in cholangiocarcinoma: a systematic literature review. J Gastrointest Oncol. 10:751–765. DOI: 10.21037/jgo.2019.03.10. PMID: 31392056. PMCID: PMC6657309.
crossref
26. Abou-Alfa GK, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, et al. 2020; Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 21:796–807. Erratum in: Lancet Oncol 2020;21:e462. DOI: 10.1016/S1470-2045(20)30157-1. PMID: 32416072.
crossref
27. Graham RP, Barr Fritcher EG, Pestova E, Schulz J, Sitailo LA, Vasmatzis G, et al. 2014; Fibroblast growth factor receptor 2 translocations in intrahepatic cholangiocarcinoma. Hum Pathol. 45:1630–1638. DOI: 10.1016/j.humpath.2014.03.014. PMID: 24837095.
crossref
28. Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. 2020; Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 21:671–684. DOI: 10.1016/S1470-2045(20)30109-1. PMID: 32203698.
crossref
29. Javle M, Lowery M, Shroff RT, Weiss KH, Springfeld C, Borad MJ, et al. 2018; Phase II study of BGJ398 in patients with FGFR-altered advanced cholangiocarcinoma. J Clin Oncol. 36:276–282. DOI: 10.1200/JCO.2017.75.5009. PMID: 29182496. PMCID: PMC6075847.
crossref
30. Ikeda M, Ioka T, Fukutomi A, Morizane C, Kasuga A, Takahashi H, et al. 2018; Efficacy and safety of trametinib in Japanese patients with advanced biliary tract cancers refractory to gemcitabine. Cancer Sci. 109:215–224. DOI: 10.1111/cas.13438. PMID: 29121415. PMCID: PMC5765304.
crossref
31. Bekaii-Saab T, Phelps MA, Li X, Saji M, Goff L, Kauh JS, et al. 2011; Multi-institutional phase II study of selumetinib in patients with metastatic biliary cancers. J Clin Oncol. 29:2357–2363. DOI: 10.1200/JCO.2010.33.9473. PMID: 21519026. PMCID: PMC3107751.
crossref
32. Subbiah V, Lassen U, Élez E, Italiano A, Curigliano G, Javle M, et al. 2020; Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 21:1234–1243. DOI: 10.1016/S1470-2045(20)30321-1. PMID: 32818466.
crossref
33. Lavingia V, Fakih M. 2016; Impressive response to dual BRAF and MEK inhibition in patients with BRAF mutant intrahepatic cholangiocarcinoma-2 case reports and a brief review. J Gastrointest Oncol. 7:E98–E102. DOI: 10.21037/jgo.2016.09.13. PMID: 28078132. PMCID: PMC5177579.
crossref
34. Churi CR, Shroff R, Wang Y, Rashid A, Kang HC, Weatherly J, et al. 2014; Mutation profiling in cholangiocarcinoma: prognostic and therapeutic implications. PLoS One. 9:e115383. DOI: 10.1371/journal.pone.0115383. PMID: 25536104. PMCID: PMC4275227.
crossref
35. Galdy S, Lamarca A, McNamara MG, Hubner RA, Cella CA, Fazio N, et al. 2017; HER2/HER3 pathway in biliary tract malignancies; systematic review and meta-analysis: a potential therapeutic target? Cancer Metastasis Rev. 36:141–157. DOI: 10.1007/s10555-016-9645-x. PMID: 27981460. PMCID: PMC5385197.
crossref
36. Amin NEL, Hansen TF, Fernebro E, Ploen J, Eberhard J, Lindebjerg J, et al. 2021; Randomized Phase II trial of combination chemotherapy with panitumumab or bevacizumab for patients with inoperable biliary tract cancer without KRAS exon 2 mutations. Int J Cancer. 149:119–126. DOI: 10.1002/ijc.33509. PMID: 33561312.
crossref
37. Golan T, Raitses-Gurevich M, Kelley RK, Bocobo AG, Borgida A, Shroff RT, et al. 2017; Overall survival and clinical characteristics of BRCA-associated cholangiocarcinoma: a multicenter retrospective study. Oncologist. 22:804–810. DOI: 10.1634/theoncologist.2016-0415. PMID: 28487467. PMCID: PMC5507643.
crossref
38. Fouassier L, Marzioni M, Afonso MB, Dooley S, Gaston K, Giannelli G, et al. 2019; Signalling networks in cholangiocarcinoma: molecular pathogenesis, targeted therapies and drug resistance. Liver Int. 39 Suppl 1:43–62. DOI: 10.1111/liv.14102. PMID: 30903728.
crossref
39. Isomoto H. 2009; Epigenetic alterations in cholangiocarcinoma-sustained IL-6/STAT3 signaling in cholangio- carcinoma due to SOCS3 epigenetic silencing. Digestion. 79 Suppl 1:2–8. DOI: 10.1159/000167859. PMID: 19153483.
crossref
40. Wang W, Zhong W, Yuan J, Yan C, Hu S, Tong Y, et al. 2015; Involvement of Wnt/β-catenin signaling in the mesenchymal stem cells promote metastatic growth and chemoresistance of cholangiocarcinoma. Oncotarget. 6:42276–42289. DOI: 10.18632/oncotarget.5514. PMID: 26474277. PMCID: PMC4747224.
crossref

Table 1
Active and Pending National Clinical Trials.gov Registered Trials for neoadjuvant chemotherapy for intrahepatic cholangiocarcinoma
Trial ID Author/institution No. of patients Arm(s) Primary outcome Status
NCT04961788 Shanghai Zhongshan Hospital 30 Toripalimab + Gemox 12-month ORR Recruiting
NCT04989218 University of Alabama at Birmingham 20 Durvalumab, Tremelimumab + platinum-based chemotherapy 12-week (4 cycle) ORR Recruiting
NCT04523402 Shen Feng, Eastern Hepatobiliary Surgery Hospital 100 Oxaliplatin + Gemcitabine & Resection versus Resection only 24-month PFS Not yet recruiting
NCT05290116 Sun Yat-sen University 17 Hepatic Arterial Infusion Chemotherapy + Tislelizumab and Apatinib 12-month ORR Recruiting
NCT04954781 Fudan University 25 Transarterial chemoembolization + Tislelizumab 24-month ORR Recruiting
NCT04546828 Samsung Medical Center 34 Gemcitabine, Cisplatin, Nab-Paclitaxel 16-week increased rate of R0 resection Withdrawn
NCT04669496 Shanghai Zhongshan Hospital 178 Gemox + Lenvatinib, Toripalimab versus Capecitabine 24-month EFS Recruiting
NCT04523402 Eastern Hepatobiliary Surgery Hospital 100 Gemox vs No neoadjuvant 24-month EFS Not yet recruiting

ORR, overall response rate; PFS, progression-free survival; EFS, event-free survival.

Table 2
Active and Pending National Clinical Trials.gov Registered Trials for immunotherapy for intrahepatic hepatic cholangiocarcinoma
Trial ID Author/institution No. of patients Arm(s) Target of drug Phase Setting Primary outcomes measured Status Preliminary outcomesa)
NCT02829918 H. Lee Moffitt Cancer Center 54 Nivolumab PD-1 II Unresectable 16-month ORR, 36-month OS, PFS Active, not recruiting ORR after 4 cycles of treatment is 22%, average PFS is 3.68 months (2.30–5.69 months)
NCT03046862 Seoul National University Hospital 31 Durvalumab + Tremelimumab + Gemcitabine/Cisplatin PD-L1, CTLA-4 II Unresectable or recurrent cancer RR at 6 weeks, PFS, OS Active, not recruiting N/A
NCT05223816 Virogin Biotech Ltd. 41 VG161 PD-L1 IIa/IIb Unresectable 3-month PFS & 12-month ORR Active, not recruiting N/A
NCT04361331 Shanghai Zhongshan Hospital 60 Lenvatinib + Toripalimab versus Lenvatinib + Gemox chemotherapy PD-1 II Unresectable 12-month ORR Active, not recruiting N/A
NCT03260712 European Organisation for Research and Treatment of Cancer 50 Gemcitabine + Cisplatin + Pembrolizumab PD-1 II Unresectable 6-month PFS Active, not recruiting N/A
NCT03473574 AIO-Studien-gGmbH 128 Durvalumab + Tremelimumab + Gemcitabine +/– Cisplatin PD-1, CTLA-4 II Unresectable 30-month ORR Active, not recruiting N/A
NCT03875235 AstraZeneca 810 Gemcitabine + Cisplatin +/– Durvalumab PD-1 III Unresectable 40-month OS Active, not recruiting Combining Durvalumab: OS 12.8 months (11.1–14.0 months), OS at 18 months 35.1%, PFS 7.2 months (6.4–7.4 months)
Without Durvalumab: OS 11.5 months (10.1–12.5 months), OS at 18 months 25.6%, PFS 5.7 months (5.6–6.7 months)
NCT03201458 National Cancer Institute 76 Atezolizumab +/– Cobimetinib PD-L1, MEK II Unresectable 12-month PFS Active, not recruiting N/A
NCT03101566 University of Michigan Rogel Cancer Center 64 Nivolumab + Gemcitabine/Cisplatin PD-1, CTLA-1 II Unresectable 6-month PFS Active, not recruiting % of patients alive and without disease progression at 6 months Gemcitabine + Cisplatin + Nivolumab 59.4% vs. Nivolumab + Ipilimumab 21.2%
NCT03785873 University of Michigan Rogel Cancer Center 34 Nivolumab + Nanoliposomal-Irinotecan, 5-FU, Leucovorin PD-1 Ib/II Unresectable 4-week AE rate, 2-year PFS Active, not recruiting N/A
NCT02821754 National Cancer Institute 54 Durvalumab + Tremelimumab +/– TACE + RFA or Cryoablation PD-1, CTLA-4 II Unresectable 6-month PFS, AE rate Active, not recruiting 6 month PFS, 36 patients in RFA/TACE group; average month survival is 2.8 months (1.3–4.8 months), only 1 patient in RFA/cryoablation group and still alive
NCT03046862 Seoul National University Hospital 31 Durvalumab + Tremelimumab + Gemcitabine/Cisplatin PD-1, CTLA-4 II Unresectable 6-week RR Active, not recruiting N/A
NCT03230318 Basilea Pharmaceutica 148 Derazantinib FGFR2 II Unresectable 32-week ORR & PFS Completed N/A
NCT03639935 University of Michigan Rogel Cancer Center 35 Rucaparib + Nivolumab PD-1, PARP II Unresectable 4-month OS, PFS Recruiting N/A
NCT03867370 Shangai Junshi Bioscience 40 Toripalimab + Lenvatinib PD-1 Ib/II Neoadjuvant Treatment Pathological Response Rate, 2-month ORR, R0 resection rate Recruiting N/A
NCT03704480 GERCOR – Multidisciplinary Oncology Group 106 Durvalumab + Tremelimumab +/– Paclitaxel PD-1, CTLA-4 II Unresectable 6-month OS Recruiting N/A
NCT03898895 Sun Yat-sen University 36 Camrelizumab + Radiotherapy PD-1 II Unresectable 12-month PFS, OS, AE rate Recruiting N/A
NCT05251662 Tianjin Medical University Cancer Institute and Hospital 90 GEMOX + Bevacizumab + Sintilimab vs Sintilimab + GEMOX PD-L1 II Unresectable 90-day ORR Recruiting N/A
NCT03482102 Massachusetts General Hospital 70 Tremelimumab + Duravalumab + Radiation PD-1, CTLA-4 II Unresectable 3-year ORR, AE rate Recruiting N/A
NCT03695952 Asan Medical Center 100 Nivolumab or Pembrolizumab PD-1 Prospective Cohort Unresectable 6-month ORR, AE rate Recruiting N/A
NCT03796429 Shanghai Zhongshan Hospital 40 Gemcitabine + Cisplatin + Toripalimab (up to 2 years) PD-1 II Unresectable 36-month PFS Recruiting N/A
NCT03478488 3D Medicines Co. 480 KN035 +/– Gemcitabine/Oxaliplatin PD-L1 III Unresectable 12-week OS Recruiting N/A
NCT04295317 Shanghai Zhongshan Hospital 65 Capecitabine + SHR-1210 PD-1 II Unresectable 24-month Recurrence free survival Recruiting N/A
NCT04301778 Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins 30 Durvalumab + SNDX-6352 PD-1, CSF-1R II Unresectable 4-year ORR and drug-AE Active, not recruiting N/A
NCT05052099 University Hospital, Essen 35 FOLFOX6 + Atezolizumab + Bevacizumab PD-1, VEGF 1b/II Unresectable 12-week ORR Recruiting N/A
NCT01853618 National Cancer Institute 61 Tremelimumab +/– RFA +/– TACE CTLA-4 I Unresectable 12-month AEs, PFS, OS Completed PFS 8.6 months for Tremelimumab + Cryoablation as compared to 3.4 months for Tremelimumab + RFA
NCT03110328 Samsung Medical Center 33 Pembrolizumab PD-1 II Unresectable, second-line treatment 12-month ORR, PFS, OS Completed N/A
NCT03111732 National Cancer Institute 11 Pembrolizumab + Oxaliplatin/Capecitabine PD-1 II Unresectable 5-month PFS Completed Median amount of time patient survives without disease progression for 5 months after treatment, Pembrolizumab + Oxaliplatin + Capectitabine, 4.54 months (2.5–9.6 months)

RR, response rate; PFS, progression-free survival; OS, overall survival; ORR, overall response rate; AE, adverse events; RFA, radiofrequency ablation; TACE, Transarterial Catheter Chemoembolization; CTLA-4, cytotoxic T-lymphocyte-associated protein 4; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1; VEGF, vascular endothelial growth factor; PARP, poly (ADP-ribose) polymerase; FGFR, fibroblast growth factor receptor.

a)Preliminary Selected Outcomes as of 12/20/2022. Not all preliminary data reported had an accompanying statistical analysis.

TOOLS
Similar articles