You are leaving Medscape Education
Cancel Continue
Log in to save activities Your saved activities will show here so that you can easily access them whenever you're ready. Log in here CME & Education Log in to keep track of your credits.
 

CME / ABIM MOC

Emerging Pathways in Advanced Cholangiocarcinoma

  • Authors: Milind Javle, MD; Tin-Yun Tang, MD; Arndt Vogel, MD; Rachna Shroff, MD, MS
  • CME / ABIM MOC Released: 3/11/2022
  • THIS ACTIVITY HAS EXPIRED
  • Valid for credit through: 3/11/2023
Start Activity


Target Audience and Goal Statement

This activity is intended for oncologists, gastroenterologists, pathologists, and other healthcare providers caring for patients with cholangiocarcinoma (CCA)

The goal of this activity is for learners to better understand biomarker testing in patients with CCA, mechanisms of action (MOAs) of emerging treatment pathways, and the efficacy and safety of new agents.

Upon completion of this activity, participants will:

  • Have increased knowledge regarding the
    • Biomarker expression within CCA
    • MOAs of novel agents in CCA
  • Have greater competence related to
    • Testing for biomarkers within CCA
  • Demonstrate greater confidence in their ability to
    • Understand the side effects of targeted therapies used to manage CCA


Disclosures

Medscape, LLC requires every individual in a position to control educational content to disclose all financial relationships with ineligible companies that have occurred within the past 24 months. Ineligible companies are organizations whose primary business is producing, marketing, selling, re-selling, or distributing healthcare products used by or on patients.

All relevant financial relationships for anyone with the ability to control the content of this educational activity are listed below and have been mitigated according to Medscape policies. Others involved in the planning of this activity have no relevant financial relationships.


Faculty

  • Milind Javle, MD

    Department of Gastrointestinal Medical Oncology
    Division of Cancer Medicine
    The University of Texas
    MD Anderson Cancer Center
    Houston, Texas, United States

    Disclosures

    Disclosure: Milind Javle, MD, has the following relevant financial relationships:
    Advisor or consultant for: AstraZeneca; Bristol Myers Squibb; EMD Serono; Meclun; Taiho; TransThera; QED
    Speaker or a member of a speakers bureau for: Agios; Incyte Corporation; QED
    Grants for clinical research from: Agios; Aslan; AstraZeneca; Basilea; EMD Serono; Eli Lilly; Incyte Corporation; Merck; Novartis; QED
    Other: Steering committee: Incyte Corporation; NuCana; PCI

  • Tin-Yun Tang, MD

    Hematology/Oncology Fellow
    The University of Texas
    MD Anderson Cancer Center
    Houston, Texas, United States

    Disclosures

    Disclosure: Tin-Yun Tang, MD, has the following relevant financial relationships:
    Stocks, stock options, or bonds from: Invitae, Editas, UniQure

  • Arndt Vogel, MD

    Professor
    Medical School Hannover
    Hannover, Germany

    Disclosures

    Disclosure: Arndt Vogel, MD, has the following relevant financial relationships:
    Advisor or consultant for: AstraZeneca; Bayer; Bristol Meyers Squibb; BTG; Eisai; GlaxoSmithKline; Imaging Equipment Ltd (AAA); Incyte Corporation; Ipsen; Lilly; Merck; Merck Sharp & Dohme; Novartis; Pierre Fabre; Roche; Sanofi; Servier; Sirtex; Terumo
    Speaker or a member of a speakers bureau for: AstraZeneca; Bayer; Bristol Myers Squibb; BTG; Eisai; GlaxoSmithKline; Imaging Equipment Ltd (AAA); Incyte Corporation; Ipsen; Lilly; Merck; Merck Sharp & Dohme; Novartis; Pierre Fabre; Roche; Sanofi; Servier; Sirtex; Terumo
    Grants for clinical research from: Servier

  • Rachna Shroff, MD, MS

    Associate Professor, Medicine
    College of Medicine Tucson
    Chief, Section of GI Medical Oncology
    Director, UACC Clinical Trials Office
    Associate Dean, Clinical and Translational Research
    The University of Arizona Health Sciences
    Tucson, Arizona, United States

    Disclosures

    Disclosure: Rachna Shroff, MD, MS, has the following relevant financial relationships:
    Advisor or consultant for: AstraZeneca, Boehringer Ingelheim, Clovis, Genentech, Incyte Corporation, Merck, QED, Servier, Taiho, Zymeworks
    Speaker or a member of a speakers bureau for: Servier
    Grants for clinical research from: Bayer, Bristol Myers Squibb, Exelixis, IMV Inc., LOXO, Novocure, NUCANA, QED, Rafael Pharm, SeaGen Inc., formerly Seattle Genetics, Inc., Taiho

Editors

  • Davecia Ragoonath Cameron, MS

    Medical Education Director, Medscape, LLC

    Disclosures

    Disclosure: Davecia Ragoonath Cameron, MS, has disclosed no relevant financial relationships.

  • Jenny Engelmoer, PhD

    Medical Education Director, Medscape, LLC

    Disclosures

    Disclosure: Jenny Engelmoer, PhD, has disclosed no relevant financial relationships.

Compliance Reviewer

  • Amanda Jett, PharmD, BCACP

    Associate Director, Accreditation and Compliance, Medscape, LLC

    Disclosures

    Amanda Jett, PharmD, BCACP has disclosed no relevant financial relationships.

Peer Reviewer

This activity has been peer reviewed and the reviewer has disclosed no relevant financial relationships.


Accreditation Statements



In support of improving patient care, Medscape, LLC is jointly accredited by the Accreditation Council for Continuing Medical Education (ACCME), the Accreditation Council for Pharmacy Education (ACPE), and the American Nurses Credentialing Center (ANCC), to provide continuing education for the healthcare team.

    For Physicians

  • Medscape, LLC designates this enduring material for a maximum of 1.0 AMA PRA Category 1 Credit(s)™ . Physicians should claim only the credit commensurate with the extent of their participation in the activity.

    Successful completion of this CME activity, which includes participation in the evaluation component, enables the participant to earn up to 1.0 MOC points in the American Board of Internal Medicine's (ABIM) Maintenance of Certification (MOC) program. Participants will earn MOC points equivalent to the amount of CME credits claimed for the activity. It is the CME activity provider's responsibility to submit participant completion information to ACCME for the purpose of granting ABIM MOC credit. Aggregate participant data will be shared with commercial supporters of this activity.

    Contact This Provider

For questions regarding the content of this activity, contact the accredited provider for this CME/CE activity noted above. For technical assistance, contact [email protected]


Instructions for Participation and Credit

There are no fees for participating in or receiving credit for this online educational activity. For information on applicability and acceptance of continuing education credit for this activity, please consult your professional licensing board.

This activity is designed to be completed within the time designated on the title page; physicians should claim only those credits that reflect the time actually spent in the activity. To successfully earn credit, participants must complete the activity online during the valid credit period that is noted on the title page. To receive AMA PRA Category 1 Credit™, you must receive a minimum score of 70% on the post-test.

Follow these steps to earn CME/CE credit*:

  1. Read about the target audience, learning objectives, and author disclosures.
  2. Study the educational content online or print it out.
  3. Online, choose the best answer to each test question. To receive a certificate, you must receive a passing score as designated at the top of the test. We encourage you to complete the Activity Evaluation to provide feedback for future programming.

You may now view or print the certificate from your CME/CE Tracker. You may print the certificate, but you cannot alter it. Credits will be tallied in your CME/CE Tracker and archived for 6 years; at any point within this time period, you can print out the tally as well as the certificates from the CME/CE Tracker.

*The credit that you receive is based on your user profile.

CME / ABIM MOC

Emerging Pathways in Advanced Cholangiocarcinoma

Authors: Milind Javle, MD; Tin-Yun Tang, MD; Arndt Vogel, MD; Rachna Shroff, MD, MSFaculty and Disclosures
THIS ACTIVITY HAS EXPIRED

CME / ABIM MOC Released: 3/11/2022

Valid for credit through: 3/11/2023

processing....

References

  1. Brindley PJ, et al. Cholangiocarcinoma. Nat Rev Dis Primers. 2021;7:65.
  2. Dondossola D, et al. Practical review for diagnosis and clinical management of perihilar cholangiocarcinoma. World J Gastroenterol. 2020;26:3542-3561.
  3. Kirstein MM, et al. Epidemiology and risk factors of cholangiocarcinoma. Visc Med. 2016;32:395-400.
  4. Kendall T, et al. Anatomical, histomorphological and molecular classification of cholangiocarcinoma. Liver Int. 2019;39(suppl 1):7-18.
  5. Valle JW, et al. Biliary cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27(suppl 5):v28-v37.
  6. Vijgen S, et al. Pathology of intrahepatic cholangiocarcinoma. Hepatobiliary Surg Nutr. 2017;6:22-34.
  7. Rizvi S, et al. Emerging technologies for the diagnosis of perihilar cholangiocarcinoma. Semin Liver Dis. 2018;38:160-169.
  8. Rizvi S, et al. Current diagnostic and management options in perihilar cholangiocarcinoma. Digestion. 2014;89:216-224.
  9. Banales JM, et al. Cholangiocarcinoma 2020: the next horizon in mechanisms and management. Nat Rev Gastroenterol Hepatol. 2020;17:557-588.
  10. National Comprehensive Cancer Network (NCCN). NCCN clinical practice guidelines in oncology: hepatobiliary cancers (Version 5.2021). 2021. Accessed February 4, 2022. https://www.nccn.org/guidelines/guidelines-detail?category=1&id=1438
  11. Rodrigues PM, et al. Next-generation biomarkers for cholangiocarcinoma. Cancers (Basel). 2021;13:3222.
  12. Nault JC, et al. Biomarkers for hepatobiliary cancers. Hepatology. 2021;73(Suppl 1):115-127.
  13. Infigratinib [prescribing information]. Approved 2021. Revised May 2021.
  14. Pemigatinib [prescribing information]. Approved 2020. Revised June 2021.
  15. Ivosidenib [prescribing information]. Approved 2018. Revised August 2021.
  16. Mosele F, et al. Recommendations for the use of next-generation sequencing (NGS) for patients with metastatic cancers: a report from the ESMO Precision Medicine Working Group. Ann Oncol. 2020;31:1491-1505.
  17. Lorenzi M, et al. Epidemiology of microsatellite instability high (MSI-H) and deficient mismatch repair (dMMR) in solid tumors: a structured literature review. J Oncol. 2020;2020:1807929.
  18. Ando Y, et al. Low prevalence of biliary tract cancer with defective mismatch repair genes in a Japanese hospital-based population. Oncol Lett. 2022;23:4.
  19. Winkelmann R, et al. Microsatellite instability occurs rarely in patients with cholangiocarcinoma: a retrospective study from a German tertiary care hospital. Int J Mol Sci. 2018;19:1421.
  20. Goeppert B, et al. Mismatch repair deficiency is a rare but putative therapeutically relevant finding in non-liver fluke associated cholangiocarcinoma. Br J Cancer. 2019;120:109-114.
  21. Tshering G, et al. Biomarkers for the diagnosis of cholangiocarcinoma: a systematic review. Am J Trop Med Hyg. 2018;98:1788-1797.
  22. Yi M, et al. Regulation of PD-L1 expression in the tumor microenvironment. J Hematol Oncol. 2021;14:10.
  23. Sangkhamanon S, et al. Programmed death-ligand 1 (PD-L1) expression associated with a high neutrophil/lymphocyte ratio in cholangiocarcinoma. Asian Pac J Cancer Prev. 2017;18:1671-1674.
  24. Malka D, et al. The importance of molecular testing in the treatment of cholangiocarcinoma. EMJ Oncol. 2020;8:82-94.
  25. Bankov K, et al. Sequencing of intraductal biopsies is feasible and potentially impacts clinical management of patients with indeterminate biliary stricture and cholangiocarcinoma. Clin Transl Gastroenterol. 2018;9:151.
  26. Cescon DW, et al. Circulating tumor DNA and liquid biopsy in oncology. Nat Cancer. 2020;1:276-290.
  27. Chakrabarti S, et al. Targeted therapies in advanced biliary tract cancer: an evolving paradigm. Cancers (Basel). 2020;12:2039.
  28. Macias RIR, et al. Diagnostic and prognostic biomarkers in cholangiocarcinoma. Liver Int. 2019;39(Suppl 1):108-122.
  29. Andersen JB, et al. Genomic and genetic characterization of cholangiocarcinoma identifies therapeutic targets for tyrosine kinase inhibitors. Gastroenterology. 2012;142:1021-1031.e15.
  30. Sia D, et al. Integrative molecular analysis of intrahepatic cholangiocarcinoma reveals 2 classes that have different outcomes. Gastroenterology. 2013;144:829-840.
  31. Yue S, et al. FGFR-TKI resistance in cancer: current status and perspectives. J Hematol Oncol. 2021;14:23.
  32. Babina IS, et al. Advances and challenges in targeting FGFR signalling in cancer. Nat Rev Cancer. 2017;17:318-332.
  33. Salati M, et al. IDH signalling pathway in cholangiocarcinoma: from biological rationale to therapeutic targeting. Cancers (Basel). 2020;12:3310.
  34. Iqbal N, et al. Human epidermal growth factor receptor 2 (HER2) in cancers: overexpression and therapeutic implications. Mol Biol Int. 2014;2014:852748.
