Panelists Discuss Emerging Therapies in BTCs

December 2022, Vol 3, No 4

Standing: Milind Javle, MD;
Seated: left to right: Rachna T. Shroff, MD, MS; Jennifer Knox, MSc, MD, FRCPC; Flavio G. Rocha, MD, FACS, FSSP; Richard Kim, MD; Mark Yarchoan, MD

Milind Javle, MD, of the MD Anderson Cancer Center, led off the roundtable dialogue on emerging therapies in biliary tract cancers (BTCs) with a discussion around methylthioadenosine phosphorylase (MTAP) loss in cholangiocarcinoma (CCA). MTAP is located on the 9p21 chromosome, where CDKN2A/B is also located. CDKN2A/B loss occurs in 19% to 27% of BTCs and is almost always associated with subsequent MTAP loss. Although CDKN2A is not a druggable target, it has been hypothesized that MTAP can be targeted using protein arginine methyltransferase 5 (PRMT5) inhibitors. MTAP enzyme loss is associated with an accumulation of methylthioadenosine substrate, which in turn inhibits PRMT5, a key methylation enzyme in the S-adenosyl methionine (SAM) cycle; therefore, PRMT5 inhibitors may be a future therapeutic approach in BTC. The folate cycle also plays an important role in the regeneration of the SAM pathway; it has been hypothesized that MTAP loss can be targeted using antifolate agents such as pemetrexed. Resistance to immune checkpoint inhibitors (ICIs) may also be of concern in MTAP loss tumors because 9p21 loss results in a “cold” immune microenvironment, which may not be responsive to immunotherapy. Intrahepatic cholangiocarcinoma (iCCA) with MTAP loss also results in reduced CD3+, CD4+, CD8+, and inducible co-stimulator T-cells, and MTAP loss and SMAD4 mutations have been associated with low PD-L1 positivity. Although CDKN2A/B is not targetable, MTAP loss is a potential therapeutic target with several therapies currently under investigation.

Rachna Shroff, MD, MS, of the University of Arizona Cancer Center, discussed MDM2 as a potential molecular target. MDM2 works hand in hand with p53, and overexpression of MDM2 leads to p53 degradation and tumor proliferation. Although MDM2 amplifications in BTC are rare, an incidence in iCCA of approximately 6% has been seen and suggests MDM2 is a negative prognostic marker in BTC1; one study demonstrated significantly shorter overall survival in patients with MDM2 amplifications compared with those without this amplification.2 BI 907828 is in development as a potent MDM2/p53 antagonist and has been shown to have a dual mode of action with direct tumor targeting plus immunomodulation in preclinical models.3 NCT03449381 is a phase 1 study evaluating BI 907828 monotherapy in patients with advanced solid tumors; cohort 2 included patients with BTC with p53 wild-type and MDM2-amplified tumors. Early data showed that 84% of patients across all dose levels achieved at least stable disease and 2 of 4 patients with BTC achieved a confirmed partial response.4 A phase 2 multi-cohort basket trial is planned for patients with MDM2-amplified and p53 wild-type tumors, which will also include a BTC cohort.

Jennifer Knox, MSc, MD, FRCPC, of the Princess Margaret Cancer Centre in Toronto, discussed targeting KRAS, which she deemed “the holy grail of undruggable genomic drivers.” The first drugs to target KRAS were mutant-specific and targeted G12C; however, there has been increasing excitement around targets for other alleles as well as pan-(K)RAS inhibitors in clinical and preclinical development.5 Dr. Knox detailed the pros and cons of pan versus mutant- or allele-selective KRAS targeting. Allele-specific KRAS drugs are better tolerated, are able to combine with other agents, have a more sustained target inhibition, and can be used in early treatment.5 However, these therapies are restricted to patients with specific mutant variants, are prone to on-target resistance, and the feasibility of developing allele-selective drugs beyond G12C/D and G13C is a concern. Pan-KRAS drugs can be used in a broader population, in cancers driven by KRAS wild type, and in heterogeneous cancers driven by multiple KRAS aberrations and can overcome on-target KRAS resistance; however, the toxic effects on normal tissues when inhibiting wild-type KRAS in humans have yet to be determined.5 The frequency of KRAS in BTC has been previously reported as anywhere from 12% to 42%, with very few reports describing specific KRAS variants. There is a need for continued research into relevant targets, and both allele-specific and pan-KRAS targeting techniques have been a focus of ongoing research and development.

