Precision Oncology for Hepatocellular Cancer: Slivering the Liver by FGF19–FGFR4–KLB Pathway Inhibition Vivek Subbiah 1 and Sumanta K

Total Page:16

File Type:pdf, Size:1020Kb

Precision Oncology for Hepatocellular Cancer: Slivering the Liver by FGF19–FGFR4–KLB Pathway Inhibition Vivek Subbiah 1 and Sumanta K VIEWS IN THE SPOTLIGHT Precision Oncology for Hepatocellular Cancer: Slivering the Liver by FGF19–FGFR4–KLB Pathway Inhibition Vivek Subbiah 1 and Sumanta K. Pal 2 Summary: This issue reports two studies, one by Hatlen and colleagues and the other by Kim and colleagues, that detail the drug-development journey of the FGF19–FGFR4 inhibitor fi sogatinib (BLU-554), from identifi cation of the drug to preclinical validation studies to fi nally the results of the proof-of-concept fi rst-in-human phase I trial of this potent and selective, type I irreversible inhibitor of FGFR4. Moreover, Hatlen and colleagues also report a resistance mechanism acquired after therapy that targets selective FGFR4 inhibition, which validates FGF as a specifi c target in hepatocellular cancer. See related article by Hatlen et al., p. 1686 (9). See related article by Kim et al., p. 1696 (8) . O sliver of liver, Get lost! Go away! in HCC. In fact, the four nonselective VEGF-based multikinase You tremble and quiver, O sliver of liver inhibitors (sorafenib, regorafenib, lenvatinib, and cabozan- –“A Sliver of Liver ” tinib) that have been approved by the FDA in HCC are in use Myra Cohn Livingston, 1926–1996 without a biomarker to guide this therapy selection. Further- more, understanding of the response and resistance mecha- The identifi cation of oncogenic activation of tyrosine nisms to these multikinase inhibitors has yet to be elucidated. kinases, such as mutations in the EGFR or BRAF genes, rear- Immune checkpoint inhibitors indicated for HCC confer very rangements of the NTRK genes, and mutations/rearrange- modest activity, and more than ten nonspecifi c, phase III tri- ments of RET, has enabled the development of targeted als in all patients with HCC have failed to meet the primary treatments for such pathway-aberrant cancers. However, such end-point, warranting an urgent biomarker-driven therapy ( 2 ). advances have had a limited impact on patients with hepato- cellular carcinoma (HCC), the cancer with the second high- est mortality in the world, with 700,000 deaths worldwide CHALLENGES OF DRUG DEVELOPMENT recorded every year. Patients with HCC have yet to derive IN THE FGFR PATHWAY meaningful durable benefi t from selective biomarker-driven targeted therapy in this era of precision oncology ( 1 ). One The family of FGFs regulates multiple biological processes, major challenge in HCC has been absence of “druggable” including cell proliferation, migration, differentiation, apop- alterations; that is, genes reported to be frequently mutated tosis, metabolism, and angiogenesis. Consequently, aberrant in HCC such as the TERT promoter, TP53 , and CTNNB1 activation of FGFR signaling has been implicated in several (β-catenin), have all remained formidable as targets with no malignancies. The FGFR tyrosine kinase family consists of effective drug ( 1 ). Additional aberrations identifi ed through four members, FGFR1–4, which are activated through 22 the efforts of The Cancer Genome Atlas include LZTR1, different FGF ligands, highlighting the complexities, both EEF1A1, SF3B1, SMARCA4, ALB, APOB , and CPS1 , in addition known and unknown, of this pathway, as evidenced by the to well-characterized driver oncogenes such as CCND1, FGF19 variable success of more than 20 compounds ( Table 1 ) that (11q13.3 ), MYC (8q24.21), MET (7q31.2), VEGFA (6p21.1), and have been in development to target FGFR ( 3–6 ). Several FGFR MCL1 (1q21.3; ref. 1 ). This wide variability in aberrations inhibitors that have entered early-phase trials have been lim- combined with the lack of effective drugs targeting them have ited by their nonselectivity and side effects from pan-FGFR resulted in a dearth of biomarker-driven drug therapy options inhibition, mainly from the on-target toxicity of hyperphos- phatemia, leading to frequent treatment interruptions and dose reductions and thereby loss of effi cacy ( 4 ). Indeed, in the 1 Department Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, landmark study of the FGFR inhibitor erdafi tinib (Balversa) Texas. 2 Department of Medical Oncology & Experimental Therapeutics, in FGFR3-mutant bladder cancer, nearly half of the patients City of Hope Comprehensive Cancer Center, Duarte, California. had treatment-related adverse events of grade 3 or higher; Corrected online December 10, 2019. nonetheless, given the confi rmed response rate of 40% with Corresponding Author: Vivek Subbiah, The University of Texas MD Ander- a medial overall survival of 13.8 months, erdafi tinib became son Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030. Phone: the fi rst-ever biomarker-based drug to be approved in blad- 713-563-0393; Fax: 713-792-0334; E-mail: [email protected] der cancer and the fi rst-in-class FGFR inhibitor to receive Cancer Discov 2019;9:1646–9 approval ( 7 ). On the basis of observed toxicities, most FGFR doi: 10.1158/2159-8290.CD-19-1156 inhibitor trials recommend mitigation strategies for these ©2019 American Association for Cancer Research. electrolyte imbalances, such as maintaining a low-phosphate 1646 | CANCER DISCOVERY DECEMBER 2019 www.aacrjournals.org Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. VIEWS Table 1. Small-molecule FGFR inhibitors in clinical development and their measured in vitro IC50 values compared with BLU-554 (fi sogatinib) FGF4 measured IC50 Measured IC50 Inhibitor (nmol/L, in vitro ) (nmol/L, in vitro ) Company JNJ-42756493 (erdafi tinib, Balversa) a 5.7 FGFR1: 1.2 Janssen FGFR2: 2.5 FGFR3: 3 BLU-554 (fi sogatinib) 5 FGFR1: 624 Blueprint Medicines FGFR2: 1202 FGFR3: 2203 BLU-9931 3 FGFR1: 591 Blueprint Medicines FGFR2: 493 FGFR3: 150 H3B-6527 <1.2 FGFR1: 320 H3 Biomedicine, Eisai Incorporation FGFR2: 1290 FGFR3: 1060 PRN1371 19.3 FGFR1: 0.6 Principia Biopharma FGFR2: 1.3 FGFR3: 4.1 TAS-120 (futibatinib) 8.3 FGFR1: 3.9 Taiho Oncology FGFR2: 1.3 FGFR3: 1.6 INCB054828 (pemigatinib) 30 FGFR1: 0.4 Incyte Pharmaceuticals FGFR2: 0.5 FGFR3: 1 AP24534 (ponatinib) 7.7 FGFR1: 2.2 Ariad Pharmaceuticals FGFR2: 1.6 FGFR3: 18.2 ARQ 087 (derazantinib) 34 FGFR1: 4.5 ArQule FGFR2: 1.8 FGFR3: 4.5 BIBF 1120 (nintedanib) 610 FGFR1: 69 Boehringer Ingelheim FGFR2: 37 FGFR3: 108 PD166866 N/A FGFR1: 52.4 Pfi zer PD173074 N/A FGFR1: 22–25 Pfi zer FGFR3: 29 NVP-FGF401 1.1 N/A Novartis NVP-BGJ398 (infi gratinib) 60 FGFR1: 0.9 Novartis FGFR2: 1.4 FGFR3: 1 Dovitinib (CHIR258, TKI258) N/A FGFR1: 8 Novartis FGFR3: 9 LYS2874455 6 FGFR1: 2.8 Eli Lilly FGFR2: 2.6 FGFR3: 6.4 AZD4547 165 FGFR1: 0.2 AstraZeneca FGFR2: 2.5 FGFR3: 1.8 Debio-1347 (CH5183284) 290 FGFR1: 9.3 Debiopharm International FGFR2: 7.6 FGFR3: 22 BAY1163877 (rogaratinib) 33 FGFR1: 12–15 Bayer FGFR2: <1 FGFR3: 19 E-3810 (lucitanib) >1,000 FGFR1: 17.5 Clovis Oncology FGFR2: 82.5 FGFR3: 237.5 aBalversa is the fi rst FGFR kinase inhibitor approved by the FDA. Balversa (erdafi tinib) received accelerated approval from the FDA for the treatment of adults with locally advanced or metastatic urothelial carcinoma that has susceptible FGFR3 or FGFR2 genetic alterations, and who have progressed during or following at least one line of prior platinum-containing chemotherapy, including within 12 months of neoadjuvant or adjuvant platinum- containing chemotherapy. DECEMBER 2019 CANCER DISCOVERY | 1647 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2019 American Association for Cancer Research. VIEWS diet that excludes products like cheese, ice cream, chocolate, ACQUIRED RESISTANCE PARADIGM IN fast foods, colas, yogurt, and meats, which likely can have FGFR4, AND NEXT STEPS major implications for most patients’ way of life and, one can argue, quality of life in those with advanced cancer. In the The initial clinical benefit seen with select targeted quest for FGFR selectivity, and to limit off-target toxicities therapies in oncology is often diminished by the eventual, such as those associated with concurrent VEGFR2 inhibition, seemingly inevitable development of acquired resistance. several observations were made—drugs that selectively target Therefore, as critical as it is to identify a novel drug to target FGFR without VEGFR2 demonstrated a loss of FGFR4 path- an aberration, so too is the almost immediate effort to iden- way inhibition. This finding presented a new need to develop tify the emerging mechanisms of resistance and to formulate a selective FGFR4 inhibitor so that the lack of VEGFR2 activ- a therapeutic strategy to overcome them in real time. In their ity does not affect the clinical outcomes regarding disease study, Hatlen and colleagues studied patients who devel- control, especially in FGFR4-driven cancers. oped disease progression on fisogatinib, identified muta- This issue of Cancer Discovery presents critical findings on tions in the gatekeeper (V550) and hinge-1 (C552) residues the FGF19–FGFR4 inhibitor fisogatinib (BLU-554), a potent of FGFR4 that confer resistance to fisogatinib in the clinical and selective, type I irreversible inhibitor of FGFR4. First, Kim setting, and validated those mechanisms of resistance in vivo and colleagues (8) report the identification, preclinical valida- and in vitro. Of the 7 patients who had imaging evidence of tion, and complete clinical data from the first-in-human response per RECIST, 2 (29%) showed evidence of on-target phase I trial of fisogatinib. Then, Hatlen and colleagues (9) resistance. Hatlen and colleagues went on to demonstrate report their findings on the novel acquired mechanisms of that LY2874455, a gatekeeper-agnostic FGFR inhibitor, over- resistance identified within the kinase domain of FGFR4 comes this resistance to fisogatinib in bothin vitro and in vivo in two patients on the aforementioned phase I trial who models. This finding further validates the theory that can- progressed on fisogatinib. Furthermore, they go on to dem- cers with acquired resistance to selective FGFR4 inhibitors onstrate in vitro evidence of overcoming this resistance by still maintain the FGF19–FGFR4–KLB pathway dependency.
Recommended publications
  • FGF19 Protects Hepatocellular Carcinoma Cells Against Endoplasmic Reticulum Stress Via Activation of FGFR4-Gsk3β-Nrf2 Signaling
    Author Manuscript Published OnlineFirst on September 26, 2017; DOI: 10.1158/0008-5472.CAN-17-2039 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. FGF19 protects hepatocellular carcinoma cells against endoplasmic reticulum stress via activation of FGFR4-GSK3β-Nrf2 signaling Yong Teng1,2#*, Huakan Zhao3,4#, Lixia Gao1, Wenfa Zhang3, Austin Y Shull5, Chloe Shay6 1. Department of Oral Biology, Augusta University, Augusta, GA, USA 2. Georgia Cancer Center, Augusta University, Augusta, GA, USA 3. School of Life Sciences, Chongqing University, Chongqing, China 4. Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China 5. Department of Biology, Presbyterian College, Clinton, SC, USA 6. Emory Children’s Center, Emory University, Atlanta, GA, USA # These authors contributed equally to this work Running Title: Role of FGF19 in ER stress Key Words: FGF19, FGFR4, Nrf2, HCC, ER stress, anticancer Abbreviations list: AARE: amino-acid-response element; ANOVA: analysis of variance; ARE: antioxidant response elements; ATF4: activating transcription factor 4; ChIP-qPCR: chromatin immunoprecipitation quantitative-PCR; CV: cyclic voltammetry; DMSO: dimethyl sulfoxide; EMT: epithelial-mesenchymal transition; ER: endoplasmic reticulum; FGF19: fibroblast growth factor 19; FGFR4: FGF receptor 4; GSK3β: glycogen synthase kinase •− 3β; HCC: hepatocellular carcinoma; Nrf2: nuclear factor E2-related factor 2; O2 : superoxide; PBS: phosphate-buffered saline; ROS: reactive free
    [Show full text]
  • ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are The
    0031-3998/07/6103-0267 PEDIATRIC RESEARCH Vol. 61, No. 3, 2007 Copyright © 2007 International Pediatric Research Foundation, Inc. Printed in U.S.A. ARTICLES Fibroblast Growth Factors 1, 2, 17, and 19 Are the Predominant FGF Ligands Expressed in Human Fetal Growth Plate Cartilage PAVEL KREJCI, DEBORAH KRAKOW, PERTCHOUI B. MEKIKIAN, AND WILLIAM R. WILCOX Medical Genetics Institute [P.K., D.K., P.B.M., W.R.W.], Cedars-Sinai Medical Center, Los Angeles, California 90048; Department of Obstetrics and Gynecology [D.K.] and Department of Pediatrics [W.R.W.], UCLA School of Medicine, Los Angeles, California 90095 ABSTRACT: Fibroblast growth factors (FGF) regulate bone growth, (G380R) or TD (K650E) mutations (4–6). When expressed at but their expression in human cartilage is unclear. Here, we deter- physiologic levels, FGFR3-G380R required, like its wild-type mined the expression of entire FGF family in human fetal growth counterpart, ligand for activation (7). Similarly, in vitro cul- plate cartilage. Using reverse transcriptase PCR, the transcripts for tivated human TD chondrocytes as well as chondrocytes FGF1, 2, 5, 8–14, 16–19, and 21 were found. However, only FGF1, isolated from Fgfr3-K644M mice had an identical time course 2, 17, and 19 were detectable at the protein level. By immunohisto- of Fgfr3 activation compared with wild-type chondrocytes and chemistry, FGF17 and 19 were uniformly expressed within the showed no receptor activation in the absence of ligand (8,9). growth plate. In contrast, FGF1 was found only in proliferating and hypertrophic chondrocytes whereas FGF2 localized predominantly to Despite the importance of the FGF ligand for activation of the resting and proliferating cartilage.
    [Show full text]
  • Up-Regulation of Fibroblast Growth Factor 19 and Its Receptor Associates with Progression from Fatty Liver to Hepatocellular Carcinoma
    www.impactjournals.com/oncotarget/ Oncotarget, Vol. 7, No. 32 Research Paper Up-regulation of fibroblast growth factor 19 and its receptor associates with progression from fatty liver to hepatocellular carcinoma Yan Li1,*, Weizhong Zhang2,*, Anne Doughtie1, Guozhen Cui3, Xuanyi Li1, Harshul Pandit1, Yingbin Yang1, Suping Li1, Robert Martin1 1Division of Surgical Oncology, Department of Surgery, School of Medicine, University of Louisville, Louisville, KY, 40202, USA 2Department of Hand Surgery, China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130022, China 3Department of Hepatology, Cancer Center, The First Hospital of Jilin University, Changchun, 130021, China *These authors contributed equally to this work Correspondence to: Yan Li, email: [email protected] Robert Martin, email: [email protected] Keywords: hepatocellular carcinoma, FGF19, FGFR4, cancer stem cell Received: February 08, 2016 Accepted: June 12, 2016 Published: July 21, 2016 ABSTRACT Background: Human fibroblast growth factor 19 (FGF19), its receptor (FGFR4) and EpCAM play an important role in cell proliferation, differentiation, motility, and overexpression have been linked to hepatocellular carcinoma (HCC). The aim of this study was to evaluate the FGF19 signals responsible for the progression of HCC arising from fatty liver. Results: FGF19 level was significantly increased in the HCC patients’ serum compared to non-HCC controls. The IHC results demonstrated significant increases of protein expressions of FGF19, FGFR4 and EpCAM in specimens with fatty liver, NASH, cirrhosis, and HCC compared to healthy liver tissue. There was a significant positive correlation between the protein expressions (FGF19, FGFR4, and EpCAM) and histopathologic changes from FL to HCC. Furthermore, FGF19 was positively correlated with FGFR4 and with EpCAM.
    [Show full text]
  • FGF21 Acts As a Negative Regulator of Bile Acid Synthesis
    237 2 Journal of M M Chen, C Hale et al. FGF21 negative regulator of bile 237:2 139–152 Endocrinology acid metabolism RESEARCH FGF21 acts as a negative regulator of bile acid synthesis Michelle M Chen*, Clarence Hale*, Shanaka Stanislaus, Jing Xu and Murielle M Véniant Department of Cardiometabolic Disorders, Amgen Inc., Thousand Oaks, California, USA Correspondence should be addressed to M M Véniant: [email protected] *(M M Chen and C Hale contributed equally to this work) Abstract Fibroblast growth factor 21 (FGF21) is a potent regulator of glucose and lipid Key Words homeostasis in vivo; its most closely related subfamily member, FGF19, is known to be a f FGF21 critical negative regulator of bile acid synthesis. To delineate whether FGF21 also plays a f Fc-fusion protein functional role in bile acid metabolism, we evaluated the effects of short- and long-term f β-klotho binding exposure to native FGF21 and long-acting FGF21 analogs on hepatic signal transduction, f bile acid gene expression and enterohepatic bile acid levels in primary hepatocytes and in rodent and monkey models. FGF21 acutely induced ERK phosphorylation and inhibited Cyp7A1 mRNA expression in primary hepatocytes and in different rodent models, although less potently than recombinant human FGF19. Long-term administration of FGF21 in mice fed a standard chow diet resulted in a 50–60% decrease in bile acid levels in the liver and small intestines and consequently a 60% reduction of bile acid pool size. In parallel, colonic and fecal bile acid was decreased, whereas fecal cholesterol and fatty acid excretions were elevated.
    [Show full text]
  • FGF Signaling Network in the Gastrointestinal Tract (Review)
    163-168 1/6/06 16:12 Page 163 INTERNATIONAL JOURNAL OF ONCOLOGY 29: 163-168, 2006 163 FGF signaling network in the gastrointestinal tract (Review) MASUKO KATOH1 and MASARU KATOH2 1M&M Medical BioInformatics, Hongo 113-0033; 2Genetics and Cell Biology Section, National Cancer Center Research Institute, Tokyo 104-0045, Japan Received March 29, 2006; Accepted May 2, 2006 Abstract. Fibroblast growth factor (FGF) signals are trans- Contents duced through FGF receptors (FGFRs) and FRS2/FRS3- SHP2 (PTPN11)-GRB2 docking protein complex to SOS- 1. Introduction RAS-RAF-MAPKK-MAPK signaling cascade and GAB1/ 2. FGF family GAB2-PI3K-PDK-AKT/aPKC signaling cascade. The RAS~ 3. Regulation of FGF signaling by WNT MAPK signaling cascade is implicated in cell growth and 4. FGF signaling network in the stomach differentiation, the PI3K~AKT signaling cascade in cell 5. FGF signaling network in the colon survival and cell fate determination, and the PI3K~aPKC 6. Clinical application of FGF signaling cascade in cell polarity control. FGF18, FGF20 and 7. Clinical application of FGF signaling inhibitors SPRY4 are potent targets of the canonical WNT signaling 8. Perspectives pathway in the gastrointestinal tract. SPRY4 is the FGF signaling inhibitor functioning as negative feedback apparatus for the WNT/FGF-dependent epithelial proliferation. 1. Introduction Recombinant FGF7 and FGF20 proteins are applicable for treatment of chemotherapy/radiation-induced mucosal injury, Fibroblast growth factor (FGF) family proteins play key roles while recombinant FGF2 protein and FGF4 expression vector in growth and survival of stem cells during embryogenesis, are applicable for therapeutic angiogenesis. Helicobacter tissues regeneration, and carcinogenesis (1-4).
    [Show full text]
  • Leukemia Insights June 2021
    JUNE 2021 In this month’s Leukemia Insights newsletter, written by Prithviraj Bose, M.D. and Srdan Verstovsek, M.D., Ph.D., and sponsored in part by the Charif Souki Cancer Research ABOUT MyMDAnderson Fund, we discuss our novel therapeutic approaches for the rare hematologic malignancies, systemic mastocytosis and myeloid/lymphoid neoplasms with myMDAnderson is a secure, eosinophilia. Learn more about our Leukemia program. personalized web site helping community physicians expedite patient referrals, as well as improve continuity Spotlight on rare, atypical, myeloid of care through information access and streamlined communications. neoplasms: systemic mastocytosis and Physicians who have referred patients myeloid/lymphoid neoplasms with to MD Anderson or plan to do so, can utilize the HIPAA compliant features eosinophilia and FGFR1 rearrangements of myMDAnderson to: • Refer a patient • View your patient's appointments Systemic Mastocytosis Access patient reports • Send and receive secure Systemic mastocytosis (SM) is a rare myeloid neoplasm messages driven in approximately 95% of cases by an activating mutation in c-KIT, usually D816V. SM is characterized as indolent, smoldering or advanced, based on the presence JOIN THE COVERSATION and number of so-called B- and C-findings. The latter Connect with us. signify organ damage and are a hallmark of advanced SM (AdvSM). AdvSM, in turn, is typically sub-classified as aggressive SM (ASM), SM with an associated hematologic neoplasm (SM-AHN, the most common subtype) and mast cell leukemia (MCL). Patients with indolent SM (ISM) and smoldering SM (SSM) enjoy much better survival than JOIN OUR MAILING LIST those with AdvSM, although symptoms in all three subtypes can be severe and debilitating.
    [Show full text]
  • Incyte Announces the European Commission Approval of Pemazyre
    3/29/2021 Incyte Announces the European Commission Approval of Pemazyre® (pemigatinib) as a Treatment for Adults with Locally Advanced or Metastatic Cholangiocarcinoma with a Fibroblast Growth Factor Receptor 2 (FGFR2) Fusion or Rearrangement - Pemazyre is the rst targeted therapy approved in the EU for this indication WILMINGTON, Del.--(BUSINESS WIRE)-- Incyte (Nasdaq:INCY) today announced that the European Commission (EC) has approved Pemazyre® (pemigatinib) for the treatment of adults with locally advanced or metastatic cholangiocarcinoma with a broblast growth factor receptor 2 (FGFR2) fusion or rearrangement that have progressed after at least one prior line of systemic therapy. The decision follows the positive opinion received from the European Medicines Agency’s Committee for Medicinal Products for Human Use in January 2021 recommending the conditional marketing authorization of Pemazyre. “Pemazyre’s approval is a crucial milestone for patients with FGFR2 positive cholangiocarcinoma. It is the rst new treatment option to be made available to these patients in the EU in over a decade and has demonstrated a high rate of durable responses in a setting where historically there has been no eective standard of care,” said Hervé Hoppenot, Chief Executive Ocer, Incyte. “We now look forward to working with individual countries in Europe to ensure eligible patients can access this new treatment as soon as possible.” The EC decision is based on data from the FIGHT-202 study evaluating the safety and ecacy of Pemazyre in adult patients with previously treated, locally advanced or metastatic cholangiocarcinoma with documented FGF/FGFR status. Interim results from FIGHT-202 demonstrated that in patients harboring FGFR2 fusions or rearrangements (Cohort A [108 patients]), Pemazyre monotherapy resulted in an overall response rate (ORR) of 37 percent (primary endpoint) and a median duration of response (DOR) of 8 months (secondary endpoint) based on an independent central radiographic review.
    [Show full text]
  • The Biology of Hepatocellular Carcinoma: Implications for Genomic and Immune Therapies Galina Khemlina1,4*, Sadakatsu Ikeda2,3 and Razelle Kurzrock2
    Khemlina et al. Molecular Cancer (2017) 16:149 DOI 10.1186/s12943-017-0712-x REVIEW Open Access The biology of Hepatocellular carcinoma: implications for genomic and immune therapies Galina Khemlina1,4*, Sadakatsu Ikeda2,3 and Razelle Kurzrock2 Abstract Hepatocellular carcinoma (HCC), the most common type of primary liver cancer, is a leading cause of cancer-related death worldwide. It is highly refractory to most systemic therapies. Recently, significant progress has been made in uncovering genomic alterations in HCC, including potentially targetable aberrations. The most common molecular anomalies in this malignancy are mutations in the TERT promoter, TP53, CTNNB1, AXIN1, ARID1A, CDKN2A and CCND1 genes. PTEN loss at the protein level is also frequent. Genomic portfolios stratify by risk factors as follows: (i) CTNNB1 with alcoholic cirrhosis; and (ii) TP53 with hepatitis B virus-induced cirrhosis. Activating mutations in CTNNB1 and inactivating mutations in AXIN1 both activate WNT signaling. Alterations in this pathway, as well as in TP53 and the cell cycle machinery, and in the PI3K/Akt/mTor axis (the latter activated in the presence of PTEN loss), as well as aberrant angiogenesis and epigenetic anomalies, appear to be major events in HCC. Many of these abnormalities may be pharmacologically tractable. Immunotherapy with checkpoint inhibitors is also emerging as an important treatment option. Indeed, 82% of patients express PD-L1 (immunohistochemistry) and response rates to anti-PD-1 treatment are about 19%, and include about 5% complete remissions as well as durable benefit in some patients. Biomarker-matched trials are still limited in this disease, and many of the genomic alterations in HCC remain challenging to target.
    [Show full text]
  • Targeting FXR and FGF19 to Treat Metabolic Diseases—Lessons
    1720 Diabetes Volume 67, September 2018 Targeting FXR and FGF19 to Treat Metabolic Diseases— Lessons Learned From Bariatric Surgery Nadejda Bozadjieva,1 Kristy M. Heppner,2 and Randy J. Seeley1 Diabetes 2018;67:1720–1728 | https://doi.org/10.2337/dbi17-0007 Bariatric surgery procedures, such as Roux-en-Y gastric diabetes (T2D) (1). Clinical data demonstrate that patients bypass (RYGB) and vertical sleeve gastrectomy (VSG), who have undergone RYGB or VSG experience increased are the most effective interventions available for sus- satiety and major glycemic improvements prior to significant tained weight loss and improved glucose metabolism. weight loss, suggesting that metabolic changes as result Bariatric surgery alters the enterohepatic bile acid cir- of these surgeries are essential to the weight loss and glycemic culation, resulting in increased plasma bile levels as well benefits (2). Therefore, it is important to identify the medi- as altered bile acid composition. While it remains unclear ators that play a role in promoting the benefits of these why both VSG and RYGB can alter bile acids, it is possible surgeries with the goal of improving current surgical that these changes are important mediators of the approaches and developing less invasive therapies that effects of surgery. Moreover, a molecular target of bile harness these effects. acid synthesis, the bile acid–activated transcription fac- The effectiveness of bariatric surgery to reduce body tor FXR, is essential for the positive effects of VSG on weight and improve glucose metabolism highlights the weight loss and glycemic control. This Perspective examines the relationship and sequence of events be- important role the gastrointestinal tract plays in regulat- tween altered bile acid levels and composition, FXR ing a wide range of metabolic processes.
    [Show full text]
  • The Roles of Fgfs in the Early Development of Vertebrate Limbs
    Downloaded from genesdev.cshlp.org on September 26, 2021 - Published by Cold Spring Harbor Laboratory Press REVIEW The roles of FGFs in the early development of vertebrate limbs Gail R. Martin1 Department of Anatomy and Program in Developmental Biology, School of Medicine, University of California at San Francisco, San Francisco, California 94143–0452 USA ‘‘Fibroblast growth factor’’ (FGF) was first identified 25 tion of two closely related proteins—acidic FGF and ba- years ago as a mitogenic activity in pituitary extracts sic FGF (now designated FGF1 and FGF2, respectively). (Armelin 1973; Gospodarowicz 1974). This modest ob- With the advent of gene isolation techniques it became servation subsequently led to the identification of a large apparent that the Fgf1 and Fgf2 genes are members of a family of proteins that affect cell proliferation, differen- large family, now known to be comprised of at least 17 tiation, survival, and motility (for review, see Basilico genes, Fgf1–Fgf17, in mammals (see Coulier et al. 1997; and Moscatelli 1992; Baird 1994). Recently, evidence has McWhirter et al. 1997; Hoshikawa et al. 1998; Miyake been accumulating that specific members of the FGF 1998). At least five of these genes are expressed in the family function as key intercellular signaling molecules developing limb (see Table 1). The proteins encoded by in embryogenesis (for review, see Goldfarb 1996). Indeed, the 17 different FGF genes range from 155 to 268 amino it may be no exaggeration to say that, in conjunction acid residues in length, and each contains a conserved with the members of a small number of other signaling ‘‘core’’ sequence of ∼120 amino acids that confers a com- molecule families [including WNT (Parr and McMahon mon tertiary structure and the ability to bind heparin or 1994), Hedgehog (HH) (Hammerschmidt et al.
    [Show full text]
  • Differential Specificity of Endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in Complex with KLB
    Differential Specificity of Endocrine FGF19 and FGF21 to FGFR1 and FGFR4 in Complex with KLB Chaofeng Yang1, Chengliu Jin1, Xiaokun Li3, Fen Wang1, Wallace L. McKeehan1,2, Yongde Luo1,2* 1 Center for Cancer and Stem Cell Biology, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America, 2 IBT Proteomics and Nanotechnology Laboratory, Institute of Biosciences and Technology, Texas A&M Health Science Center, Houston, Texas, United States of America, 3 School of Pharmacy, Wenzhou Medical College, Wenzhou, China Abstract Background: Recent studies suggest that betaKlotho (KLB) and endocrine FGF19 and FGF21 redirect FGFR signaling to regulation of metabolic homeostasis and suppression of obesity and diabetes. However, the identity of the predominant metabolic tissue in which a major FGFR-KLB resides that critically mediates the differential actions and metabolism effects of FGF19 and FGF21 remain unclear. Methodology/Principal Findings: We determined the receptor and tissue specificity of FGF21 in comparison to FGF19 by using direct, sensitive and quantitative binding kinetics, and downstream signal transduction and expression of early response gene upon administration of FGF19 and FGF21 in mice. We found that FGF21 binds FGFR1 with much higher affinity than FGFR4 in presence of KLB; while FGF19 binds both FGFR1 and FGFR4 in presence of KLB with comparable affinity. The interaction of FGF21 with FGFR4-KLB is very weak even at high concentration and could be negligible at physiological concentration. Both FGF19 and FGF21 but not FGF1 exhibit binding affinity to KLB. The binding of FGF1 is dependent on where FGFRs are present.
    [Show full text]
  • Clinicogenomic Analysis of FGFR2-Rearranged Cholangiocarcinoma Identifies Correlates of Response and Mechanisms of Resistance to Pemigatinib
    Published OnlineFirst November 20, 2020; DOI: 10.1158/2159-8290.CD-20-0766 RESEARCH ARTICLE Clinicogenomic Analysis of FGFR2- Rearranged Cholangiocarcinoma Identifies Correlates of Response and Mechanisms of Resistance to Pemigatinib Ian M. Silverman1, Antoine Hollebecque2, Luc Friboulet2, Sherry Owens1, Robert C. Newton1, Huiling Zhen3, Luis Féliz4, Camilla Zecchetto5, Davide Melisi5, and Timothy C. Burn1 Downloaded from cancerdiscovery.aacrjournals.org on September 30, 2021. © 2020 American Association for Cancer Research. Published OnlineFirst November 20, 2020; DOI: 10.1158/2159-8290.CD-20-0766 ABSTRACT Pemigatinib, a selective FGFR1–3 inhibitor, has demonstrated antitumor activity in FIGHT-202, a phase II study in patients with cholangiocarcinoma harboring FGFR2 fusions/rearrangements, and has gained regulatory approval in the United States. Eligibility for FIGHT- 202 was assessed using genomic profiling; here, these data were utilized to characterize the genomic landscape of cholangiocarcinoma and to uncover unique molecular features of patients harboring FGFR2 rearrangements. The results highlight the high percentage of patients with cholangiocarcinoma harboring potentially actionable genomic alterations and the diversity in gene partners that rear- range with FGFR2. Clinicogenomic analysis of pemigatinib-treated patients identified mechanisms of primary and acquired resistance. Genomic subsets of patients with other potentially actionable FGF/ FGFR alterations were also identified. Our study provides a framework for molecularly guided clinical trials and underscores the importance of genomic profiling to enable a deeper understanding of the molecular basis for response and nonresponse to targeted therapy. SIGNIFICANCE: We utilized genomic profiling data from FIGHT-202 to gain insights into the genomic landscape of cholangiocarcinoma, to understand the molecular diversity of patients with FGFR2 fusions or rearrangements, and to interrogate the clinicogenomics of patients treated with pemi- gatinib.
    [Show full text]