  35. Galdy S, et al. HER2/HER3 pathway in biliary tract malignancies; systematic review and meta-analysis: a potential therapeutic target? Cancer Metastasis Rev. 2017;36:141-157.
  36. Uprety D, et al. KRAS: from undruggable to a druggable cancer target. Cancer Treat Rev. 2020;89:102070.
  37. Wang J, et al. Targeting the FGFR signaling pathway in cholangiocarcinoma: promise or delusion? Ther Adv Med Oncol. 2020;12:1758835920940948.
  38. Goyal L, et al. Targeting FGFR inhibition in cholangiocarcinoma. Cancer Treat Rev. 2021;95:102170.
  39. Jain A, et al. Molecular profiling of biliary tract cancer: a target rich disease. J Gastrointest Oncol. 2016;7:797-803.
  40. Rizzo A, et al. Targeting BRAF-mutant biliary tract cancer: recent advances and future challenges. Cancer Control. Jan-Dec 2020;27:1073274820983013.
  41. Savoia P, et al. Targeting the ERK signaling pathway in melanoma. Int J Mol Sci. 2019;20:1483.
  42. Chang, F, et al. Signal transduction mediated by the Ras/Raf/MEK/ERK pathway from cytokine receptors to transcription factors: potential targeting for therapeutic intervention. Leukemia. 2003;17:1263-1293.
  43. Turner N, et al. Fibroblast growth factor signalling: from development to cancer. Nat Rev Cancer. 2010;10:116-129.
  44. Liu PCC, 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;15:e0231877.
  45. Abou-Alfa GK, et al. Pemigatinib for previously treated, locally advanced or metastatic cholangiocarcinoma: a multicentre, open-label, phase 2 study. Lancet Oncol. 2020;21:671-684.
  46. Abou-Alfa GK, et al. Pemigatinib for previously treated locally advanced/metastatic cholangiocarcinoma (CCA): update of FIGHT-202. J Clin Oncol. 2021;39(suppl):4086.
  47. Javle M, et al. Infigratinib (BGJ398) in previously treated patients with advanced or metastatic cholangiocarcinoma with FGFR2 fusions or rearrangements: mature results from a multicentre, open-label, single-arm, phase 2 study. Lancet Gastroenterol Hepatol. 2021;6:803-815.
  48. Sootome H, et al. Futibatinib is a novel irreversible FGFR 1-4 inhibitor that shows selective antitumor activity against FGFR-deregulated tumors. Cancer Res. 2020;80:4986-4997.
  49. Goyal L, et al. FOENIX-CCA2: a phase II, open-label, multicenter study of futibatinib in patients (pts) with intrahepatic cholangiocarcinoma (iCCA) harboring FGFR2 gene fusions or other rearrangements. J Clin Oncol. 2020;38(suppl):108.
  50. Goyal L, et al. Primary results of phase 2 FOENIX-CCA2: the irreversible FGFR1-4 inhibitor futibatinib in intrahepatic cholangiocarcinoma (iCCA) with FGFR2 fusions/rearrangements. Cancer Res. 2021;81:13 (Abs CT010).
  51. Braun S, et al. Derazantinib: an investigational drug for the treatment of cholangiocarcinoma. Expert Opin Investig Drugs. 2021;30:1071-1080.
  52. Droz Dit Busset M, et al. Derazantinib for patients with intrahepatic cholangiocarcinoma harboring FGFR2 fusions/rearrangements: primary results from the phase II study FIDES-01. Ann Oncol. 2021;32(suppl 5):Abstract 47P.
  53. Abou-Alfa GK, 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.
  54. Zhu AX, et al. Final overall survival efficacy results of ivosidenib for patients with advanced cholangiocarcinoma with IDH1 mutation: the phase 3 randomized clinical ClarIDHy trial. JAMA Oncol. 2021;7:1669-1677.
  55. Durvalumab [prescribing information]. Approved 2017. Revised July 2021.
  56. ClinicalTrials.gov. Durvalumab or placebo in combination with gemcitabine/cisplatin in patients with 1st line advanced biliary tract cancer (TOPAZ-1). Accessed February 4, 2022. https://clinicaltrials.gov/ct2/show/NCT03875235
  57. Oh D-Y, et al. A phase 3 randomized, double-blind, placebo-controlled study of durvalumab in combination with gemcitabine plus cisplatin (GemCis) n patients with advanced biliary tract cancer (BTC): TOPAZ-1. J Clin Oncol. 2022;39(suppl):3783004.
  58. ClinicalTrials.gov. My Pathway: a study evaluating herceptin/perjeta, tarceva, zelboraf/cotellic, erivedge, alecensa, and tecentriq treatment targeted against certain molecular alterations in participants with advanced solid tumors. Accessed February 4, 2022. https://clinicaltrials.gov/ct2/show/NCT02091141
  59. Meric-Bernstam F, et al. MyPathway HER2 basket study: pertuzumab (P) + trastuzumab (H) treatment of a large, tissue-agnostic cohort of patients with HER2-positive advanced solid tumors. J Clin Oncol. 2021;39(suppl):3004.
  60. Subbiah V, et al. Dabrafenib plus trametinib in patients with BRAF V600E-mutated biliary tract cancer (ROAR): a phase 2, open-label, single-arm, multicentre basket trial. Lancet Oncol. 2020;21:1234-1243.
« Return to: Emerging Pathways in Advanced Cholangiocarcinoma
  • Print