Mark Yarchoan, MD, from Johns Hopkins Medicine, discussed novel immunotherapies in BTC. CCA is an immunosuppressed tumor type characterized by low tumor-mutation burden and PD-L1 expression.6 With results from TOPAZ-1, there is now evidence that iCCA can benefit from immunotherapy.7 In this regard, there are a number of novel ICIs currently being investigated in CCA, including antagonists of T-cell immunoglobulin and mucin domain 3 (TIM3), lymphocyte activation gene-3 (LAG-3), and T-cell immunoreceptor with Ig and ITIM domains (TIGIT). Data from other tumor types may help develop a rationale for using other ICIs in CCA. For example, nivolumab plus a LAG-3 inhibitor (relatlimab) showed a small benefit over nivolumab alone in metastatic melanoma.8 However, there may be a declining benefit of consecutive ICI use; a subgroup of patients with melanoma who received nivolumab plus relatlimab eventually progressed and then received nivolumab plus ipilimumab, but demonstrated almost no response, although first-line treatment with nivolumab plus ipilimumab has been shown to result in a 50% objective response ratio (ORR) in these patients.8

To convert CCA into an immune-sensitive tumor, a variety of components need to be considered: there needs to be induction of a T-cell response, reversal of immune deficits, and modulation of the tumor microenvironment (TME). Adoptive cell therapies have been used in other tumor types to initiate the immune cascade, which suggests a possible approach in CCA. Finally, it has been shown that there are 4 distinct, molecularly defined subsets of CCA, categorized into 4 clusters, in which the TME differs, suggesting the need for an individualized approach to immunotherapy in CCA.

Finally, Richard Kim, MD, from the Moffitt Cancer Center, discussed a novel approach with dual blockade of vascular endothelial growth factor (VEGF) and delta-like protein-4 (DLL4). DLL4 plays an important role in angiogenesis, and animal studies have shown that DLL4 is controlled by VEGF and that DLL4/Notch signal transduction supports VEGF function. It has been hypothesized that joint inhibition of the 2 pathways may be synergistic. Currently, 3 bispecific antibodies are under development that utilize this strategy, with CTX-009 being the most advanced. Interim phase 2 data of CTX-009 in combination with paclitaxel in BTC demonstrated an ORR of 42%.9 Based on this single-arm study, there is a phase 2/3 trial of CTX-009 plus paclitaxel versus paclitaxel alone in patients with unresectable advanced metastatic BTC to confirm the efficacy of this combination.

References

  1. Pu X, Zhu L, Li F, et al. Target molecular treatment markers in intrahepatic cholangiocarcinoma based on Chinese population. Pathol Res Pract. 2020;216(9):153116.
  2. Kim SJ, Akita M, Sung Y-N, et al. MDM2 amplification in intrahepatic cholangiocarcinomas: its relationship with large-duct type morphology and uncommon KRAS mutations. Am J Surg Pathol. 2018;42(4):512-521.
  3. Rudolph D, Reschke M, Blake S, et al. BI 907828: a novel, potent MDM2 inhibitor that induces antitumor immunologic memory and acts synergistically with an antiPD-1 antibody in syngeneic mouse models of cancer. Cancer Res. 2018;78:4866.
  4. Gounder MM, Yamamoto N, Patel MR, et al. A phase Ia/Ib, dose-escalation/expansion study of the MDM2–p53 antagonist BI 907828 in patients with solid tumors, including advanced/metastatic liposarcoma (LPS). J Clin Oncol. 2022;40(16):3004-3004.
  5. Hofmann MH, Gerlach D, Misale S, et al. Expanding the reach of precision oncology by drugging all KRAS mutants. Cancer Discov. 2022;12(4):924-937.
  6. Yarchoan M, Albacker LA, Hopkis AC, et al. PD-L1 expression and tumor mutational burden are independent biomarkers in most cancers. JCI Insight. 2019;4(6):e126908.
  7. Oh D-Y, He AR, Qin S, et al. A phase 3 randomized, double-blind, placebo-controlled study of durvalumab in combination with gemcitabine plus cisplatin (GemCis) in patients (pts) with advanced biliary tract cancer (BTC): TOPAZ-1. J Clin Oncol. 2022;40(4):378.
  8. Menzies AM, da Silva IP, Trojaniello C, et al. CTLA-4 blockade resistance after relatlimab and nivolumab. N Engl J Med. 2022;386(17):1668-1669.
  9. Compass Therapeutics reports positive interim phase 2 data of CTX-009 in combination with paclitaxel in biliary tract cancers. News release. May 4, 2022. Accessed Nov 30, 2022. https://bit.ly/3Fmlgcq.

Related Items

Surgical Approaches to CCA, Part 1: Integration with Medical Therapy
December 2022, Vol 3, No 4
A multidisciplinary approach to treating localized disease in cholangiocarcinoma (CCA) is important, and management strategies for advanced disease have the potential to be used in patients who undergo surgical resection.
Surgical Approaches to CCA, Part 2: Innovations in Surgical Techniques
December 2022, Vol 3, No 4
Improving early return to function and minimizing complications are important considerations for patients with resectable cholangiocarcinoma (CCA), but data on minimally invasive liver surgery have been limited to single or multi-institutional retrospective studies. Of note, 2 randomized studies have shown that laparoscopic removal of colorectal liver metastases improved outcomes in complication rates, length of stay, and pain burden without compromising margin status compared with open surgery.
Surgical Approaches to CCA, Part 3: Liver Transplant
December 2022, Vol 3, No 4
Historically, liver transplantation in patients with cholangiocarcinoma (CCA) was considered a contraindication at most US medical centers; however, with recent advances, the United Network for Organ Sharing (UNOS) liver intestine committee now recognizes that transplantation is indicated in hilar CCA when performed under institutional protocols with neoadjuvant chemoradiation. Maria Majella Doyle, MD, MBA, of Washington University School of Medicine and Mid America Transplant Services, discussed liver transplant in her presentation, detailing protocols and efficacy results from the Mayo Clinic and other centers in hilar CCA and beyond.
Advances in Medical Therapy of CCA, Part 2: Treatment Challenges
December 2022, Vol 3, No 4
Systemic control has always been the mainstay in the treatment of advanced biliary tract cancer (BTC), with several first- and second-line therapies approved by the US Food and Drug Administration.
Keynote: Crossfire Debates on the Management of CCA
December 2022, Vol 3, No 4
Teresa Macarulla, MD, PhD, of the Vall d’Hebron University Hospital in Barcelona, Spain, and Mark Yarchoan, MD, from Johns Hopkins Medicine, debated the question, “Should immunotherapy be standard of care in cholangiocarcinoma (CCA)?”
Supportive Care and Management of Complications in BTC: A Multidisciplinary Team Approach
December 2022, Vol 3, No 4
Mark Schattner, MD, FASGE, AGAF, of Memorial Sloan Kettering Cancer Center, discussed endoscopic biliary drainage (EBD) in cholangiocarcinoma (CCA), specifically in hilar obstruction, emphasizing the different goals and techniques in patients with resectable versus unresectable disease. The ultimate goals for management of obstruction in patients with resectable CCA are to optimize them for surgery and avoid perioperative complications and unnecessary procedures.

Subscribe Today!

To sign up for our newsletter or print publications, please enter your contact information below.

I'd like to receive: