Loading

Commentary Open Access
Volume 3 | Issue 2 | DOI: https://doi.org/10.46439/cancerbiology.3.046

Combination immunotherapy: The new roadmap for the treatment of intrahepatic cholangiocarcinoma

  • 1Department of Genetics and Genomic Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, USA
  • 2Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, USA
  • 3Division of Liver Diseases, Department of Medicine, Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mt Sinai, New York, USA
+ Affiliations - Affiliations

*Corresponding Author

Daniela Sia, daniela.sia@mssm.edu

Received Date: November 18, 2022

Accepted Date: November 25, 2022

Abstract

Intrahepatic cholangiocarcinoma (iCCA) is a catastrophic malignancy of the intrahepatic bile ducts and one of the neoplasms with incidence rates that have been rising faster than almost any other cancer. This steady increase combined with the high mortality rates underscore the fact that optimal management of iCCA remains a challenge. While immune checkpoint inhibitors (ICIs) as single agents have elicited discouraging results in patients with iCCA, the combination of chemotherapy plus anti-PD-L1 blockade has recently become the new standard of care, underscoring the importance of designing rational combination strategies able to increase the therapeutic efficacy of ICIs. To do so, a thorough understanding of the tumor-immune interactions is becoming critical. This commentary aims to discuss recent advancements in our understanding of the complex relationship between tumor cells and the surrounding immune microenvironment of iCCA, with particular emphasis on our recent manuscript published in the journal Gut (Martin-serrano et al., 2022) [1].

