Next Article in Journal
RARβ Expression in Keratinocytes from Potentially Malignant Oral Lesions: The Functional Consequences of Re-Expression by De-Methylating Agents
Next Article in Special Issue
Quality of Life and Symptom Management in Advanced Biliary Tract Cancers
Previous Article in Journal
Role of Chemotherapy in Vulvar Cancers: Time to Rethink Standard of Care?
Previous Article in Special Issue
Novel Pharmacological Options in the Treatment of Cholangiocarcinoma: Mechanisms of Resistance
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Patterns of Whole Exome Sequencing in Resected Cholangiocarcinoma

1
Department of Surgery, City of Hope National Medical Center, Duarte, CA 91010, USA
2
Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
3
Department of Pathology, City of Hope National Medical Center, Duarte, CA 91010, USA
4
Department of Radiation Oncology, City of Hope National Medical Center, Duarte, CA 91010, USA
5
Division of Gastroenterology, City of Hope National Medical Center, Duarte, CA 91010, USA
*
Author to whom correspondence should be addressed.
Cancers 2021, 13(16), 4062; https://doi.org/10.3390/cancers13164062
Submission received: 18 June 2021 / Revised: 29 July 2021 / Accepted: 3 August 2021 / Published: 12 August 2021
(This article belongs to the Special Issue Updates in Biliary Tract Cancers)

Abstract

:

Simple Summary

Cholangiocarcinomas are rare cancers that harbor a significant number of potentially targetable mutations. In this study, we assessed the frequency of genomic profiling for resected cholangiocarcinomas. We found that, over the past decade, a third of patients underwent tumor genomic profiling, among whom 89% harbored a targetable mutation. Mutations were associated with a median of one approved drug. A quarter of eligible sequenced patients were treated with therapy targeting tumor-specific mutations.

Abstract

Background: With minimally effective chemotherapy options, cholangiocarcinoma patients have 5 year survival rate of 10%. Tumor genetic profiling (TGP) can identify mutations susceptible to targeted therapies. We sought to describe the use of TGP and frequency of actionable results in resected cholangiocarcinoma. Methods: A retrospective review of patients undergoing curative intent resection at a comprehensive cancer center (2010–2020). Clinicopathologic and partial or whole exome sequencing data were reviewed. Results: 114 patients (mean age 65 ± 11 years, 45% female) underwent resection of cholangiocarcinoma (46% poorly differentiated, 54% intrahepatic, 36% node positive, 75% margin negative). Additionally, 32% of patients underwent TGP, yielding a mean of 3.1 actionable mutations per patient (range 0–14). Mutations aligned with a median of one drug per patient (range 0–11). Common mutations included TP53 (33%), KRAS (31%), IDH1/2 (14%), FGFR (14%), and BRAF (8%). Targeted therapies were administered in only 4% of patients (23% of eligible sequenced patients). After a median 22 months, 23% had recurrence and 29% were deceased. Discussion: TGP for cholangiocarcinoma has increased over the last decade with targeted therapies identified in most sequenced tumors, impacting treatment in a quarter of eligible patients. Precision medicine will play a central role in the future care of cholangiocarcinoma.

1. Introduction

While cholangiocarcinoma represents only 3% of gastrointestinal malignancies, its incidence is increasing worldwide [1,2,3]. The overall prognosis for patients with cholangiocarcinoma remains poor with a 5 year survival rate of approximately 8–10% [4]. Due to its insidious growth, most cholangiocarcinomas are typically diagnosed at a late stage, and only 30% are eligible for curative intent resection at diagnosis due to vascular involvement or intraperitoneal, lymphatic, or distant spread [2,3]. While surgery remains the only curative option for these patients, rates of post-surgical recurrence are high (>60%) [2,5]. Therefore, surgery alone is unlikely to be a definitive modality for treatment in the future. As more is understood about the biology of these tumors, identifying effective systemic therapies, including chemotherapy, immunotherapy, and targeted therapies, will play a key role in the treatment of nearly all biliary tract cancers [2].
Despite decades of investigation, three recent trials have demonstrated no overall survival benefit to adjuvant cytotoxic systemic therapies for resected cholangiocarcinoma (BCAT trial, PRODIGE 12-ACCORD 18-UNICANCER-GI trial, BILCAP trial) [6,7,8]. Regardless of the fact only limited data exists to support its effectiveness, capecitabine is now established as the new standard of care for adjuvant therapy following curative intent resection for cholangiocarcinoma [9]. While the combination of gemcitabine and cisplatinum is considered the standard for palliative treatment based upon the ABC-02 and BT22 trials [10,11], the search for more effective first-line agents is ongoing [12]. Immunotherapy is currently being explored in biliary tract cancers, particularly in tumors with a high tumor mutational burden [13]; however, mismatch repair deficiency and microsatellite instability in cholangiocarcinoma are uncommon (<5%) [14].
There is a desperate need for new targets for treatment of cholangiocarcinoma [15]. In light of this need, it is noteworthy that cholangiocarcinoma is a mutation-rich cancer with a relatively high frequency of mutations [16]. Some have estimated that two-thirds of intrahepatic cholangiocarcinomas harbor actionable mutations [17]. To date, there have been a number of investigations into targeted therapies in the general population of patients with biliary cancer (HER/ERBB, HGF/c-MET, mTOR, etc.) but most have been largely unsuccessful [18]. Due to the failure of trials of targeted therapies in biologically unselected patients, there is a growing interest in testing such therapies on patients with specific mutations [12]. A precision medicine-based strategy for cholangiocarcinoma is supported by data from the MOSCATO-01 trial, which identified druggable mutations in 68% of patients with advanced cholangiocarcinoma (primarily intrahepatic) and demonstrated a median overall survival benefit of 17 months compared with 5 months among patients who were treated with therapy not matched to any mutation (p = 0.008) [19].
Mutational analysis has been widely available to clinicians for more than a decade as a means of identifying mutations and to connect patients to clinical trials related to their personal mutations [20]. It is not known how clinicians have used these genomic sequence analyses in patients with bile duct cancers or how commonly patients are directed towards targeted therapies matching identified mutations. We hypothesized that the application of genetic sequence analysis has increased over recent years, but that use of targeted therapies in cholangiocarcinoma remains relatively uncommon. We sought to understand patterns of use and mutations among patients treated with surgical intent at a tertiary cancer center.

2. Materials and Methods

We performed a retrospective case series of all patients treated for cholangiocarcinoma via curative intent resection at a tertiary cancer center between 2010 and 2020. Patients were included if they had pathologically confirmed intrahepatic, hilar, or extrahepatic ductal cholangiocarcinoma. Ampullary carcinomas, gallbladder carcinomas, and hepatocellular carcinomas were excluded. Patients were excluded if they never underwent any surgical resection or if surgery was performed with palliative intention only. The cases were reviewed by trained abstractors for inclusion and charts sampled for patient demographics, cholangiocarcinoma risk factors, and tumor features and staging. The patient charts were all sampled for the use of TGP. Genomic sequencing data was collected, including number of genes sequenced, tumor mutational burden (TMB), the number of actionable mutations identified, and the number of drugs associated with those identified mutations. For those genomic sequencing tests that reported such detail, we recorded the number of clinical trials associated with identified mutations. For each identified mutation, we documented the gene and specific codon mutation.
Patient treatment details were collected, including the use of systemic chemotherapy, radiation therapy, and immunotherapies. Surgical resection details included type of resection and margin status. Lastly, any targeted therapies used in treatment—in particular, therapies associated with identified mutations—were documented. We estimated the rate of targeted therapy use among eligible patients, i.e., patients with recurrent or persistent (R1+) disease following surgery who had targetable mutations identified upon sequencing. Patient charts were reviewed for evidence of cancer recurrence and/or death at date of last follow-up. The differences between patients who did and did not undergo genomic sequence testing were reported by a two-sided Student’s t-test (continuous variables) and Chi-squared tests (categorical variables). Log-rank tests were used to estimate differences in overall survival. This study was approved by the Institutional Review Board at City of Hope National Medical Center. All statistical analysis was performed using commercially available software (Stata v16.1, StataCorp LLC, College Station, TX, USA).

3. Results

A total of 114 patients were treated with curative intent resection for cholangiocarcinoma between 2010 and 2020. The mean age was 64.5 years (±11.8 years) and 44.7% were female. The prevalence of cholangiocarcinoma-specific risk factors was relatively low, including viral hepatitis (15.8%), alcohol abuse (4.4%), and primary sclerosing cholangitis (0.9%). Other risk factors reflected the general population, including obesity (22.1%) and diabetes (23.7%). The majority of tumors were intrahepatic (53.5%) with the remainder extrahepatic distal cholangiocarcinomas (31.6%) and hilar (14.9%). Biliary obstruction (total bilirubin > 3) was seen in 40.4% of patients at presentation. Histologic grade was most commonly moderately (45.6%) or poorly differentiated (46.5%). Mean tumor size was 4.6cm (±2.5cm), and a minority were classified as multifocal (14.0%) or bilobar (3.5%). Patient demographics and tumor characteristics of all patients enrolled in this study are summarized in Table 1. AJCC 8th edition T-stage and N-stage, along with pathological stages are also summarized in Table 1. Two patients (1.9%) were designated as pathological stage IV based upon findings of distant disease on final pathology.
In this study, 36 patients (31.6%) with cholangiocarcinoma underwent TGP. Patients who underwent genetic sequencing were significantly younger than those patients who did not undergo tumor sequencing (60.7 years versus 66.3 years, p = 0.02). We did not identify any other differences in demographics or tumor characteristics between sequenced and unsequenced patients. Table 2 summarizes treatment characteristics for all patients in this study. Three-quarters of patients received systemic chemotherapy at some point in their treatment course (72.8%). Radiation therapy was administered to 38.4% of patients in either the neoadjuvant or adjuvant setting. The most common operation was liver resection without bile duct resection (53.5%), and 75.4% of patients were found to have microscopically negative margins (i.e., R0). There were no significant differences in treatment characteristics between those patients who underwent TGP and the remainder of patients. Though most (91%) genetic sequence tests were performed during the mid- (2014–2016) to late-period (2017–2020) of the study, this was not significant (p = 0.06).
Table 3 summarizes the TGP results performed on the tumors of the 36 sequenced patients in this study. The breadth of genes sequenced ranged from small gene panels (e.g., 48 genes) to whole exome sequencing (performed in 12 patients). The tumor mutational burden ranged from less than one to five mutations per megabase pair. A total of 41 unique mutations were identified. A median of two actionable mutations was identified in each sample (range 0–9). Targeted sequencing and whole exome sequencing identified a similar number of mutations (mean 2.9 and 1.8, respectively). Common mutations included 12 TP53 mutations/amplifications (33.3%), 11 KRAS mutations (30.6%), five IDH1/2 mutations (13.9%), and three BRAF V600E mutations (8.3%). Three patients had FGFR alterations (8.3%). Figure 1 displays the gene plot of identified mutations across 36 sequenced tumors. An average of seven clinical trials per patient were identified (range 0–36). A median of one drug per patient was associated with identified mutations (range 0–11). Among 26 eligible patients (i.e., underwent TGP and had an identified target), six received drugs targeting identified mutations in the setting of recurrent or persistent disease (23%, or 4.4% of the entire cohort).
Patients were followed for a median of 22 months (Interquartile range 12–42). At the last follow-up, 42 patients exhibited no evidence of disease (36.8%), while 38 were alive with disease (33.3%), and 34 had died (29.8%). Median overall survival was estimated as 59 months. Patients who underwent genetic sequence testing had a median overall survival of 42 months, while median survival was not reached among patients without TGP (i.e., greater than 50% surviving at last follow-up) [21]. There was no significant difference in the estimates of median survival between groups (p = 0.16). Figure 2 demonstrates the Kaplan Meier curves for survival among patients in this series, including those who underwent TGP and those who did not undergo TGP.

4. Discussion

In this single center case series of resected cholangiocarcinoma over the last decade, we found that a third of patients underwent TGP. Testing was more common among younger patients, which may reflect the trend toward use of TGP in patients who have a poor prognosis yet for whom clinicians are searching for a chance at effective therapy. TGP was also more common during the later years of the 2010s, which reflects the increasing use over time as this technology becomes more widely known and available. Among eligible patients, a quarter were treated with targeted therapies related to mutations identified via TGP.
The TGP reported in this study varied in the extent of genome that was sequenced. Commonly available early-generation tests include panels of 48–50 commonly mutated genes. By comparison, FoundationOne testing (Foundation Medicine, Cambridge, MA, USA) has been commercially available since 2012 and provides a panel of several hundred genes. A third of patients in this study were tested with a FoundationOne assay. Lastly, twelve patients in this study were tested with the GEM ExTra whole exome sequence test (Ashion Analytics LLC, Phoenix, AZ, USA). This assay is designed to detect tumor-specific mutations in both DNA and RNA and, therefore, samples variants not only in the genome of cells within the tumor but also the transcriptome. This assay was provided free-of-charge to patients in this study. In the MOSCATO-01 trial, whole exome sequencing identified additional variants in relevant genes in 38% of patients beyond those identified through targeted gene sequencing [19,22]. In this study, both targeted gene sequencing and whole exome sequencing identified a similar mean number of targetable mutations. It is, nonetheless, expected that whole exome sequencing will play an important role in personalized medicine for a wide variety of solid malignancies.
In this study, a median of one drug per patient was associated with efficacy in identified mutations. However, we found that only a quarter of patients who were eligible, were treated with a targeted therapy. This finding might be explained by a number of possible observations. The first is that, while TGP may have become more commonplace, the general knowledge and perceived utility of targeted agents may still be low among medical oncologists. Second, targeted agents are expensive and are likely not covered by many insurers. This may be a primary reason that these targeted agents used in an “off label” fashion are only accessible to a fraction of patients with associated mutations. The third issue raises the question of timing of administration of targeted therapies. In our practice, we find that the use of TGP in patients with resected cholangiocarcinoma most commonly comes into play in the setting of recurrence. This observation bears out in the assessment of comparative survival between sequenced an unsequenced patients. While no significant difference was found, Figure 2 suggests that those patients who are sequenced have the worst survival. While many patients who develop recurrence following resection will be treated with palliative measures, it remains to be seen whether targeted therapies will be integrated into the first-line in the adjuvant setting. It is not known how mutations vary between principally resected and recurrent tumors. Assays such as circulating tumor DNA for detecting molecular residual disease following resection of cholangiocarcinoma may aid in identifying patients that may benefit from targeted therapies prior to clinically evident recurrence.
The most common mutations were TP53, KRAS, IDH, BRAF, and PIK3CA. Isocitrate dehydrogenase mutations (IDH1/2) are common in intrahepatic cholangiocarcinoma (13%) [23,24]. Mutant IDH blocks liver progenitor cells from undergoing hepatocyte differentiation. In vitro dasantanib treated IDH mutated xenografts demonstrate pronounced apoptosis and tumor regression [25]. In a recent phase III clinical trial for patients with progression on chemotherapy, ivosidinib was associated with significant improvement in progression-free survival compared with placebo (ClarIDHy trial, 2.7 months vs. 1.4 months, HR 0.37, 95% CI 0.25–0.54, p < 0.0001) [23]. One patient in this study with recurrence of previously resected cholangiocarcinoma was found to have an IDH1 R132L mutation and is under treatment with ivosidinib.
Fibroblast growth factor receptors (FGFR) are a family of transmembrane proteins with tyrosine kinase domains, which are commonly mutated or fused in urothelial, breast, and gynecologic cancers, as well as cholangiocarcinomas (commonly FGFR2) [12]. In addition to non-selective tyrosine kinase inhibitors, which have shown activity in cholangiocarcinoma with FGFR2 fusion, a number of phase II trials have demonstrated efficacy for targeted FGFR inhibitors [12]. Selective FGFR kinase inhibitor Infigratinib (BGJ398) showed efficacy in some patients with FGFR2 mutations [26,27]. Among patients receiving Pemigatinib (FGFR inhibitor) in the setting of an FGFR alteration in cholangiocarcinoma, 35.5% achieved an objective response [28]. Futibatinib (TAS-120, highly selective inhibition of FGFR1-4) demonstrated a disease control rate of 79% [29]. Other FGFR-targeting agents have demonstrated lesser effectiveness [12].
In this study, three patients undergoing TGP were found to have fibroblast growth factor-related mutations, but none were treated with associated drugs.
BRAF mutations are present in 5% of cholangiocarcinomas [30]. These patients have a higher stage of tumor at resection, greater likelihood of lymph node involvement, and worse overall survival compared with non-BRAF mutated cholangiocarcinoma. In a basket trial of patients with V600E mutated cancers, BRAF inhibitor (dabrafenib) and MEK inhibitor (trametinib) combination demonstrated a clinical response in 51% of patients [31]. Two of three patients with V600E mutations in this study were treated with BRAF/MEK inhibition.
There are reports of patient response to mTOR inhibition (everolimus) in tumors with PIK3CA mutations. This mutation is found in as many as 8% of cholangiocarcinomas and occurred in 8% of our cohort [32]. None, however, were treated with targeted therapies. Other areas of investigation for targeted therapies in cholangiocarcinoma include human epidermal growth factor receptor 2 (HER2) and RNF2 mutations [12].
While there is great optimism about the future of precision medicine for difficult to treat malignancies, there are a number of limitations to this tool as it pertains to cholangiocarcinoma. Some have argued that the genetic heterogeneity of biliary tract cancers may hamper efforts to elucidate which mutational targets will yield effects for patients [33]. Further, there is potential heterogeneity in the frequency of targetable mutations between intra and extrahepatic lesions; however, this study was not intended to detect such a difference. Lack of a “stereotyped” genetic signature to cholangiocarcinoma may slow development of trials of targeted therapies, further compounded by the relative rarity of this tumor overall. While IDH mutations and FGFR2 fusions have promise for targeted therapies, these represent a small minority of identified mutations [12]. An additional limitation of the use of TGP in cholangiocarcinoma is tissue availability [34]. For patients with a locally advanced disease that is unresectable, targeted therapies might provide a chance at disease control but require additional testing after routine pathologic diagnosis. For patients whose diagnosis is made based upon cytologic specimens such as fine needle aspirate or duct brushings, obtaining cell block material or dedicated fine needle biopsy material reserved for genomic or molecular sequence testing is key. Appropriate stewardship of diagnostic materials will require pre-testing coordination to preclude additional or repeated intervention. Lastly, while targeted genomic testing may cost only a few hundred US dollars, whole exome sequencing often costs 2–5 times more per assay. While this limitation may be prohibitive at present, we take the approach that whole exome-based TGP may provide valuable information for patients about emerging targets and new clinical trials on the immediate horizon.

5. Conclusions

Overall, our study shows that the use of TGP in cholangiocarcinoma has increased over the past decade. While the majority of patients were found to have potentially actionable mutations, this testing resulted in an altered treatment in only a quarter of eligible patients. The findings of this study must be interpreted in the context of a single institution experience. While our care teams had ready access to whole exome sequencing for several years before it was commonly available elsewhere, the application of TGP and choice to prescribe targeted therapies can be expected to vary between providers and institutions. This sample of patients may not represent the natural distribution of mutations seen in the larger population of patients with cholangiocarcinoma. It should also be emphasized that tumors from surgically resected patients in this study may not have the same mutational burden seen in locally advanced or metastatic disease. Furthermore, some patients may have been treated with targeted therapies at other institutions following resection at our quaternary center and, therefore, may not have been captured by this analysis.
Future directions for targeted therapies in cholangiocarcinoma include the continued trial of drugs among patients with unique mutations. To address the issue of limited tissue available for TGP, ‘liquid biopsies’ will likely play a role in identifying tumor-based mutations through circulating tumor DNA [35]. While patients await more effective first-line agents for cholangiocarcinoma, precision methods may play a more active role in up-front therapy for patients with resected tumors [12]. To facilitate this, payors and providers will need to continue the trend of increasing application of TGP in the coming years.

Author Contributions

Conceptualization, L.W.T. and L.G.M.; Methodology, L.W.T. and L.G.M.; Investigation, L.W.T. and L.G.M.; Data curation, L.W.T. and P.W.; Writing—original draft preparation, L.W.T.; Writing—review and editing, L.W.T., P.W., D.L., S.G.W., S.C., M.R., J.K., A.A., J.L., V.C., G.S., Y.F., and L.G.M.; Supervision, L.G.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Institutional Review Board of City of Hope (protocol code 20229, date 3 June 2020).

Informed Consent Statement

Patient consent was waived due to non-identifiable nature of the data categorized as low-risk by the Institutional Review Board.

Data Availability Statement

Data is available on request due to restrictions regarding patient privacy.

Conflicts of Interest

Thornblade: none. Wong: none. Li: Receiving research funding to institution from Brooklyn Immunotherapeutics and AstraZeneca; consulting fees for Advanced Accelerator Applications, Novartis, Bayer, Genentech USA, Taiho Oncology, QED, TerSera, Coherus, Merck, Sun Pharma, Eisai, Exelixis, Ipsen, and Lexicon. Warner: Spouse receives consulting fees for Coloplast, Olympus, and Boston Scientific. Chang: none. Raoof: none. Kessler: none. Amini: Consulting fees AstraZeneca, RefleXion, Takeda Pharmaceuticals. Lin: none. Chung: Consulting fees Pfizer, Ipsen, Coherus, Perthera, AstraZeneca; Research funding Merck. Singh: none. Fong: Serves as a paid scientific advisor to Medtronic, Johnson and Johnson, and Olympus, unrelated to the content of this paper. Melstrom: none.

References

  1. Tyson, G.L.; Ilyas, J.A.; Duan, Z.; Green, L.K.; Younes, M.; El-Serag, H.B.; Davila, J.A. Secular trends in the incidence of cholangiocarcinoma in the USA and the impact of misclassification. Dig. Dis. Sci. 2014, 59, 3103–3110. [Google Scholar] [CrossRef]
  2. Ejaz, A.; Cloyd, J.M.; Pawlik, T.M. Advances in the Diagnosis and Treatment of Patients with Intrahepatic Cholangiocarcinoma. Ann. Surg. Oncol. 2020, 27, 552–560. [Google Scholar] [CrossRef] [PubMed]
  3. Poultsides, G.A.; Zhu, A.X.; Choti, M.A.; Pawlik, T.M. Intrahepatic cholangiocarcinoma. Surg. Clin. N. Am. 2010, 90, 817–837. [Google Scholar] [CrossRef]
  4. Diaz-Gonzalez, A.; Vilana, R.; Bianchi, L.; Garcia-Criado, A.; Rimola, J.; Rodriguez de Lope, C.; Ferrer, J.; Ayuso, C.; Da Fonseca, L.G.; Reig, M.; et al. Thermal Ablation for Intrahepatic Cholangiocarcinoma in Cirrhosis: Safety and Efficacy in Non-Surgical Patients. J. Vasc. Interv. Radiol. 2020, 31, 710–719. [Google Scholar] [CrossRef] [PubMed]
  5. Blechacz, B. Cholangiocarcinoma: Current Knowledge and New Developments. Gut Liver 2017, 11, 13–26. [Google Scholar] [CrossRef] [PubMed]
  6. Ebata, T.; Hirano, S.; Konishi, M.; Uesaka, K.; Tsuchiya, Y.; Ohtsuka, M.; Kaneoka, Y.; Yamamoto, M.; Ambo, Y.; Shimizu, Y.; et al. Randomized clinical trial of adjuvant gemcitabine chemotherapy versus observation in resected bile duct cancer. Br. J. Surg. 2018, 105, 192–202. [Google Scholar] [CrossRef] [PubMed]
  7. Edeline, J.; Benabdelghani, M.; Bertaut, A.; Watelet, J.; Hammel, P.; Joly, J.P.; Boudjema, K.; Fartoux, L.; Bouhier-Leporrier, K.; Jouve, J.L.; et al. Gemcitabine and Oxaliplatin Chemotherapy or Surveillance in Resected Biliary Tract Cancer (PRODIGE 12-ACCORD 18-UNICANCER GI): A Randomized Phase III Study. J. Clin. Oncol. 2019, 37, 658–667. [Google Scholar] [CrossRef]
  8. Primrose, J.N.; Fox, R.P.; Palmer, D.H.; Malik, H.Z.; Prasad, R.; Mirza, D.; Anthony, A.; Corrie, P.; Falk, S.; Finch-Jones, M.; et al. Capecitabine compared with observation in resected biliary tract cancer (BILCAP): A randomised, controlled, multicentre, phase 3 study. Lancet Oncol. 2019, 20, 663–673. [Google Scholar] [CrossRef] [Green Version]
  9. Shroff, R.T.; Kennedy, E.B.; Bachini, M.; Bekaii-Saab, T.; Crane, C.; Edeline, J.; El-Khoueiry, A.; Feng, M.; Katz, M.H.G.; Primrose, J.; et al. Adjuvant Therapy for Resected Biliary Tract Cancer: ASCO Clinical Practice Guideline. J. Clin. Oncol. 2019, 37, 1015–1027. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Valle, J.; Wasan, H.; Palmer, D.H.; Cunningham, D.; Anthoney, A.; Maraveyas, A.; Madhusudan, S.; Iveson, T.; Hughes, S.; Pereira, S.P.; et al. Cisplatin plus gemcitabine versus gemcitabine for biliary tract cancer. N. Engl. J. Med. 2010, 362, 1273–1281. [Google Scholar] [CrossRef] [Green Version]
  11. Okusaka, T.; Nakachi, K.; Fukutomi, A.; Mizuno, N.; Ohkawa, S.; Funakoshi, A.; Nagino, M.; Kondo, S.; Nagaoka, S.; Funai, J.; et al. Gemcitabine alone or in combination with cisplatin in patients with biliary tract cancer: A comparative multicentre study in Japan. Br. J. Cancer 2010, 103, 469–474. [Google Scholar] [CrossRef] [Green Version]
  12. Lamarca, A.; Barriuso, J.; McNamara, M.G.; Valle, J.W. Molecular targeted therapies: Ready for "prime time" in biliary tract cancer. J. Hepatol. 2020, 73, 170–185. [Google Scholar] [CrossRef] [Green Version]
  13. Ueno, M.; Ikeda, M.; Morizane, C.; Kobayashi, S.; Ohno, I.; Kondo, S.; Okano, N.; Kimura, K.; Asada, S.; Namba, Y.; et al. Nivolumab alone or in combination with cisplatin plus gemcitabine in Japanese patients with unresectable or recurrent biliary tract cancer: A non-randomised, multicentre, open-label, phase 1 study. Lancet Gastroenterol. Hepatol. 2019, 4, 611–621. [Google Scholar] [CrossRef]
  14. Marabelle, A.; Le, D.T.; Ascierto, P.A.; Di Giacomo, A.M.; De Jesus-Acosta, A.; Delord, J.P.; Geva, R.; Gottfried, M.; Penel, N.; Hansen, A.R.; 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. [Google Scholar] [CrossRef]
  15. Banales, J.M.; Cardinale, V.; Carpino, G.; Marzioni, M.; Andersen, J.B.; Invernizzi, P.; Lind, G.E.; Folseraas, T.; Forbes, S.J.; Fouassier, L.; et al. Expert consensus document: Cholangiocarcinoma: Current knowledge and future perspectives consensus statement from the European Network for the Study of Cholangiocarcinoma (ENS-CCA). Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 261–280. [Google Scholar] [CrossRef] [PubMed]
  16. Nakamura, H.; Arai, Y.; Totoki, Y.; Shirota, T.; Elzawahry, A.; Kato, M.; Hama, N.; Hosoda, F.; Urushidate, T.; Ohashi, S.; et al. Genomic spectra of biliary tract cancer. Nat. Genet. 2015, 47, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  17. Ross, J.S.; Wang, K.; Gay, L.; Al-Rohil, R.; Rand, J.V.; Jones, D.M.; Lee, H.J.; Sheehan, C.E.; Otto, G.A.; Palmer, G.; et al. New routes to targeted therapy of intrahepatic cholangiocarcinomas revealed by next-generation sequencing. Oncologist 2014, 19, 235–242. [Google Scholar] [CrossRef] [Green Version]
  18. Valle, J.W.; Lamarca, A.; Goyal, L.; Barriuso, J.; Zhu, A.X. New Horizons for Precision Medicine in Biliary Tract Cancers. Cancer Discov. 2017, 7, 943–962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Verlingue, L.; Malka, D.; Allorant, A.; Massard, C.; Ferté, C.; Lacroix, L.; Rouleau, E.; Auger, N.; Ngo, M.; Nicotra, C.; et al. Precision medicine for patients with advanced biliary tract cancers: An effective strategy within the prospective MOSCATO-01 trial. Eur. J. Cancer (Oxf. Engl. 1990) 2017, 87, 122–130. [Google Scholar] [CrossRef]
  20. Simile, M.M.; Bagella, P.; Vidili, G.; Spanu, A.; Manetti, R.; Seddaiu, M.A.; Babudieri, S.; Madeddu, G.; Serra, P.A.; Altana, M.; et al. Targeted Therapies in Cholangiocarcinoma: Emerging Evidence from Clinical Trials. Medicina (Kaunas) 2019, 55, 42. [Google Scholar] [CrossRef] [Green Version]
  21. Thornblade, L.W.; Kessler, J.J.D.M.R. Thermal ablation of intrahepatic cholangiocarcinoma: A narrative review. Dig. Med. Res. 2021, 4. [Google Scholar] [CrossRef]
  22. Koeppel, F.; Bobard, A.; Lefebvre, C.; Pedrero, M.; Deloger, M.; Boursin, Y.; Richon, C.; Chen-Min-Tao, R.; Robert, G.; Meurice, G.; et al. Added Value of Whole-Exome and Transcriptome Sequencing for Clinical Molecular Screenings of Advanced Cancer Patients With Solid Tumors. Cancer J. (Sudbury Mass.) 2018, 24, 153–162. [Google Scholar] [CrossRef]
  23. Abou-Alfa, G.K.; Macarulla, T.; Javle, M.M.; Kelley, R.K.; Lubner, S.J.; Adeva, J.; Cleary, J.M.; Catenacci, D.V.; Borad, M.J.; Bridgewater, J.; et al. Ivosidenib in IDH1-mutant, chemotherapy-refractory cholangiocarcinoma (ClarIDHy): A multicentre, randomised, double-blind, placebo-controlled, phase 3 study. Lancet Oncol. 2020, 21, 796–807. [Google Scholar] [CrossRef]
  24. Banales, J.M.; Marin, J.J.G.; Lamarca, A.; Rodrigues, P.M.; Khan, S.A.; Roberts, L.R.; Cardinale, V.; Carpino, G.; Andersen, J.B.; Braconi, C.; et al. Cholangiocarcinoma 2020: The next horizon in mechanisms and management. Nat. Rev. Gastroenterol. Hepatol. 2020, 17, 557–588. [Google Scholar] [CrossRef] [PubMed]
  25. Saha, S.K.; Gordan, J.D.; Kleinstiver, B.P.; Vu, P.; Najem, M.S.; Yeo, J.C.; Shi, L.; Kato, Y.; Levin, R.S.; Webber, J.T.; et al. Isocitrate Dehydrogenase Mutations Confer Dasatinib Hypersensitivity and SRC Dependence in Intrahepatic Cholangiocarcinoma. Cancer Discov. 2016, 6, 727–739. [Google Scholar] [CrossRef] [Green Version]
  26. Goyal, L.; Saha, S.K.; Liu, L.Y.; Siravegna, G.; Leshchiner, I.; Ahronian, L.G.; Lennerz, J.K.; Vu, P.; Deshpande, V.; Kambadakone, A.; et al. Polyclonal Secondary FGFR2 Mutations Drive Acquired Resistance to FGFR Inhibition in Patients with FGFR2 Fusion-Positive Cholangiocarcinoma. Cancer Discov. 2017, 7, 252–263. [Google Scholar] [CrossRef] [Green Version]
  27. Javle, M.; Lowery, M.; Shroff, R.T.; Weiss, K.H.; Springfeld, C.; Borad, M.J.; Ramanathan, R.K.; Goyal, L.; Sadeghi, S.; Macarulla, T.; et al. Phase II Study of BGJ398 in Patients With FGFR-Altered Advanced Cholangiocarcinoma. J. Clin. Oncol. 2018, 36, 276–282. [Google Scholar] [CrossRef]
  28. Abou-Alfa, G.K.; Sahai, V.; Hollebecque, A.; Vaccaro, G.; Melisi, D.; Al-Rajabi, R.; Paulson, A.S.; Borad, M.J.; Gallinson, D.; Murphy, A.G.; et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: A multicentre, open-label, phase 2 study. Lancet Oncol. 2020, 21, 671–684. [Google Scholar] [CrossRef]
  29. Goyal, L.; Shi, L.; Liu, L.Y.; Fece de la Cruz, F.; Lennerz, J.K.; Raghavan, S.; Leschiner, I.; Elagina, L.; Siravegna, G.; Ng, R.W.S.; et al. TAS-120 Overcomes Resistance to ATP-Competitive FGFR Inhibitors in Patients with FGFR2 Fusion-Positive Intrahepatic Cholangiocarcinoma. Cancer Discov. 2019, 9, 1064–1079. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Javle, M.; Bekaii-Saab, T.; Jain, A.; Wang, Y.; Kelley, R.K.; Wang, K.; Kang, H.C.; Catenacci, D.; Ali, S.; Krishnan, S.; et al. Biliary cancer: Utility of next-generation sequencing for clinical management. Cancer 2016, 122, 3838–3847. [Google Scholar] [CrossRef] [Green Version]
  31. Subbiah, V.; Lassen, U.; Élez, E.; Italiano, A.; Curigliano, G.; Javle, M.; de Braud, F.; Prager, G.W.; Greil, R.; Stein, A.; et al. Dabrafenib plus trametinib in patients with BRAFV600E-mutated biliary tract cancer (ROAR): A phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 2020, 21, 1234–1243. [Google Scholar] [CrossRef]
  32. Bian, J.L.; Wang, M.M.; Tong, E.J.; Sun, J.; Li, M.; Miao, Z.B.; Li, Y.L.; Zhu, B.H.; Xu, J.J. Benefit of everolimus in treatment of an intrahepatic cholangiocarcinoma patient with a PIK3CA mutation. World J. Gastroenterol. 2017, 23, 4311–4316. [Google Scholar] [CrossRef]