Commentary

With an occurrence of 1–2 new cases per 100,000 persons annually in the US, intrahepatic cholangiocarcinoma (iCCA) – a cancer that forms in the small bile ducts within the liver – is relatively rare but highly fatal, accounting for less than 3% of all gastrointestinal cancers, yet nearly 20% of all deaths [2]. Despite the low prevalence of iCCA in most high-income countries (global age standardized incidence rate of 1.4 cases per 100,000 person-years) [3], a steady increase in its incidence rates has been recorded over the past decades. In the US the incidence of iCCA has increased by 128% over the past 40-years [4]. iCCAs are rarely diagnosed at early stage due to the lack of specific symptoms. Late diagnosis compromises the efficacy of hepatic resection, which represents the only curative option for early stage iCCA2, and palliative chemotherapy with gemcitabine plus cisplatin (GemCis) has been the standard of care (SOC) for advanced disease for the past 12 years, with a median overall survival (OS) of just 12 months [5]. Therefore, patients with iCCA face a highly dismal prognosis, with 95% of patients dying within five years [6]. 
In the past decade, the high incidence and mortality rates have sparked renewed interest in this long-forgotten disease. As a result, multi-institutional collaborations as well as international consortia have assembled human tissue collections and provided detailed information on the molecular landscape of human iCCA, ultimately laying the groundwork for the evaluation of targeted therapies. In this regard, our group and others identified recurrent and targetable genetic alterations (i.e., gene fusions in FGFR2, NRTK, mutations in IDH, etc) [7-13]. Despite such breakthroughs and the increase in clinical trials investigating targeted therapies, progress has been modest with the most promising drugs, FGFR and IDH inhibitors, conferring a survival benefit of only 2-4 months [14-16]. 
Immune checkpoint inhibitors (ICIs) have revolutionized the management of several solid cancers [17,18]; yet, results in iCCA have been modest. ICIs are a new class of immunotherapeutic antibodies that block T cell surface-expressed inhibitory receptors or their ligands to unleash the patient’s own immune response against the tumor [16]. As a result, monoclonal antibodies (moAb) blocking the inhibitory receptor programmed cell death protein 1 (PD1) or its ligand PD-L1 have quickly become the new SOC in many cancers [19]. Unfortunately, despite the great success and long-lasting clinical benefit, only a small fraction of patients responds to PD1/PD-L1 checkpoint blockade. ICIs are particularly effective in patients with mismatch repair (MMR) deficiency, with an observed clinical response in up to 40% of patients [20]. Unfortunately, MMR deficiency occurs in only 3% of iCCA patients [21-24],  and initial clinical results in unselected CCA populations treated with the anti-PD1 moAb pembrolizumab as single agent have been disappointing, with only 6% to 13% of clinical responses [25]. Recently, the TOPAZ-1 trial found that the addition of durvalumab, a moAb targeting PD-L1, to the SOC chemotherapy significantly prolongs overall survival and increases objective response rates from 19% to 27% in patients with iCCA [26]. Based on these results, the Food and Drug Administration (FDA) approved this combination as new SOC for advanced iCCA in September 2022. Despite this breakthrough, these results are far from optimal given the mild improvement in median overall survival (from 11.5 to 12.8 months26). Hence, the identification of biomarkers for optimal patient selection as well as the design of rational combination strategies able to increase the efficacy of ICIs in iCCA remain top priorities.
The lack of understanding of the interaction between tumor cells and the immune microenvironment represents one of the main reasons for unsatisfactory clinical trial outcomes in iCCA. The first pan-cancer immunogenomic analyses characterized the intratumoral immune landscape of over 20 solid cancers (so-called Cancer Immunome Atlas) and suggested that tumor-intrinsic genomic alterations determine the tumor immunophenotype and escape mechanisms [27,28]. Unfortunately, no iCCA cases were included in these studies. To fill this knowledge gap, our group virtually deconstructed the tumor immune microenvironment (TIME) of a large collection of human iCCA samples [1]. By considering elements of the Stroma, Tumor and Immune Microenvironment, our algorithm successfully classified iCCA tumors into 5 robust groups, called STIM classes, encompassing both immune inflamed (or hot) and non-inflamed (or cold) profiles [1]. Compared to previous studies which described distinct iCCA classes based on either tumor [29,30] or immune-only [31] related features, our unsupervised approach unveiled actionable genotype-immunophenotype relationships, with each class being defined by a unique TIME associated with distinct underlying genetic alterations. In line with emerging single cell RNA-sequencing-based analyses [32,33] and the immunotolerant nature of the liver [34], we observed that more than half of iCCAs were non-inflamed tumors with abundance of immunosuppressive components (i.e. macrophages, regulatory T cells, etc.). The non-inflamed tumors entail 3 distinct classes with the largest being the “hepatic stem-like” class, so called because of the enrichment of stem cell features along with high rate of genetic alterations in driver genes, including mutations in BAP1, IDH1/2 and FGFR2 fusions. Immunophenotypically, the tumors in this class are characterized by low cytolytic activity and an abundance of immunosuppressive macrophages. Interestingly, a recent study found that pharmacologic inhibition of mutant IDH1 reactivates CD8+ T cells in an iCCA subcutaneous murine model [35] confirming the validity of the genotype-immunophenotype relationships identified by our algorithm, and that the immunogenicity of this neoplasm is indeed influenced by its underlying genotype. Similarly, recent studies confirmed the association between FGFR2 fusions and reduced immune infiltration [36]. Evidence from other tumors points to an important immunosuppressive role for oncogenic FGFR signaling in shaping the TIME (i.e., upregulation of PD-L1, inhibition of IFN? signaling, increase of myeloid-derived suppressive cells, T cell exclusion) [37-40]. In this regard, emerging experimental [41-43] and clinical [44] evidence from lung, renal and head and neck carcinoma showed promising results with the addition of FGFR inhibitors to ICIs. As an example, in a FGFR2-driven autochthonous mouse model of lung cancer [41] treatment with the FGFR inhibitor erdafitinib elicited unique changes in the TIME (i.e., reduction of regulatory T cells, increase of T cell infiltration, etc.), and synergized with anti-PD1 ICI by inducing tumor cell killing that enhanced tumor antigen presentation and antitumor T-cell responses. Unfortunately, similar studies in iCCA are missing. Major hurdle to these clinical goals is that current preclinical models of FGFR2 fusion-driven iCCA include human cell lines [7,45-47] and patient-derived xenografts (PDX) models [48], which albeit useful, lack an immunocompetent host environment and therefore, do not fully mimic the immunobiology of iCCA patients. In a recent study, a series of murine organoid lines have been engineered from TP53 null mice with FGFR2 fusions constructs [49], which could be potentially injected into immunocompetent syngeneic hosts (i.e., same genetic background as the organoids). Nonetheless, loss of TP53 and FGFR2 fusion rarely co-occur in human iCCA [50] while orthotopic transplantation of fully developed tumor bypasses the initial steps of tumorigenesis, potentially leading to aberrant inflammatory responses. Future studies in iCCA aimed at establishing clinically-relevant FGFR2 fusion-driven models are anxiously awaited since they will offer the valuable opportunity to understand the impact of this alteration on the TIME of iCCA and provide the rationale for more effective ICI combinations able to enhance the extent of initial responses in FGFR2-driven iCCA patients. The other two non-inflamed classes, named Tumor classical and Desert-like, showed enrichment of co-occurrent mutations in KRAS and TP53 genes, and were significantly associated with abundance of regulatory T cells. Cooperation between TP53 and KRAS in driving immune evasion has been described in ovarian cancer [51], although no studies have been reported in iCCA.
The ‘inflamed class’ represents less than 35% of this cohort and encompassed two classes named “Immune classical” and “Inflammatory Stroma”. Despite that both classes were enriched in immune infiltration, great heterogeneity was observed in terms of their stromal characteristics, with the Inflammatory Stroma being enriched in myofibroblastic CAFs, signatures of resistance to ICIs and KRAS mutations. In this regard, emerging evidence suggests that gain-of-function KRAS mutations contribute to the immunosuppressive TIME that supports cancer growth through various mechanisms and thus, compromise clinical responses to ICIs [52-55]. Accordingly, treatment of colorectal cancer murine models with sotorasib, the first FDA-approved KRASG12C inhibitor, results in an increase of actively proliferating T cells and produces durable responses in combination with PD1 moAb [56]. In line with this, we demonstrated that KRAS inhibition with the potent BI3406 was able to sensitize KRAS-mutant iCCA to anti-PD1 in a subcutaneous model of the disease and in a KRAS-mutant iCCA patient-derived organoid co-culture with autologous T cells, further confirming a key immunosuppressive role for mutant KRAS in iCCA. 
Considering the emerging data with selective inhibitors in both KRAS [1]- and IDH1 [35]-mutant iCCA, it is clear that targeting tumor-intrinsic vulnerabilities that modulate anti-tumor T cell responses may identify new combinations for improving the efficacy of ICIs in this malignancy. To do so, preclinical models able to reproduce the underlying genotype and tumor immune cell interactions of iCCA are urgently needed. In our study, we performed a comprehensive immunogenomic cross-species analysis between human iCCAs and the most commonly used hydrodynamic gene delivery-based mouse models [1]. These models offer several advantages compared to traditional germline-based genetically engineered mouse models since they are able to quickly develop autochthonous and mosaic liver tumors harboring specific and customizable genetic alterations. In these models, plasmid DNA either targeting tumor suppressors or inducing overexpression of oncogenes are delivered to the hepatocytes within the liver by hydrodynamic tail vein injection [57]. Transient transduction of the hepatocytes induces their trans-differentiation into cholangiocytes and subsequent neoplastic transformation as demonstrated in previous lineage tracing studies [58-60]. The hepatocytic origin of iCCA is consistent with genomic studies of human samples [61] suggesting not only that iCCA can derive from both hepatocytes and cholangiocytes, but also that hepatocytes may actually represent the main source, ultimately generating a lot of controversy in the field. Interestingly, in further support of genomic studies in human samples [61,62], we identified hydrodynamic gene delivery-based mouse models able to closely recapitulate the immunobiology of two largest STIM classes, Inflammatory Stroma and Hepatic Stem-like, suggesting that, regardless of their cell-of-origin, hepatocyte-derived iCCAs are indeed bone fide iCCA and highly resemble the human disease. In particular, we described that murine tumors driven by activating KRAS mutations and p19 loss closely resemble iCCA tumors of the Inflammatory Stroma, which are enriched in KRAS mutations with abundant stroma and immune infiltration. Similar to their human counterpart, KRAS-driven murine tumors showed a significant increase in in CD8+ T cells and enrichment of dysfunctional CD8+PD1+ T cells. On the other end, murine tumors carrying NOTCH1 or YAP1 activation in combination with AKT1 activation resembled cold tumors of the Hepatic Stem-like. Overall, this analysis demonstrates validity of these mouse models in general, and for pre-clinical studies of ICIs in particular.
The discovery of targetable FGFR2 fusions and IDH1 mutations has accelerated the use of personalized treatment approaches in iCCA. However, whether our STIM classifications of iCCA could effectively guide the management of the diseases still remains unclear. Similarly, the predictive role of tumor mutational burden (TMB), neo-antigen burden (TNB), and chromosomal aberration load in predicting response to ICI in iCCA requires further elucidation. Interestingly, while we did not observe any association between our STIM classes and TMB or TNB [1], a recent study reported that highly infiltrated tumors are associated with significantly higher TNB, suggesting that the immunogenicity of the underlying tumor mutations might direct immune infiltration [36]. Similarly, we observed that focal chromosomal losses impacting genes of the interferon pathway were more frequent in the non-inflamed tumor classes, potentially impacting their immunogenicity [1]. The use of immunogenomic information in clinical practice would require validation in prospective clinical trials using pre- and post-treatment biopsies and high-resolution technologies, such as single cell profiling. However, great challenges exist to guarantee routine access to tissue biopsies and overcome the limitation of high intratumor genomic and immune heterogeneity. A valid alternative would be to rely on the use of liquid biopsies to detect circulating biomarkers [63]. Given the emerging association between the genotype and immunogenicity of iCCA, an alternative would be the detection of oncogenic mutations (i.e. KRAS, TP53, IDH1) or mutation/neoantigen load using circulating tumor DNA to guide the selection of patients for ICI combination strategies. To achieve these clinical goals, improved pre-clinical models that faithfully recapitulate diverse patient-specific and tumor-intrinsic genotypes will significantly advance our understanding of the mechanisms underpinning how the underlying genotype influences immunogenicity as well as the efficacy of ICI in iCCA. 
While extremely valuable, murine tumors can feasibly model only few genetic alterations and therefore reproducing the genetic heterogeneity of human iCCA in mice remains difficult. Given that the crosstalk between oncogenic pathways instructs the composition of the TIME and dictate therapeutic efficacy [51,64], precision modeling of tumor genetic heterogeneity in a patient-specific manner is pivotal for the discovery of optimal treatment strategies. To overcome these limitations, recent advances in 3D cell cultures have led to the development of cancer patient-derived organoid models (PDO) that reproduce the underlying (epi)genetic, proteomic, morphological and pharmacotypic features of primary patient tumors [65-68], including CCA [69]. Comparison between drug responses in patients with gastrointestinal cancers and matched PDOs showed >80% accuracy in predicting positive treatment response and 100% accuracy in predicting no response [70]. Thereby, PDOs have been extensively used in drug screens and have shown feasibility for drug discovery in different cancers [71-73]. A significant improvement in PDO modeling is the incorporation of immune cells to study tumor-immune cell interactions and responses to ICIs in a patient-specific manner. However, only few approaches leverage PDO/immune cell co-cultures. In air-liquid interface cultures, PDOs are grown in collagen in an inner trans-well from minced tumor fragments [74] and preserve diversity of endogenous immune cells for a maximum of ~4-6 weeks [75]. Alternatively, a key study demonstrated the feasibility of generating PDO-reactive T cells from peripheral blood of colorectal and lung cancer patients [76], and CD8 T cell responses were observed in few samples upon treatment with PD1 ICI [76]. In iCCA, the only study that attempted PDO/T cell co-cultures from three patients used allogeneic blood–derived T cells, which were polyclonally stimulated to elicit a response [77]. Such PDO/T cell interactions do not recapitulate tumor-specific T cell (TST) interactions and immune synapse formation, which are key to functional T cell responses to ICI. Furthermore, the latter studies do not use intratumoral T cells that have been exposed to the TIME of the primary tumor and lack functional readouts that capture the breadth of anti-tumor T cell responses. Lastly, major challenge in these approaches is reproducing the full spectrum of cellular interactions within the TIME. Nevertheless, T cells are key effectors of anti-tumor immunity and optimizing autologous PDO/T cell co-cultures is advantageous in modeling tumor-T cell interactions, identifying TST and testing strategies targeting tumor vulnerabilities in combination with ICI.
In conclusion, despite the great advancements in our understanding of the immunobiology of iCCA at the bulk and single cell level, a lot more work remains to be done. Future studies should focus on dissecting the mechanisms underpinning immune evasion and immunosuppression driven by different tumor-intrinsic alterations. At the same time, more accurate preclinical platforms for large ICI screenings are anxiously awaited. Clinical trials should include genomic analyses of pre-treatment biopsies to shed light on potential biomarkers of response and resistance to ultimately optimize patient selection and improve the outcome of patients affected by this fatal malignancy.

 

Acknowledgments

Figures 1 was created with Biorender. 

Disclosure

The authors declare no conflict of interest.

References

1. Martin-Serrano MA, Kepecs B, Torres-Martin M, Bramel ER, Haber PK, Merritt E, et al. Novel microenvironment-based classification of intrahepatic cholangiocarcinoma with therapeutic implications. Gut. 2022 May 18;gutjnl-2021-326514.

2. Banales JM, Marin JJ, Lamarca A, Rodrigues PM, Khan SA, Roberts LR, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nature Reviews Gastroenterology & Hepatology. 2020 Sep;17(9):557-88.

3. Rumgay H, Ferlay J, de Martel C, Georges D, Ibrahim AS, Zheng R, et al. Global, regional and national burden of primary liver cancer by subtype. European Journal of Cancer. 2022 Jan 1;161:108-18.

4. Saha SK, Zhu AX, Fuchs CS, Brooks GA. Forty-year trends in cholangiocarcinoma incidence in the US: intrahepatic disease on the rise. The oncologist. 2016 May;21(5):594-9.

5. Valle J, Wasan H, Palmer DH, Cunningham D, Anthoney A, Maraveyas A, et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. New England Journal of Medicine. 2010 Apr 8;362(14):1273-81.

6. Vithayathil M, Khan SA. Current epidemiology of cholangiocarcinoma in western countries. Journal of Hepatology. 2022 Aug 14.

7. Sia D, Losic B, Moeini A, Cabellos L, Hao K, Revill K, et al. Massive parallel sequencing uncovers actionable FGFR2–PPHLN1 fusion and ARAF mutations in intrahepatic cholangiocarcinoma. Nature communications. 2015 Jan 22;6(1):1-1.

8. Borger DR, Tanabe KK, Fan KC, Lopez HU, Fantin VR, Straley KS, et al. Frequent mutation of isocitrate dehydrogenase (IDH) 1 and IDH2 in cholangiocarcinoma identified through broad-based tumor genotyping. The oncologist. 2012 Jan;17(1):72-9.

9. Nakamura H, Arai Y, Totoki Y, Shirota T, Elzawahry A, Kato M, et al. Genomic spectra of biliary tract cancer. Nature Genetics. 2015 Sep;47(9):1003-10.

10. Chan-On W, Nairismägi ML, Ong CK, Lim WK, Dima S, Pairojkul C, et al. Exome sequencing identifies distinct mutational patterns in liver fluke–related and non-infection-related bile duct cancers. Nature Genetics. 2013 Dec;45(12):1474-8.

11. Jiao Y, Pawlik TM, Anders RA, Selaru FM, Streppel MM, Lucas DJ, et al. Exome sequencing identifies frequent inactivating mutations in BAP1, ARID1A and PBRM1 in intrahepatic cholangiocarcinomas. Nature Genetics. 2013 Dec;45(12):1470-3.

12. Arai Y, Totoki Y, Hosoda F, Shirota T, Hama N, Nakamura H, et al. Fibroblast growth factor receptor 2 tyrosine kinase fusions define a unique molecular subtype of cholangiocarcinoma. Hepatology. 2014 Apr;59(4):1427-34.

13. Wu YM, Su F, Kalyana-Sundaram S, Khazanov N, Ateeq B, Cao X, et al. Identification of targetable FGFR gene fusions in diverse cancers. Cancer Discovery. 2013 Jun;3(6):636-47.

14. Abou-Alfa GK, Sahai V, Hollebecque A, Vaccaro G, Melisi D, Al-Rajabi R, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. The Lancet Oncology. 2020 May 1;21(5):671-84.

15. Abou-Alfa GK, Macarulla T, Javle MM, Kelley RK, Lubner SJ, Adeva J, et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): a multicentre, randomised, double-blind, placebo-controlled, phase 3 study. The Lancet Oncology. 2020 Jun 1;21(6):796-807.

16. Makawita S, K Abou-Alfa G, Roychowdhury S, Sadeghi S, Borbath I, Goyal L, et al. Infigratinib in patients with advanced cholangiocarcinoma with FGFR2 gene fusions/translocations: the PROOF 301 trial. Future Oncology. 2020 Oct;16(30):2375-84.

17. Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature Reviews Cancer. 2012 Apr;12(4):252-64.

18. Sharma P, Allison JP. The future of immune checkpoint therapy. Science. 2015 Apr 3;348(6230):56-61.

19. Tang J, Yu JX, Hubbard-Lucey VM, Neftelinov ST, Hodge JP, Lin Y. Trial watch: the clinical trial landscape for PD1/PDL1 immune checkpoint inhibitors. Nature Reviews Drug Discovery. 2018 Dec 1;17(12):854-6.

20. Le DT, Uram JN, Wang H, Bartlett BR, Kemberling H, Eyring AD, et al. PD-1 blockade in tumors with mismatch-repair deficiency. New England Journal of Medicine. 2015 Jun 25;372(26):2509-20.

21. Goeppert B, Roessler S, Renner M, Singer S, Mehrabi A, Vogel MN, Pathil A, Czink E, Köhler B, Springfeld C, Pfeiffenberger J. Mismatch repair deficiency is a rare but putative therapeutically relevant finding in non-liver fluke associated cholangiocarcinoma. British journal of cancer. 2019 Jan;120(1):109-14.

22. 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. Hepato-Gastroenterology. 2002 May 1;49(45):604-8.

23. Momoi H, Itoh T, Nozaki Y, Arima Y, Okabe H, Satoh S, et al. Microsatellite instability and alternative genetic pathway in intrahepatic cholangiocarcinoma. Journal of Hepatology. 2001 Aug 1;35(2):235-44.

24. Weinberg BA, Xiu J, Lindberg MR, Shields AF, Hwang JJ, Poorman K, et al. Molecular profiling of biliary cancers reveals distinct molecular alterations and potential therapeutic targets. Journal of Gastrointestinal Oncology. 2019 Aug;10(4):652.

25. Piha‐Paul SA, Oh DY, Ueno M, Malka D, Chung HC, Nagrial A, et al. Efficacy and safety of pembrolizumab for the treatment of advanced biliary cancer: Results from the KEYNOTE‐158 and KEYNOTE‐028 studies. International Journal of Cancer. 2020 Oct 15;147(8):2190-8.

26. 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 Evidence. 2022 Jun 1:EVIDoa2200015.

27. Charoentong P, Finotello F, Angelova M, Mayer C, Efremova M, Rieder D, et al. Pan-cancer immunogenomic analyses reveal genotype-immunophenotype relationships and predictors of response to checkpoint blockade. Cell Reports. 2017 Jan 3;18(1):248-62.

28. Yang J, Zhao S, Wang J, Sheng Q, Liu Q, Shyr Y. A pan-cancer immunogenomic atlas for immune checkpoint blockade immunotherapy. Cancer Research. 2021 Dec 13.

29. Sia D, Hoshida Y, Villanueva A, Roayaie S, Ferrer J, Tabak B, et al. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes. Gastroenterology. 2013 Apr 1;144(4):829-40.

30. Andersen JB, Spee B, Blechacz BR, Avital I, Komuta M, Barbour A, et al. Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology. 2012 Apr 1;142(4):1021-31.

31. Job S, Rapoud D, Dos Santos A, Gonzalez P, Desterke C, Pascal G, et al. Identification of four immune subtypes characterized by distinct composition and functions of tumor microenvironment in intrahepatic cholangiocarcinoma. Hepatology. 2020 Sep;72(3):965-81.

32. Zhang M, Yang H, Wan L, Wang Z, Wang H, Ge C, et al. Single-cell transcriptomic architecture and intercellular crosstalk of human intrahepatic cholangiocarcinoma. Journal of Hepatology. 2020 Nov 1;73(5):1118-30.

33. Ma L, Hernandez MO, Zhao Y, Mehta M, Tran B, Kelly M, et al. Tumor cell biodiversity drives microenvironmental reprogramming in liver cancer. Cancer Cell. 2019 Oct 14;36(4):418-30.

34. Zheng M, Tian Z. Liver-mediated adaptive immune tolerance. Frontiers in Immunology. 2019 Nov 5;10:2525.

35. Wu MJ, Shi L, Dubrot J, Merritt J, Vijay V, Wei TY, et al. Mutant IDH inhibits IFNγ–TET2 signaling to promote immunoevasion and tumor maintenance in cholangiocarcinoma. Cancer Discovery. 2022 Mar 1;12(3):812-35.

36. Lin Y, Peng L, Dong L, Liu D, Ma J, Lin J, et al. Geospatial Immune Heterogeneity Reflects the Diverse Tumor–Immune Interactions in Intrahepatic Cholangiocarcinoma. Cancer Discovery. 2022 Oct 5;12(10):2350-71.

37. Akhand SS, Liu Z, Purdy SC, Abdullah A, Lin H, Cresswell GM, et al. Pharmacologic inhibition of FGFR modulates the metastatic immune microenvironment and promotes response to immune checkpoint blockade. Cancer Immunology Research. 2020 Dec;8(12):1542-53.

38. Li P, Huang T, Zou Q, Liu D, Wang Y, Tan X, et al. FGFR2 promotes expression of PD-L1 in colorectal cancer via the JAK/STAT3 signaling pathway. The Journal of Immunology. 2019 May 15;202(10):3065-75.

39. Ye T, Wei X, Yin T, Xia Y, Li D, Shao B, et al. Inhibition of FGFR signaling by PD173074 improves antitumor immunity and impairs breast cancer metastasis. Breast Cancer Research and Treatment. 2014 Feb;143(3):435-46.

40. Liu LI, Ye TH, Han YP, Song H, Zhang YK, Xia Y, et al. Reductions in myeloid-derived suppressor cells and lung metastases using AZD4547 treatment of a metastatic murine breast tumor model. Cellular Physiology and Biochemistry. 2014;33(3):633-45.

41. Palakurthi S, Kuraguchi M, Zacharek SJ, Zudaire E, Huang W, Bonal DM, et al. The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor ImmunityErdafitinib plus Anti–PD-1 Enhances Antitumor Responses. Cancer Immunology Research. 2019 Sep 1;7(9):1457-71.

42. Adachi Y, Kamiyama H, Ichikawa K, Fukushima S, Ozawa Y, Yamaguchi S, et al. Inhibition of FGFR Reactivates IFNg Signaling in Tumor Cells to Enhance the Combined Antitumor Activity of Lenvatinib with Anti-PD-1 Antibodies. Cancer Research. 2022 Jan 15;82(2):292-306.

43. Kono M, Komatsuda H, Yamaki H, Kumai T, Hayashi R, Wakisaka R, et al. Immunomodulation via FGFR inhibition augments FGFR1 targeting T-cell based antitumor immunotherapy for head and neck squamous cell carcinoma. Oncoimmunology. 2022 Dec 31;11(1):2021619.

44. Liu Y, Long L, Liu J, Zhu L, Luo F. Case report: anlotinib reverses nivolumab resistance in advanced primary pulmonary lymphoepithelioma-like carcinoma with FGFR3 gene amplification. Frontiers in Oncology. 2021:4041.

45. Goyal L, Saha SK, Liu LY, Siravegna G, Leshchiner I, Ahronian LG, et al. Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion–Positive CholangiocarcinomaAcquired FGFR2 Resistance Mutations in Cholangiocarcinoma. Cancer Discovery. 2017 Mar 1;7(3):252-63.

46. Liu PC, Koblish H, Wu L, Bowman K, Diamond S, DiMatteo D, et al. INCB054828 (pemigatinib), a potent and selective inhibitor of fibroblast growth factor receptors 1, 2, and 3, displays activity against genetically defined tumor models. PloS One. 2020 Apr 21;15(4):e0231877.

47. Goyal L, Shi L, Liu LY, Fece de la Cruz F, Lennerz JK, Raghavan S, et al. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion–Positive Intrahepatic CholangiocarcinomaTAS-120 Efficacy in FGFR Inhibitor–resistant Biliary Cancer. Cancer Discovery. 2019 Aug 1;9(8):1064-79.

48. Wu Q, Zhen Y, Shi L, Vu P, Greninger P, Adil R, et al. EGFR Inhibition Potentiates FGFR Inhibitor Therapy and Overcomes Resistance in FGFR2 Fusion–Positive Cholangiocarcinoma. Cancer Discovery. 2022 May 2;12(5):1378-95.

49. Cristinziano G, Porru M, Lamberti D, Buglioni S, Rollo F, Amoreo CA, et al. FGFR2 fusion proteins drive oncogenic transformation of mouse liver organoids towards cholangiocarcinoma. Journal of Hepatology. 2021 Aug 1;75(2):351-62.

50. Silverman IM, Hollebecque A, Friboulet L, Owens S, Newton RC, Zhen H, et al. Clinicogenomic Analysis of FGFR2-Rearranged Cholangiocarcinoma Identifies Correlates of Response and Mechanisms of Resistance to PemigatinibGenomic Profiling in FGFR2-Rearranged Cholangiocarcinoma. Cancer Discovery. 2021 Feb 1;11(2):326-39.

51. Blagih J, Zani F, Chakravarty P, Hennequart M, Pilley S, Hobor S, et al. Cancer-specific loss of p53 leads to a modulation of myeloid and T cell responses. Cell Reports. 2020 Jan 14;30(2):481-96.

52. Liao W, Overman MJ, Boutin AT, Shang X, Zhao D, Dey P, et al. KRAS-IRF2 axis drives immune suppression and immune therapy resistance in colorectal cancer. Cancer Cell. 2019 Apr 15;35(4):559-72.

53. Kortlever RM, Sodir NM, Wilson CH, Burkhart DL, Pellegrinet L, Swigart LB, et al. Myc cooperates with Ras by programming inflammation and immune suppression. Cell. 2017 Nov 30;171(6):1301-15.

54. Dias Carvalho P, Guimaraes CF, Cardoso AP, Mendonca S, Costa AM, Oliveira MJ, et al. KRAS Oncogenic Signaling Extends beyond Cancer Cells to Orchestrate the MicroenvironmentParacrine Signaling from KRAS-Mutant Cancer Cells. Cancer Research. 2018 Jan 1;78(1):7-14.

55. Wellenstein MD, de Visser KE. Cancer-cell-intrinsic mechanisms shaping the tumor immune landscape. Immunity. 2018 Mar 20;48(3):399-416.

56. Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS (G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019 Nov;575(7781):217-23.

57. Chen X, Calvisi DF. Hydrodynamic transfection for generation of novel mouse models for liver cancer research. The American Journal of Pathology. 2014 Apr 1;184(4):912-23.

58. Fan B, Malato Y, Calvisi DF, Naqvi S, Razumilava N, Ribback S, et al. Cholangiocarcinomas can originate from hepatocytes in mice. The Journal of Clinical Investigation. 2012 Aug 1;122(8):2911-5.

59. Hill MA, Alexander WB, Guo B, Kato Y, Patra K, O'Dell MR, et al. Kras and Tp53 mutations cause cholangiocyte-and hepatocyte-derived cholangiocarcinoma. Cancer Research. 2018 Aug 15;78(16):4445-51.

60. Sekiya S, Suzuki A. Intrahepatic cholangiocarcinoma can arise from Notch-mediated conversion of hepatocytes. The Journal of Clinical Investigation. 2012 Nov 1;122(11):3914-8.

61. Wardell CP, Fujita M, Yamada T, Simbolo M, Fassan M, Karlic R, et al. Genomic characterization of biliary tract cancers identifies driver genes and predisposing mutations. Journal of Hepatology. 2018 May 1;68(5):959-69.

62. Moeini A, Haber PK, Sia D. Cell of origin in biliary tract cancers and clinical implications. JHEP Reports. 2021 Apr 1;3(2):100226.

63. Ignatiadis M, Sledge GW, Jeffrey SS. Liquid biopsy enters the clinic—Implementation issues and future challenges. Nature reviews Clinical Oncology. 2021 May;18(5):297-312.

64. Mansouri S, Heylmann D, Stiewe T, Kracht M, Savai R. Cancer genome and tumor microenvironment: Reciprocal crosstalk shapes lung cancer plasticity. Elife. 2022 Sep 8;11:e79895.

65. Van de Wetering M, Francies HE, Francis JM, Bounova G, Iorio F, Pronk A, et al. Prospective derivation of a living organoid biobank of colorectal cancer patients. Cell. 2015 May 7;161(4):933-45.

66. Boj SF, Hwang CI, Baker LA, Chio II, Engle DD, Corbo V, Jager M, Ponz-Sarvise M, Tiriac H, Spector MS, Gracanin A. Organoid models of human and mouse ductal pancreatic cancer. Cell. 2015 Jan 15;160(1-2):324-38.

67. Yan HH, Siu HC, Law S, Ho SL, Yue SS, Tsui WY, et al. A comprehensive human gastric cancer organoid biobank captures tumor subtype heterogeneity and enables therapeutic screening. Cell Stem Cell. 2018 Dec 6;23(6):882-97.

68. Ooft SN, Weeber F, Dijkstra KK, McLean CM, Kaing S, van Werkhoven E, Schipper L, Hoes L, Vis DJ, van de Haar J, Prevoo W. Patient-derived organoids can predict response to chemotherapy in metastatic colorectal cancer patients. Science Translational Medicine. 2019 Oct 9;11(513):eaay2574.

69. Broutier L, Mastrogiovanni G, Verstegen M, Francies HE, Gavarró LM, Bradshaw CR, et al. Human primary liver cancer–derived organoid cultures for disease modeling and drug screening. Nature Medicine. 2017 Dec;23(12):1424-35.

70. Vlachogiannis G, Hedayat S, Vatsiou A, Jamin Y, Fernández-Mateos J, Khan K, et al. Patient-derived organoids model treatment response of metastatic gastrointestinal cancers. Science. 2018 Feb 23;359(6378):920-6.

71. Sakamoto N, Feng Y, Stolfi C, Kurosu Y, Green M, Lin J, et al. BRAFV600E cooperates with CDX2 inactivation to promote serrated colorectal tumorigenesis. Elife. 2017 Jan 10;6:e20331.

72. Nadauld LD, Garcia S, Natsoulis G, Bell JM, Miotke L, Hopmans ES, et al. Metastatic tumor evolution and organoid modeling implicate TGFBR2as a cancer driver in diffuse gastric cancer. Genome Biology. 2014 Aug;15(8):1-8.

73. Sachs N, de Ligt J, Kopper O, Gogola E, Bounova G, Weeber F, et al. A living biobank of breast cancer organoids captures disease heterogeneity. Cell. 2018 Jan 11;172(1-2):373-86.

74. Li X, Ootani A, Kuo C. An air–liquid interface culture system for 3D organoid culture of diverse primary gastrointestinal tissues. InGastrointestinal Physiology and Diseases 2016 (pp. 33-40). Humana Press, New York, NY.

75. Neal JT, Li X, Zhu J, Giangarra V, Grzeskowiak CL, Ju J, et al. Organoid modeling of the tumor immune microenvironment. Cell. 2018 Dec 13;175(7):1972-88.

76. Dijkstra KK, Cattaneo CM, Weeber F, Chalabi M, van de Haar J, Fanchi LF, Slagter M, van der Velden DL, Kaing S, Kelderman S, van Rooij N. Generation of tumor-reactive T cells by co-culture of peripheral blood lymphocytes and tumor organoids. Cell. 2018 Sep 6;174(6):1586-98.

77. Zhou G, Lieshout R, van Tienderen GS, de Ruiter V, van Royen ME, Boor PP, et al. Modelling immune cytotoxicity for cholangiocarcinoma with tumour-derived organoids and effector T cells. British Journal of Cancer. 2022 May 21:1-2.

Author Information X