  33. Brandi, G.; Farioli, A.; Astolfi, A.; Biasco, G.; Tavolari, S. Genetic heterogeneity in cholangiocarcinoma: A major challenge for targeted therapies. Oncotarget 2015, 6, 14744–14753. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Forner, A.; Vidili, G.; Rengo, M.; Bujanda, L.; Ponz-Sarvisé, M.; Lamarca, A. Clinical presentation, diagnosis and staging of cholangiocarcinoma. Liver Int. Off. J. Int. Assoc. Study Liver 2019, 39 (Suppl. 1), 98–107. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Ettrich, T.J.; Schwerdel, D.; Dolnik, A.; Beuter, F.; Blätte, T.J.; Schmidt, S.A.; Stanescu-Siegmund, N.; Steinacker, J.; Marienfeld, R.; Kleger, A.; et al. Genotyping of circulating tumor DNA in cholangiocarcinoma reveals diagnostic and prognostic information. Sci. Rep. 2019, 9, 13261. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Gene plot of all identified mutations among 36 sequenced cholangiocarcinomas. Red squares represent gene mutations, blue squares represent gene fusion, and yellow squares represent gene loss.
Figure 1. Gene plot of all identified mutations among 36 sequenced cholangiocarcinomas. Red squares represent gene mutations, blue squares represent gene fusion, and yellow squares represent gene loss.
Cancers 13 04062 g001
Figure 2. Kaplan Meier curves for overall survival among patients who underwent tumor genomic profiling (red line) and did not undergo tumor genomic profiling (blue line).
Figure 2. Kaplan Meier curves for overall survival among patients who underwent tumor genomic profiling (red line) and did not undergo tumor genomic profiling (blue line).
Cancers 13 04062 g002
Table 1. Demographics, distribution of cholangiocarcinoma risk factors, clinical characteristics, and pathologic stage of patients with resected cholangiocarcinoma, including those who did and did not receive genomic sequencing of their tumor.
Table 1. Demographics, distribution of cholangiocarcinoma risk factors, clinical characteristics, and pathologic stage of patients with resected cholangiocarcinoma, including those who did and did not receive genomic sequencing of their tumor.
All Patients
n = 114
Patients Receiving Sequencing
n = 36
Patients Not Receiving Sequencing
n = 78
p-Value
Demographics
Mean age (years ± SD) (at diagnosis)64.5 ± 11.860.7 (12.9)66.3 (10.9)0.02
Female, n (%)51 (44.7)17 (47.2)34. (43.6)0.72
Race, n (%)
Asian30 (26.3)9 (25.0)21 (26.9)0.71
Black0 (0)0 (0)0 (0)
Hispanic30 (26.3)8 (22.2)22 (28.2)
Middle Eastern0 (0)0 (0)0 (0)
White54 (47.4)19 (52.3)35 (44.9)
Cholangiocarcinoma risk factors, n (%)
Viral hepatitis18 (15.8)5 (13.9)13 (16.7)0.71
Primary sclerosing cholangitis1 (0.9)1 (2.8)0 (0)0.14
Alcohol abuse5 (4.4)2 (5.6)3 (3.9)0.68
Obese (BMI > 30)25 (22.1)6 (16.7)19 (24.7)0.34
Diabetes27 (23.7)5 (13.9)22 (28.2)0.10
Clinical characteristics, n (%)
Obstructive jaundice at diagnosis46 (40.4)13 (36.1)33 (42.3)0.53
Tumor site 0.69
Intrahepatic, n (%)61 (53.5)21 (58.3)40 (51.3)
Hilar, n (%)17 (14.9)4 (11.1)13 (16.7)
Extrahepatic distal, n (%)36 (31.6)11 (30.6)25 (32.1)
Tumor grade, n (%) 0.73
Well differentiated 9 (7.9)2 (5.6)7 (9.0)
Moderately differentiated52 (45.6)18 (50.0)34 (43.6)
Poorly differentiated53 (46.5)16 (44.4)37 (47.4)
Mean tumor size, cm (SD)4.6 (2.5)4.5 (2.9)4.4 (2.3)0.80
Multifocal disease16 (14.0)8 (22.2)8 (10.3)0.29
Bilobar disease4 (3.5)1 (2.8)3 (3.9)0.77
T-stage 0.40
126 (22.9)8 (22.8)18 (24.3)
240 (36.7)16 (45.7)24 (32.4)
340 (36.7)11 (31.4)29 (39.2)
43 (2.8)0 (0)3 (4.1)
N-stage 0.34
057 (58.2)22 (68.8)35 (53.0)
137 (37.8)9 (28.1)28 (42.4)
24 (4.1)1 (3.1)3 (4.6)
AJCC 8th Edition—Pathological Stage 0.40
IA12 (11.1)3 (8.8)9 (12.2)
IB28 (25.9)7 (20.6)21 (28.4)
IIA25 (23.2)12 (35.3)4 (5.4)
IIB28 (25.9)7 (20.6)21 (28.4)
IIIA10 (9.3)2 (5.9)8 (10.8)
IIIB21 (19.4)7 (20.6)14 (18.9)
IIIC3 (2.8)0 (0)3 (4.1)
IV2 (1.9)0 (0)2 (2.7)
Table 2. Therapies and follow-up for patients with resected cholangiocarcinoma.
Table 2. Therapies and follow-up for patients with resected cholangiocarcinoma.
All Patients
n = 114
Patients Receiving Sequencing
n = 36
Patients Not Receiving Sequencing
n = 78
p-Value
Systemic chemotherapy83 (72.8)28 (77.8)55 (70.5)0.42
Radiation therapy43 (38.4)15 (41.7)28 (36.8)0.62
Surgery 0.64
Liver resection without bile duct resection61 (53.5)21 (58.3)40 (51.3)
Resection requiring bile duct resection/reconstruction21 (18.4)7 (19.4)14 (18.0)
Pancreaticoduodenectomy32 (28.1)8 (22.2)24 (30.8)
Margin status 0.32
R086 (75.4)26 (72.2)60 (76.9)
R125 (21.9)10 (27.8)15 (19.2)
R23 (2.6)0 (0)3 (3.9)
Period of treatment 0.06
Early (2010–2013)25 (21.9)3 (8.3)22 (28.2)
Mid (2014–2016)41 (36.0)16 (44.4)25 (32.1)
Late (2017–2020)48 (42.1)17 (47.2)31 (39.7)
Median overall survival—months (25%, 75%)59 (25, NR)42 (25, NR)NR (26, NR)0.16
Table 3. Summary of patients undergoing molecular pathological assessment, identified mutations, associated drugs and trials, therapy received, and patient survival.
Table 3. Summary of patients undergoing molecular pathological assessment, identified mutations, associated drugs and trials, therapy received, and patient survival.
Genes Assessed
(WE: Whole Exome)
Actionable Mutations IdentifiedTumor Mutational Burden (per MBP)Identified Mutations/AlterationsDrugs Associated with Identified MutationsTrials IdentifiedTherapy ReceivedSurvival (Months)
* AWD, † NED
at Last Follow-up
1.3242-KRAS G12S
LRP1B D478N
KRAS
Cobimetinib
Trametinib
8Gemcitabine/Cisplatinum
5-FU
FOLFOX
Pembrolizumab
53
2.480--00Gemcitabine/Cisplatinum68
3.3242-KRAS G12V
TP53 R248Q
KRAS
Trametinib
6Gemcitabine/Cisplatinum
Capecitabine
21
4.WE11NRAS Q61R011Gemcitabine/Cisplatinum
FOLFOX
76 *
5.3242-BRAF V600E
CDKN2A/B
BRAF
Dabrafenib
Regorafenib
Trametinib
Vemurafenib
10Gemcitabine/Cisplatinum
Capecitabine
Dabrafenib/Trametinib
31
6.502-TP53 R213
NRAS G12D
MEK
Trametinib
mTOR
Everolimus
Temsirolimus
5Gemcitabine/Cisplatinum13
7.3246-ARID1A E1763 & Q372
NF2 447
PTPN11 G503V CDKN2A
EP300
NF2
Everolimus
Lapatinib
Temsirolimus
Trametinib
PTPN11
Trametinib
10Sorafenib
Pembrolizumab
45
8.WE41KRAS G12V
TP53 S125G
CDK6 amplification
CDKN2A L16
CDK6
Abemaciclib
Palbociclib
Ribociclib
28-68 *
9.3242-BRAF V600E
CDKN2A loss
BRAF
Cobimetinib
Dabrafenib
Regorafenib
Trametinib
Vemurafenib
4Gemcitabine/Cisplatinum59 *
10.3245-ARID1A Y1719
IDH2 R172W
TP53 Y220C
BAP1 123-1
LRP1B R295
None1-8 *
11.3246-PIK3CA M1004I
TP53 C141W
ATRX A419V
GATA6 amplification MCL1 amplification U2AF1 S34F
PIK3CA
Everolimus
Temsirolimus
4Gemcitabine/Cisplatinum
Pembrolizumab
53 *
12.32475ERBB2 amplification
KRAS amplification
MET amplification
CDKN2A p16INK4a & p14ARF
TET2 R1572W
ERBB2
Afatinib
Lapatinib
Neratinib
Pertuzumab
Traztuzumab
Ado-traztuzumab
Traztuzumab-dkst
KRAS
Cobimetinib
Trametinib
MET
Cabozantinib
Crizotinib
19Gemcitabine/Cisplatinum
FOFOX
Capecitabine
35 *
13.3241-IDH1None0Gemcitabine/Cisplatinum
CAPOX
36
14.-2-KRAS
TP53
None0Gemcitabine/Cisplatinum
FOLFOX
Atezolizumab
Cobimetinib
42
15.491-PIK3CA E542KNone0-10 *
16.481-KRAS G12VNone0Gemcitabine/Cisplatinum
FOLFIRI
Capecitabine
25
17.WE0<1-None0Gemcitabine/Cisplatinum
FOLFOX
5-FU
41 *
18.324 & 4853EGFR A289V
BAP1 V616
TP53 V173M
MET amplification
MET
Crizotinib
Cabozantinib
EGFR
Pantitumumab
Osimertinib
Lapatinib
Gefitinib
Erlotinib
Cetuximab
Afatinib
10Gemcitabine/Cisplatinum
FOLFOX
FOLFIRI
11
19.15915IDH1 R132LRRM1 positive
Gemcitabine
4Gemcitabine/Cisplatinum
Capecitabine
43 *
20.15915BAP1 E278None0Gemcitabine/Paclitaxel
FOLFOX
FOLFIRI
Sorafenib
30
21.483-KRAS G12D
TP53 H193R
SMAD4 R361C
None0-11
22.WE1<1TP53 V216GNone1Gemcitabine/Cisplatinum33 *
23.3243-KRAS G12V
TP53 A276D
KDM6A P334
KRAS
Cobimetinib
Trametinib
0-18 *
24.32494PIK3CA E545K
AKT1 W80R
FGFR2 C382R
CDKN2A/B loss
SMAD4 R361C & R361H
TP53 R213
AKT1 & PIK3CA
Everolimus
Temsirolimus
FGFR2
Pazopanib
Ponatinib
19Gemcitabine/Cisplatinum
Capecitabine
12
25.3242-BRAF V600E
TBX3 942-1G
BRAF
Cobimetinib
Dabrafenib
Trametinib
Regorafenib
Vemurafenib
9Gemcitabine
Capecitabine
Pembrolizumab
Dabrafenib/Trametinib
Lenvatinib
25
26.WE32CCND1 amplification
TERT C124C4
TSC1 Y48
CCND1
Abemaciclib
Palbociclib
Ribociclib
TSC1
Everolimus
Temsirolimus
17Pembrolizumab14 *
27.492-KRAS G13D
GNAS R201H
None36Gemcitabine/Cisplatinum
Capecitabine
CAPOX
25 *
28.WE21KRAS G12D
PIK3CA H1047R
PIK3CA
Copanlisib
Everolimus
Temsirolimus
10Gemcitabine/Cisplatinum
Capecitabine
22 *
29.WE1<1BAP1 F15_T1604Gemcitabine/Cisplatinum17
30.16800MDM4 amplification
NOTCH2 amplification
FAM48C amplification
PDGFRA amplification
KIT amplification
HIST2H3D amplification
HIST2H3C amplification
MCL1 amplification
IL10 amplification
FGFR3 amplification
WHSC1 gain
FGFR2 loss
NFKB1A loss
FGFR3 TACC3
00Gemcitabine
Floxuridine
49
31.WE31CDKN2A H83N
FGF3 amplification
FGF4 amplification
FGF3
Sorafenib
FGF4
Pazopanib
Sorafenib
10Gemcitabine/Cisplatinum
Capecitabine
15
32.WE1<1GNAS R844CNone6-7
33.WE11IDH1 R132CIDH1
Ivosidinib
0Capecitabine3 *
34.484-TP53 P152
ARID2 F1537
GNAS amplification
ZNF217 amplificaiton
None6Gemcitabine/Cisplatinum
5-FU
Pembrolizumab
36
35.WE0<1-None0Gemcitabine/Cisplatinum22
36.WE4<1KRAS G12D
IDH1 R132L
KDM5C G1452
MDM2 amplification
IDH1
Ivosidinib
KDM5C
Sunitinib
15Gemcitabine/Cisplatinum
Ivosidinib
26 *
WE: Whole exome. The * stands for “alive with disease” and the † stands for “No evidence of disease”.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Thornblade, L.W.; Wong, P.; Li, D.; Warner, S.G.; Chang, S.; Raoof, M.; Kessler, J.; Amini, A.; Lin, J.; Chung, V.; et al. Patterns of Whole Exome Sequencing in Resected Cholangiocarcinoma. Cancers 2021, 13, 4062. https://doi.org/10.3390/cancers13164062

AMA Style

Thornblade LW, Wong P, Li D, Warner SG, Chang S, Raoof M, Kessler J, Amini A, Lin J, Chung V, et al. Patterns of Whole Exome Sequencing in Resected Cholangiocarcinoma. Cancers. 2021; 13(16):4062. https://doi.org/10.3390/cancers13164062

Chicago/Turabian Style

Thornblade, Lucas W., Paul Wong, Daneng Li, Susanne G. Warner, Sue Chang, Mustafa Raoof, Jonathan Kessler, Arya Amini, James Lin, Vincent Chung, and et al. 2021. "Patterns of Whole Exome Sequencing in Resected Cholangiocarcinoma" Cancers 13, no. 16: 4062. https://doi.org/10.3390/cancers13164062

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop