1
|
Rasool W, Alami Idrissi Y, Ahmad O, Rath S, Saeed F, Sayed MS, Sharma B, Ibrahim AA, Kumari A, Ullah I, Saeed A. Safety and efficacy of pemigatinib in patients with cholangiocarcinoma: a systematic review. J Gastrointest Oncol 2025; 16:699-710. [PMID: 40386584 PMCID: PMC12078806 DOI: 10.21037/jgo-2024-923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 02/19/2025] [Indexed: 05/20/2025] Open
Abstract
Background Cholangiocarcinoma (CCA) is an aggressive bile duct cancer with limited therapeutic options and poor prognosis. pemigatinib, a selective FGFR inhibitor, has emerged as a promising targeted therapy for CCA patients harboring FGFR2 fusions or rearrangements. This systematic review evaluated the safety and efficacy of pemigatinib in this patient population. Methods A comprehensive systematic review was conducted across PubMed, Scopus, Embase, Cochrane Library, and Web of Science to identify studies investigating pemigatinib in CCA patients. Five studies involving a total of 459 patients met the inclusion criteria. Results Pemigatinib demonstrated an overall objective response rate (ORR) of 43.2%, with a complete response (CR) achieved in 3% of patients. Stable disease was observed in 36.9% of patients, while 14.9% experienced disease progression. Median progression-free survival (PFS) varied across studies, due to differences in patient cohorts. The most common adverse effects (AEs) included hyperphosphatemia (48%), diarrhea (28.6%), fatigue (33%), and dry eyes (20.1%). Conclusions This systematic review suggests that pemigatinib has modest therapeutic efficacy in CCA patients, with a considerable proportion achieving disease control. However, the ORR of less than 50% highlights the potential need for combination or sequential therapies to improve outcomes. Close monitoring and management of AEs, particularly hyperphosphatemia, are crucial for optimizing treatment. Further large-scale randomized trials and research are warranted to identify predictive biomarkers and optimize pemigatinib-based treatment strategies for CCA patients with FGFR2 alterations.
Collapse
Affiliation(s)
- Warda Rasool
- Faculty of Medicine, King Edward Medical University, Lahore, Pakistan
| | - Yassine Alami Idrissi
- University of Pittsburgh Medical Center (UPMC) & UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Owais Ahmad
- Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| | - Shree Rath
- All India Institute of Medical Sciences, Bhubaneswar, India
| | - Fatima Saeed
- United Medical and Dental College, Karachi, Pakistan
| | | | - Bhavya Sharma
- Baroda Medical College and SSG Hospital, Vadodara, India
| | - Arwa Amer Ibrahim
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Ajanta Kumari
- Dow University of Health Sciences, Karachi, Pakistan
| | - Irfan Ullah
- Department of Internal Medicine, Khyber Teaching Hospital, Peshawar, Pakistan
| | - Anwaar Saeed
- University of Pittsburgh Medical Center (UPMC) & UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| |
Collapse
|
2
|
Chaturantabut S, Oliver S, Frederick DT, Kim JJ, Robinson FP, Sinopoli A, Song TY, He Y, Chang YC, Rodriguez DJ, Chang L, Kesar D, Ching M, Dzvurumi R, Atari A, Tseng YY, Bardeesy N, Sellers WR. Identification of potent biparatopic antibodies targeting FGFR2 fusion-driven cholangiocarcinoma. J Clin Invest 2025; 135:e182417. [PMID: 40014401 PMCID: PMC11996885 DOI: 10.1172/jci182417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 02/11/2025] [Indexed: 03/01/2025] Open
Abstract
Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, response rates and durability are limited due to the emergence of resistance, typically involving FGFR2 kinase domain mutations, and to suboptimal dosing, relating to drug adverse effects. Here, we develop biparatopic antibodies targeting the FGFR2 extracellular domain (ECD) as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of bivalent monospecific antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies targeting distinct epitope pairs in FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2 fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, we believe that biparatopic antibodies may serve as an innovative treatment option for patients with FGFR2-altered cholangiocarcinoma.
Collapse
MESH Headings
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/immunology
- Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/drug therapy
- Cholangiocarcinoma/immunology
- Cholangiocarcinoma/pathology
- Humans
- Mice
- Animals
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/drug therapy
- Bile Duct Neoplasms/immunology
- Bile Duct Neoplasms/pathology
- Oncogene Proteins, Fusion/immunology
- Oncogene Proteins, Fusion/genetics
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Cell Line, Tumor
Collapse
Affiliation(s)
- Saireudee Chaturantabut
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Faculty of Pharmacy, Silpakorn University, Nakhon Pathom, Thailand
| | - Sydney Oliver
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Jiwan J. Kim
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Foxy P. Robinson
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Tian-Yu Song
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Yao He
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yuan-Chen Chang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | | | - Liang Chang
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Devishi Kesar
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Meilani Ching
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Ruvimbo Dzvurumi
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Adel Atari
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yuen-Yi Tseng
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Nabeel Bardeesy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts, USA
| | - William R. Sellers
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
3
|
Seraji N, Berger I. FGFR as a Predictive Marker for Targeted Therapy in Gastrointestinal Malignancies: A Systematic Review. J Gastrointest Cancer 2025; 56:96. [PMID: 40205008 PMCID: PMC11982104 DOI: 10.1007/s12029-025-01214-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
BACKGROUND Gastrointestinal (GI) cancers constitute approximately 25% of cancers worldwide. The fibroblast growth factor receptor (FGFR) family is a promising target for immunotherapy aiming to enhance survival rates. FGFR alterations are associated with GI carcinomas. Their predictive value in different malignancies remains a focus area. While FGFR inhibitors have been approved for cholangiocarcinoma (CC) therapy, uncertainties remain regarding other GI cancers. METHODS A systematic review was conducted using the following databases: CINAHL, Embase, Medline, Cochrane Library, PubMed, and Web of Science. The search terms included "FGFR" and each of the GI malignancies. A total of 18 studies were included in this review. RESULTS The efficacy of FGFR-targeted therapy is evident. Strong evidence supports the use of FGFR inhibitors in CC, gastro-oesophageal cancer (GC/OC), and hepatocellular cancer, while there is limited evidence for pancreatic cancer (PC) and colorectal cancer (CRC). Alteration forms like FGFR2 fusion or rearrangement are associated with CC, while FGFR2 amplification and FGFR2b overexpression are associated with GC/OC. The administration of multi-kinase inhibitors without prior genomic testing, makes distinct study outcomes not solely attributable to the FGFR blockade. CONCLUSION FGFRs have a predictive value for GI cancers. Certain FGFR alterations are predictable for specific GI cancers. The most established FGFR-targeted therapy is for CC. It is essential to expand the FGFR research field for PC and CRC. Consistent molecular diagnostics in clinical trials are vital to comprehend the patient population with the highest efficacy.
Collapse
Affiliation(s)
- Nika Seraji
- Faculty of Medicine, University of Southampton, Southampton, UK.
| | - Irina Berger
- Department of Pathology, Klinikum Kassel, Kassel, Germany
| |
Collapse
|
4
|
Kumbham S, Md Mahabubur Rahman K, Foster BA, You Y. A Comprehensive Review of Current Approaches in Bladder Cancer Treatment. ACS Pharmacol Transl Sci 2025; 8:286-307. [PMID: 39974639 PMCID: PMC11833730 DOI: 10.1021/acsptsci.4c00663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/21/2025]
Abstract
Bladder cancer is one of the most common malignant tumors of the urinary system globally. It is also one of the most expensive cancers to manage, due to the need for extensive treatment and follow-ups that often involve invasive and costly procedures. Although there have been some improvements in treatment options, the quality of life they offer has not improved at the same rate as other cancers. Therefore, there is an urgent need to find new alternatives to ease the burden of bladder cancer on patients. Recent discoveries have opened new avenues for the diagnosis and management of bladder cancer even though the clinical approach has largely remained the same for years. The decline in bladder cancer-specific mortality in regions that promote social awareness of risk factors and reduction of carcinogenic exposure demonstrates the effectiveness of such measures. New agents have been approved for patients who have undergone radical cystectomy after Bacillus Calmette-Guérin failure. Current best practices for diagnosing and treating bladder cancer are presented in this review. The review discusses radiation therapy, photodynamic therapy, gene therapy, chemotherapy, and nanomedicine in relation to non muscle-invasive cancers and muscle-invasive bladder cancers, as well as systemic treatments.
Collapse
Affiliation(s)
- Soniya Kumbham
- Department
of Pharmaceutical Sciences, University at
Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Kazi Md Mahabubur Rahman
- Department
of Pharmaceutical Sciences, University at
Buffalo, The State University of New York, Buffalo, New York 14214, United States
| | - Barbara A. Foster
- Department
of Pharmacology & Therapeutics, Roswell
Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Youngjae You
- Department
of Pharmaceutical Sciences, University at
Buffalo, The State University of New York, Buffalo, New York 14214, United States
| |
Collapse
|
5
|
Kim Y, Song J, Kim N, Sim T. Recent progress in emerging molecular targeted therapies for intrahepatic cholangiocarcinoma. RSC Med Chem 2025:d4md00881b. [PMID: 39925737 PMCID: PMC11800140 DOI: 10.1039/d4md00881b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 01/11/2025] [Indexed: 02/11/2025] Open
Abstract
Cholangiocarcinoma (CCA) is a diverse group of epithelial malignant tumors arising from the biliary tract, characterized by high molecular heterogeneity. It is classified into intrahepatic (iCCA) and extrahepatic CCA (eCCA) based on the location of the primary tumor. CCA accounts for approximately 15% of all primary liver cancers, with iCCA comprising 10-20% of all CCAs. iCCA is especially known for its characteristic aggressiveness and refractoriness, leading to poor prognosis. Despite the increasing global incidence and mortality rates, surgery remains the only available standard treatment approach for a subset (25%) of patients with early-stage, resectable iCCA. The paucity of effective systemic medical therapies restricts therapeutic options for patients with advanced or metastatic iCCA. In the past decade, advances in the understanding of the molecular complexity of these tumors have provided fruitful insights for the identification of promising new druggable targets and the development of feasible therapeutic strategies that may improve treatment outcomes for patients with iCCA. In this review, we aim to highlight critical up-to-date studies and medicinal chemistry aspects, focusing on novel targeted approaches utilizing promising candidates for molecular targeted therapy in iCCA. These candidates include aberrations in isocitrate dehydrogenase (IDH) 1/2, fibroblast growth factor receptor (FGFR), B-Raf proto-oncogene (BRAF), neurotrophic tyrosine receptor kinase (NTRK), human epidermal growth factor receptor 2 (HER2), and programmed cell death protein 1 (PD-1)/programmed cell death-ligand 1 (PD-L1). Furthermore, this review provides an overview of potential inhibitors aimed at overcoming acquired drug resistance in these actionable targets for iCCA.
Collapse
Affiliation(s)
- Younghoon Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
| | - Jaewon Song
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| | - Namkyoung Kim
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
| | - Taebo Sim
- KU-KIST Graduate School of Converging Science and Technology, Korea University 145 Anam-ro, Seongbuk-gu Seoul 02841 Korea
- Department of Biomedical Sciences, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea +822 2228 0797
- Clinical Candidate Discovery & Development Institute, Yonsei University College of Medicine Seoul Korea
- Graduate School of Clinical Drug Discovery & Development, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine 50 Yonsei-ro, Seodaemun-gu Seoul 03722 Republic of Korea
| |
Collapse
|
6
|
Mihara T, Tsuru Y, Kurosawa T, Nonoshita Y, Yamakawa Y, Hori M. Pemigatinib suppresses liver fibrosis and subsequent osteodystrophy in mice. Hepatol Commun 2025; 9:e0610. [PMID: 39774090 PMCID: PMC11717528 DOI: 10.1097/hc9.0000000000000610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 11/09/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Liver fibrosis could lead to serious secondary diseases, including osteodystrophy. The interaction between liver and bone has not been fully elucidated, thus existing therapies for osteodystrophy secondary to liver fibrosis are often ineffective. FGF23 was initially found as an endocrine regulator of phosphate homeostasis, but recently, its involvement in fibrosis has been suggested. In this study, we hypothesized that the FGF23 level increases with liver injury, which in turn induces liver fibrosis and osteodystrophy. METHODS Liver fibrosis model mice were generated via carbon tetrachloride administration and bile duct ligation. Fibrosis was assessed using Masson trichrome staining and hydroxyproline assay. The bone structure was evaluated using dual-energy x-ray absorptiometry and microcomputed tomography. Human HSC lines LX-2 and primary rat HSCs were used for in vitro analyses. RESULTS Carbon tetrachloride-induced and bile duct ligation-induced liver injury increased the serum FGF23 level compared with that in control mice. RNA sequencing analysis of FGF23-treated LX-2 showed that FGF23 promotes the production of matrisome, which helps in forming the extracellular matrix. The FGF receptor antagonist pemigatinib alleviated carbon tetrachloride-induced and bile duct ligation-induced liver fibrosis and the deleterious alterations in bone density and microstructure in mice. CONCLUSIONS The serum FGF23 level increased with liver injury, and FGF23 promoted liver fibrosis. Moreover, pemigatinib alleviated liver fibrosis and hepatic osteodystrophy. These findings suggest that FGF23 mediates the communication between the liver and bone and that FGF23 may be a new therapeutic target for liver fibrosis and subsequent osteodystrophy.
Collapse
Affiliation(s)
- Taiki Mihara
- Department of Veterinary Medical Science, Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yoshiharu Tsuru
- Primetech Life Science Laboratory, Primetech Corporation, Tokyo, Japan
| | - Tamaki Kurosawa
- Department of Veterinary Medical Science, Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuma Nonoshita
- Department of Veterinary Medical Science, Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuki Yamakawa
- Department of Veterinary Medical Science, Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Masatoshi Hori
- Department of Veterinary Medical Science, Veterinary Pharmacology, Graduate School of Agriculture and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
7
|
Bansal K, Chaudhary N, Bhati H, Singh V. Unveiling FDA-approved Drugs and Formulations in the Management of Bladder Cancer: A Review. Curr Pharm Biotechnol 2025; 26:48-62. [PMID: 38797905 DOI: 10.2174/0113892010314650240514053735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024]
Abstract
Urological cancers are one of the most prevalent malignancies around the globe. Specifically, bladder cancer severely threatens the health of humans because of its heterogeneous and aggressive nature. Extensive studies have been conducted for many years in order to address the limitations associated with the treatment of solid tumors with selective substances. This article aims to provide a summary of the therapeutic drugs that have received FDA approval or are presently in the testing phase for use in the prevention or treatment of bladder cancer. In this review, FDA-approved drugs for bladder cancer treatment have been listed along with their dose protocols, current status, pharmacokinetics, action mechanisms, and marketed products. The article also emphasizes the novel preparations of these drugs that are presently under clinical trials or are in the approval stage. Thus, this review will serve as a single point of reference for scientists involved in the formulation development of these drugs.
Collapse
Affiliation(s)
- Keshav Bansal
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Neeraj Chaudhary
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Hemant Bhati
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| | - Vanshita Singh
- Institute of Pharmaceutical Research, GLA University, Mathura-281406, Uttar Pradesh, India
| |
Collapse
|
8
|
Peng M, Deng J, Li X. Clinical advances and challenges in targeting FGF/FGFR signaling in lung cancer. Mol Cancer 2024; 23:256. [PMID: 39543657 PMCID: PMC11566285 DOI: 10.1186/s12943-024-02167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
Fibroblast growth factors (FGFs) and their receptors regulate numerous cellular processes, such as metabolism and signal transduction, but can also drive tumorigenesis. Specifically, in lung cancer, the overexpression of FGFs, as well as the amplification, mutation and fusion of FGFR genes, are closely linked to the initiation, progression and resistance of the disease, suggesting that targeting FGF/FGFR is an attractive therapeutic strategy for lung cancer treatment. Nintedanib, a multitarget tyrosine kinase inhibitor (TKI) used in combination with docetaxel, has shown some success as a second-line therapy for lung cancer. However, clinical trials evaluating other FGFR inhibitors have yielded mixed results, indicating substantial complexity in targeting aberrant FGF/FGFR signaling. In this review, we describe the aberrations in FGF/FGFR signaling in lung cancer and summarize the clinical efficacy of FGFR inhibitors, such as multitarget TKIs, selective FGFR-TKIs and biological agents. We also discuss various challenges associated with FGFR targeting in lung cancer, including precision patient selection, toxicity and resistance. Finally, we provide perspectives on future directions, namely, developing novel FGFR-targeting drugs, such as FGFR degraders and more specific FGFR-TKIs, adopting combination therapy and targeting FGFs.
Collapse
Affiliation(s)
- Mei Peng
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| | - Jun Deng
- Department of Pharmacy, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan, 410000, P. R. China
| | - Xiangping Li
- Department of Pharmacy, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, P. R. China.
| |
Collapse
|
9
|
Hung KL, Jones MG, Wong ITL, Curtis EJ, Lange JT, He BJ, Luebeck J, Schmargon R, Scanu E, Brückner L, Yan X, Li R, Gnanasekar A, Chamorro González R, Belk JA, Liu Z, Melillo B, Bafna V, Dörr JR, Werner B, Huang W, Cravatt BF, Henssen AG, Mischel PS, Chang HY. Coordinated inheritance of extrachromosomal DNAs in cancer cells. Nature 2024; 635:201-209. [PMID: 39506152 PMCID: PMC11541006 DOI: 10.1038/s41586-024-07861-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 07/19/2024] [Indexed: 11/08/2024]
Abstract
The chromosomal theory of inheritance dictates that genes on the same chromosome segregate together while genes on different chromosomes assort independently1. Extrachromosomal DNAs (ecDNAs) are common in cancer and drive oncogene amplification, dysregulated gene expression and intratumoural heterogeneity through random segregation during cell division2,3. Distinct ecDNA sequences, termed ecDNA species, can co-exist to facilitate intermolecular cooperation in cancer cells4. How multiple ecDNA species within a tumour cell are assorted and maintained across somatic cell generations is unclear. Here we show that cooperative ecDNA species are coordinately inherited through mitotic co-segregation. Imaging and single-cell analyses show that multiple ecDNAs encoding distinct oncogenes co-occur and are correlated in copy number in human cancer cells. ecDNA species are coordinately segregated asymmetrically during mitosis, resulting in daughter cells with simultaneous copy-number gains in multiple ecDNA species before any selection. Intermolecular proximity and active transcription at the start of mitosis facilitate the coordinated segregation of ecDNA species, and transcription inhibition reduces co-segregation. Computational modelling reveals the quantitative principles of ecDNA co-segregation and co-selection, predicting their observed distributions in cancer cells. Coordinated inheritance of ecDNAs enables co-amplification of specialized ecDNAs containing only enhancer elements and guides therapeutic strategies to jointly deplete cooperating ecDNA oncogenes. Coordinated inheritance of ecDNAs confers stability to oncogene cooperation and novel gene regulatory circuits, allowing winning combinations of epigenetic states to be transmitted across cell generations.
Collapse
Affiliation(s)
- King L Hung
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Matthew G Jones
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Ivy Tsz-Lo Wong
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Ellis J Curtis
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
- School of Medicine, University of California at San Diego, La Jolla, CA, USA
| | - Joshua T Lange
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Britney Jiayu He
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA, USA
| | - Rachel Schmargon
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Elisa Scanu
- Department of Mathematics, Queen Mary University of London, London, UK
| | - Lotte Brückner
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
| | - Xiaowei Yan
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Aditi Gnanasekar
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Rocío Chamorro González
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Julia A Belk
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA
| | - Zhonglin Liu
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
| | - Bruno Melillo
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
- Chemical Biology and Therapeutics Science Program, Broad Institute, Cambridge, MA, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA, USA
| | - Jan R Dörr
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Benjamin Werner
- Evolutionary Dynamics Group, Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Weini Huang
- Department of Mathematics, Queen Mary University of London, London, UK
- Group of Theoretical Biology, The State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Benjamin F Cravatt
- Department of Chemistry, Scripps Research, La Jolla, CA, USA
- Vividion Therapeutics, San Diego, CA, USA
| | - Anton G Henssen
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin and German Cancer Research Center DKFZ, Heidelberg, Germany
- Berlin Institute of Health, Berlin, Germany
| | - Paul S Mischel
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA.
- Department of Pathology, Stanford University, Stanford, CA, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA, USA.
- Department of Genetics, Stanford University, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
10
|
Kosaka K, Takayama N, Paul SK, Kanashiro MA, Oshima M, Fukuyo M, Rahmutulla B, Tajiri I, Mukai M, Kubota Y, Akita S, Furuyama N, Kaneda A, Iwama A, Eto K, Mitsukawa N. iPSC-derived megakaryocytes and platelets accelerate wound healing and angiogenesis. Stem Cell Res Ther 2024; 15:364. [PMID: 39402677 PMCID: PMC11477011 DOI: 10.1186/s13287-024-03966-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 09/30/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Platelet-rich plasma (PRP), which is prepared by concentrating platelets in autologous blood, shows efficacy in chronic skin wounds via multiple growth factors. However, it exhibits heterogeneity across patients, leading to unstable therapeutic efficacy. Human induced pluripotent stem cell (iPSC)-derived megakaryocytes and platelets (iMPs) are capable of providing a stable supply, holding promise as materials for novel platelet concentrate-based therapies. In this context, we evaluated the effect of iMPs on wound healing and validated lyophilization for clinical applications. METHODS The growth factors released by activated iMPs were measured. The effect of the administration of iMPs on human fibroblasts and human umbilical vein endothelial cells (HUVECs) was investigated in vitro. iMPs were applied to dorsal skin defects of diabetic mice to assess the wound closure rate and quantify collagen deposition and angiogenesis. Following the storage of freeze-dried iMPs (FD-iMPs) for three months, the stability of growth factors and their efficacy in animal models were determined. RESULT Multiple growth factors that promote wound healing were detected in activated iMPs. iMPs specifically released FGF2 and exhibited a superior enhancement of HUVEC proliferation compared to PRP. Moreover, an RNA-seq analysis revealed that iMPs induce polarization to stalk cells and enhance ANGPTL4 gene expression in HUVECs. Animal studies demonstrated that iMPs promoted wound closure and angiogenesis in chronic wounds caused by diabetes. We also confirmed the long-term stability of growth factors in FD-iMPs and their comparable effects to those of original iMPs in the animal model. CONCLUSION Our study demonstrates that iMPs promote angiogenesis and wound healing through the activation of vascular endothelial cells. iMPs exhibited more effectiveness than PRP, an effect attributed to the exclusive presence of specific factors including FGF2. Lyophilization enabled the long-term maintenance of the composition of the growth factors and efficacy of the iMPs, therefore contributing to stable supply for clinical application. These findings suggest that iMPs provide a novel treatment for chronic wounds.
Collapse
Affiliation(s)
- Kentaro Kosaka
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan.
| | - Naoya Takayama
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Sudip Kumar Paul
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | - Motohiko Oshima
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Masaki Fukuyo
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Bahityar Rahmutulla
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Ikuko Tajiri
- Department of Orthopedic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Michiaki Mukai
- Department of Orthopedic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Yoshitaka Kubota
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Shinsuke Akita
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | - Atsushi Kaneda
- Department of Molecular Oncology, Chiba University Graduate School of Medicine, Chiba, Japan
- Health and Disease Omics Center, Chiba University, Chiba, Japan
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Koji Eto
- Department of Regenerative Medicine, Chiba University Graduate School of Medicine, Chiba, Japan.
- Department of Clinical Application, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan.
| | - Nobuyuki Mitsukawa
- Department of Plastic, Reconstructive, and Aesthetic Surgery, Chiba University Graduate School of Medicine, Chiba, Japan.
| |
Collapse
|
11
|
Gnagni L, Ruscito I, Zizzari IG, Nuti M, Napoletano C, Rughetti A. Precision oncology targeting FGFRs: A systematic review on pre-clinical activity and clinical outcomes of pemigatinib. Crit Rev Oncol Hematol 2024; 202:104464. [PMID: 39094670 DOI: 10.1016/j.critrevonc.2024.104464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024] Open
Abstract
Fibroblast Growth Factor Receptors (FGFRs) are emerging as key factors involved in tumorigenesis, tumor microenvironment remodeling and acquired resistance to targeted therapies. Pemigatinib is a Tyrosine-Kinase Inhibitor that selectively targets aberrant FGFR1, FGFR2 and FGFR3. Pemigatinib is now approved for advanced-stage cholangiocarcinoma (CCA) but data suggests that other tumor histotypes exhibit FGFR alterations, thus hypothesizing its potential efficacy in other cancer settings. The present systematic review, based on PRISMA guidelines, aims to synthetize and critically interpret the results of all available preclinical and clinical evidence regarding Pemigatinib use in cancer. In April 2024, an extensive search was performed in PubMed, MEDLINE, and Scopus databases using the keyword "Pemigatinib". Twenty-seven studies finally met all inclusion criteria. The promising results emerging from Pemigatinib preclinical and clinical studies pave the way for Pemigatinib extension to multiple solid cancer settings.
Collapse
Affiliation(s)
- Ludovica Gnagni
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy
| | - Ilary Ruscito
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy.
| | - Ilaria Grazia Zizzari
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy.
| | - Aurelia Rughetti
- Department of Experimental Medicine, Sapienza University of Rome, Viale Regina Elena, Rome 324 -00161, Italy
| |
Collapse
|
12
|
Guo YJ, Ma MX, Tian T, Zhang JN, Guo XN, Qiao S. Myeloid/lymphoid neoplasms with eosinophilia and FGFR1 rearrangement t(8;13)(p11;q12): A case report and literature review. Oncol Lett 2024; 28:468. [PMID: 39119236 PMCID: PMC11306990 DOI: 10.3892/ol.2024.14601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
8p11 myeloproliferative syndrome (EMS) is a rare and aggressive hematological malignancy, characterized by myeloproliferative neoplasms, and associated with eosinophilia and T- or B-cell lineage lymphoblastic lymphoma. The pathogenesis is defined by the presence of chromosomal translocations associated with the fibroblast growth factor-1 (FGFR1) gene, located in the 8p11-12.1 chromosomal locus. At present, only ~100 cases have been reported globally. At least 15 partner genes have been identified, including the most common, the zinc finger MYM-type containing 2 (ZNF198)-FGFR1 fusion gene formed by t(8;13)(p11;q12). Different fusion genes determine the clinical manifestations and prognosis of the disease. Patients with EMS with t(8;13)(p11;q12) commonly present with lymphadenopathy and T-lymphoblastic lymphoma, which usually converts to acute myeloid leukemia (AML) with the progression of the disease. The present study describes the case of an elderly female patient with EMS with t(8;13)(p11;q12), presenting with myeloid/lymphoid syndrome (myeloproliferative neoplasms and T lymphoblastic lymphoma). The patient received the CHOPE regimen combined with tyrosine kinase inhibitor (dasatin) treatment and obtained short-term complete remission. However, 6 months later, the disease progressed from EMS to AML and the patient died due to ineffective induction therapy. The present study also reviews the relevant literature about this unusual entity to enhance the understanding of EMS.
Collapse
Affiliation(s)
- Yu-Jie Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Meng-Xue Ma
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Tian Tian
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Jing-Nan Zhang
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiao-Nan Guo
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| | - Shukai Qiao
- Department of Hematology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, P.R. China
| |
Collapse
|
13
|
Chaturantabut S, Oliver S, Frederick DT, Kim J, Robinson FP, Sinopoli A, Song TY, Rodriguez DJ, Chang L, Kesar D, He Y, Ching M, Dzvurumi R, Atari A, Tseng YY, Bardeesy N, Sellers WR. Identification of potent biparatopic antibodies targeting FGFR2 fusion driven cholangiocarcinoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613045. [PMID: 39345400 PMCID: PMC11429734 DOI: 10.1101/2024.09.16.613045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Translocations involving FGFR2 gene fusions are common in cholangiocarcinoma and predict response to FGFR kinase inhibitors. However, the rate and durability of response are limited due to the emergence of resistance, typically involving acquired FGFR2 kinase domain mutations, and to sub-optimal dosing, relating to drug adverse effects. Here, we report the development of biparatopic antibodies targeting the FGFR2 extracellular domain (ECD), as candidate therapeutics. Biparatopic antibodies can overcome drawbacks of standard bivalent monoparatopic antibodies, which often show poor inhibitory or even agonist activity against oncogenic receptors. We show that oncogenic transformation by FGFR2 fusions requires an intact ECD. Moreover, by systematically generating biparatopic antibodies that target distinct epitope pairs along the FGFR2 ECD, we identified antibodies that effectively block signaling and malignant growth driven by FGFR2-fusions. Importantly, these antibodies demonstrate efficacy in vivo, synergy with FGFR inhibitors, and activity against FGFR2 fusions harboring kinase domain mutations. Thus, biparatopic antibodies may serve as new treatment options for patients with FGFR2-altered cholangiocarcinoma. Summary We identify biparatopic FGFR2 antibodies that are effective against FGFR2 fusion driven cholangiocarcinoma.
Collapse
|
14
|
Niu S, Zhang Y, Li Z, Wang T. Prognostic value of FGFR2 alterations in patients with iCCA undergoing surgery or systemic treatments: A meta-analysis. Liver Int 2024; 44:2208-2219. [PMID: 38829010 DOI: 10.1111/liv.15984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 04/25/2024] [Accepted: 05/13/2024] [Indexed: 06/05/2024]
Abstract
BACKGROUND Over recent years, there has been a notable rise in the incidence of intrahepatic cholangiocarcinoma (iCCA), which presents a significant challenge in treatment due to its complex disease characteristics and prognosis. Notably, the identification of fibroblast growth factor receptor 2 (FGFR2) fusion/rearrangement, a potential oncogenic driver primarily observed in iCCA, raises questions about its impact on the prognostic outcomes of patients undergoing surgical intervention or other therapeutic approaches. METHODS A comprehensive search from inception to July 2023 was conducted across PubMed, Embase, Web of Science, and the Cochrane Library databases. The objective was to identify relevant publications comparing the prognosis of FGFR2 alterations and no FGFR2 alterations groups among patients with iCCA undergoing surgical resection or other systemic therapies. The primary outcome indicators, specifically Overall Survival (OS) and Disease-Free Survival (DFS), were estimated using Hazard Ratios (HRs) with 95% confidence intervals (CIs), and statistical significance was defined as p < .05. Study quality was assessed using the Newcastle-Ottawa Quality Assessment Scale. Statistical analyses were performed using Review Manager 5.4 software and Stata, version 12.0. RESULTS Six studies, involving 1314 patients (FGFR2 alterations group n = 173 and no FGFR2 alterations group n = 1141), were included in the meta-analysis. The analysis revealed that the FGFR2 alterations group exhibited a significantly better OS prognosis compared to the no FGFR2 alterations group, with a fixed-effects combined effect size HR = 1.31, 95%CI = 1.001-1.715, p = .049. Furthermore, meta-regression and subgroup analysis showed that the length of the follow-up period did not introduce heterogeneity into the results. This finding indicates the stability and reliability of the study outcomes. CONCLUSION The current study provides compelling evidence that FGFR2 alterations are frequently associated with improved survival outcomes for patients with iCCA undergoing surgical resection or other systemic treatments. Additionally, the study suggests that FGFR2 holds promise as a safe and dependable therapeutic target for managing metastatic, locally advanced or unresectable iCCA. This study offers a novel perspective in the realm of targeted therapy for iCCA, presenting a new and innovative approach to its treatment.
Collapse
Affiliation(s)
- Sen Niu
- Department of General Surgery, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Ye Zhang
- Department of General Surgery, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Zengyao Li
- Department of General Surgery, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Tong Wang
- Department of General Surgery, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|
15
|
Wang J, Liu S, Cao Y, Chen Y. Overcoming treatment resistance in cholangiocarcinoma: current strategies, challenges, and prospects. Front Cell Dev Biol 2024; 12:1408852. [PMID: 39156971 PMCID: PMC11327014 DOI: 10.3389/fcell.2024.1408852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024] Open
Abstract
Significant advancements in our understanding and clinical treatment of cholangiocarcinoma (CCA) have been achieved over the past 5 years. Groundbreaking studies have illuminated the immune landscape and pathological characteristics of the tumor microenvironment in CCA. The development of immune- and metabolism-based classification systems has enabled a nuanced exploration of the tumor microenvironment and the origins of CCA, facilitating a detailed understanding of tumor progression modulation. Despite these insights, targeted therapies have not yet yielded satisfactory clinical results, highlighting the urgent need for innovative therapeutic strategies. This review delineates the complexity and heterogeneity of CCA, examines the current landscape of therapeutic strategies and clinical trials, and delves into the resistance mechanisms underlying targeted therapies. Finally, from a single-cell and spatial transcriptomic perspective, we address the challenge of therapy resistance, discussing emerging mechanisms and potential strategies to overcome this barrier and enhance treatment efficacy.
Collapse
Affiliation(s)
- Jiayi Wang
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Siyan Liu
- International Medical College, Chongqing Medical University, Chongqing, China
| | - Yi Cao
- Second Clinical College, Chongqing Medical University, Chongqing, China
| | - Yong Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Gudivada IP, Amajala KC. Integrative Bioinformatics Analysis for Targeting Hub Genes in Hepatocellular Carcinoma Treatment. Curr Genomics 2024; 26:48-80. [PMID: 39911278 PMCID: PMC11793067 DOI: 10.2174/0113892029308243240709073945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/24/2024] [Accepted: 06/11/2024] [Indexed: 02/07/2025] Open
Abstract
Background The damage in the liver and hepatocytes is where the primary liver cancer begins, and this is referred to as Hepatocellular Carcinoma (HCC). One of the best methods for detecting changes in gene expression of hepatocellular carcinoma is through bioinformatics approaches. Objective This study aimed to identify potential drug target(s) hubs mediating HCC progression using computational approaches through gene expression and protein-protein interaction datasets. Methodology Four datasets related to HCC were acquired from the GEO database, and Differentially Expressed Genes (DEGs) were identified. Using Evenn, the common genes were chosen. Using the Fun Rich tool, functional associations among the genes were identified. Further, protein-protein interaction networks were predicted using STRING, and hub genes were identified using Cytoscape. The selected hub genes were subjected to GEPIA and Shiny GO analysis for survival analysis and functional enrichment studies for the identified hub genes. The up-regulating genes were further studied for immunohistopathological studies using HPA to identify gene/protein expression in normal vs HCC conditions. Drug Bank and Drug Gene Interaction Database were employed to find the reported drug status and targets. Finally, STITCH was performed to identify the functional association between the drugs and the identified hub genes. Results The GEO2R analysis for the considered datasets identified 735 upregulating and 284 downregulating DEGs. Functional gene associations were identified through the Fun Rich tool. Further, PPIN network analysis was performed using STRING. A comparative study was carried out between the experimental evidence and the other seven data evidence in STRING, revealing that most proteins in the network were involved in protein-protein interactions. Further, through Cytoscape plugins, the ranking of the genes was analyzed, and densely connected regions were identified, resulting in the selection of the top 20 hub genes involved in HCC pathogenesis. The identified hub genes were: KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Further, GEPIA and Shiny GO analyses provided insights into survival ratios and functional enrichment studied for the hub genes. The HPA database studies further found that upregulating genes were involved in changes in protein expression in Normal vs HCC tissues. These findings indicated that hub genes were certainly involved in the progression of HCC. STITCH database studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, could be used as leads to identify novel drugs, and identified hub genes could also be considered as potential and promising drug targets as they are involved in the gene-chemical interaction networks. Conclusion The present study involved various integrated bioinformatics approaches, analyzing gene expression and protein-protein interaction datasets, resulting in the identification of 20 top-ranked hubs involved in the progression of HCC. They are KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Gene-chemical interaction network studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, can be used as leads to identify novel drugs, and the identified hub genes can be promising drug targets. The current study underscores the significance of targeting these hub genes and utilizing existing molecules to generate new molecules to combat liver cancer effectively and can be further explored in terms of drug discovery research to develop treatments for HCC.
Collapse
Affiliation(s)
- Indu Priya Gudivada
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Krishna Chaitanya Amajala
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| |
Collapse
|
17
|
Esmail A, Badheeb M, Alnahar BW, Almiqlash B, Sakr Y, Al-Najjar E, Awas A, Alsayed M, Khasawneh B, Alkhulaifawi M, Alsaleh A, Abudayyeh A, Rayyan Y, Abdelrahim M. The Recent Trends of Systemic Treatments and Locoregional Therapies for Cholangiocarcinoma. Pharmaceuticals (Basel) 2024; 17:910. [PMID: 39065760 PMCID: PMC11279608 DOI: 10.3390/ph17070910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Cholangiocarcinoma (CCA) is a hepatic malignancy that has a rapidly increasing incidence. CCA is anatomically classified into intrahepatic (iCCA) and extrahepatic (eCCA), which is further divided into perihilar (pCCA) and distal (dCCA) subtypes, with higher incidence rates in Asia. Despite its rarity, CCA has a low 5-year survival rate and remains the leading cause of primary liver tumor-related death over the past 10-20 years. The systemic therapy section discusses gemcitabine-based regimens as primary treatments, along with oxaliplatin-based options. Second-line therapy is limited but may include short-term infusional fluorouracil (FU) plus leucovorin (LV) and oxaliplatin. The adjuvant therapy section discusses approaches to improve overall survival (OS) post-surgery. However, only a minority of CCA patients qualify for surgical resection. In comparison to adjuvant therapies, neoadjuvant therapy for unresectable cases shows promise. Gemcitabine and cisplatin indicate potential benefits for patients awaiting liver transplantation. The addition of immunotherapies to chemotherapy in combination is discussed. Nivolumab and innovative approaches like CAR-T cells, TRBAs, and oncolytic viruses are explored. We aim in this review to provide a comprehensive report on the systemic and locoregional therapies for CCA.
Collapse
Affiliation(s)
- Abdullah Esmail
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Mohamed Badheeb
- Department of Internal Medicine, Yale New Haven Health, Bridgeport Hospital, Bridgeport, CT 06610, USA
| | | | - Bushray Almiqlash
- Zuckerman College of Public Health, Arizona State University, Tempe, AZ 85287, USA;
| | - Yara Sakr
- Department of GI Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ebtesam Al-Najjar
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
| | - Ali Awas
- Faculty of Medicine and Health Sciences, University of Science and Technology, Sanaa P.O. Box 15201-13064, Yemen
| | | | - Bayan Khasawneh
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
| | | | - Amneh Alsaleh
- Department of Medicine, Desert Regional Medical Center, Palm Springs, CA 92262, USA
| | - Ala Abudayyeh
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yaser Rayyan
- Department of Gastroenterology & Hepatology, Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Maen Abdelrahim
- Section of GI Oncology, Houston Methodist Neal Cancer Center, Houston Methodist Hospital, Houston, TX 77030, USA
| |
Collapse
|
18
|
Lindley A, Prager G, Bitzer M, Burn TC, Lihou CF, Croft E. Global Expanded Access Program for Pemigatinib in Patients with Previously Treated Locally Advanced or Metastatic Cholangiocarcinoma and Fibroblast Growth Factor Receptor Gene Alterations. Cancer Res Treat 2024; 56:847-855. [PMID: 38351684 PMCID: PMC11261204 DOI: 10.4143/crt.2023.1197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 02/06/2024] [Indexed: 07/18/2024] Open
Abstract
PURPOSE Pemigatinib is a fibroblast growth factor receptor-2 (FGFR2) inhibitor approved for use in patients with previously treated cholangiocarcinoma (CCA) and FGFR2 fusions or rearrangements. This ongoing global Expanded Access Program (EAP) allows physicians in regions where pemigatinib is not commercially available to request pemigatinib for patients with locally advanced or metastatic CCA who, in the physician's opinion, could benefit from pemigatinib treatment. MATERIALS AND METHODS Eighty-nine patients from Europe, North America, and Israel were treated from January 2020 through September 2021. RESULTS Patients had FGFR gene fusions (68.5%), rearrangements (12.4%), translocations (5.6%), amplifications (2.2%), and other alterations (11.2%). Median duration of treatment in the EAP was 4.0 months (range, 0.1 to 13.6 months). The most frequently reported adverse event (AE) was hyperphosphatemia (22.5%); the most common serious AE was cholangitis (3.4%). Treatment discontinuation was associated with reports of AEs for seven patients (7.9%). AEs associated with pemigatinib were consistent with those observed in clinical trials. CONCLUSION Efficacy was not assessed in this EAP. However, some patients remained on treatment for up to a year, suggesting that they observed a benefit from treatment. Patients with CCA should undergo molecular testing to identify those who could benefit from targeted treatments such as pemigatinib.
Collapse
Affiliation(s)
| | - Gerald Prager
- Medical University of Vienna, Department of Internal Medicine I, Comprehensive Cancer Center Vienna, Vienna, Austria
| | - Michael Bitzer
- Department of Internal Medicine I, Eberhard-Karls University, Tübingen, Germany
| | | | | | | |
Collapse
|
19
|
Shang S, Zhang L, Liu K, Lv M, Zhang J, Ju D, Wei D, Sun Z, Wang P, Yuan J, Zhu Z. Landscape of targeted therapies for advanced urothelial carcinoma. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2024; 5:641-677. [PMID: 38966172 PMCID: PMC11220318 DOI: 10.37349/etat.2024.00240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/06/2024] [Indexed: 07/06/2024] Open
Abstract
Bladder cancer (BC) is the tenth most common malignancy globally. Urothelial carcinoma (UC) is a major type of BC, and advanced UC (aUC) is associated with poor clinical outcomes and limited survival rates. Current options for aUC treatment mainly include chemotherapy and immunotherapy. These options have moderate efficacy and modest impact on overall survival and thus highlight the need for novel therapeutic approaches. aUC patients harbor a high tumor mutation burden and abundant molecular alterations, which are the basis for targeted therapies. Erdafitinib is currently the only Food and Drug Administration (FDA)-approved targeted therapy for aUC. Many potential targeted therapeutics aiming at other molecular alterations are under investigation. This review summarizes the current understanding of molecular alterations associated with aUC targeted therapy. It also comprehensively discusses the related interventions for treatment in clinical research and the potential of using novel targeted drugs in combination therapy.
Collapse
Affiliation(s)
- Shihao Shang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Lei Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Kepu Liu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Maoxin Lv
- Department of Urology, First Affiliated Hospital of Kunming Medical University, Kunming 65000, Yunnan, China
| | - Jie Zhang
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
- College of Life Sciences, Northwest University, Xi’an 710068, Shaanxi, China
| | - Dongen Ju
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Di Wei
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zelong Sun
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Pinxiao Wang
- School of Clinical Medicine, Xi’an Medical University, Xi’an 710021, Shaanxi, China
| | - Jianlin Yuan
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Zheng Zhu
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| |
Collapse
|
20
|
Zhang P, Yue L, Leng Q, Chang C, Gan C, Ye T, Cao D. Targeting FGFR for cancer therapy. J Hematol Oncol 2024; 17:39. [PMID: 38831455 PMCID: PMC11149307 DOI: 10.1186/s13045-024-01558-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/21/2024] [Indexed: 06/05/2024] Open
Abstract
The FGFR signaling pathway is integral to cellular activities, including proliferation, differentiation, and survival. Dysregulation of this pathway is implicated in numerous human cancers, positioning FGFR as a prominent therapeutic target. Here, we conduct a comprehensive review of the function, signaling pathways and abnormal alterations of FGFR, as well as its role in tumorigenesis and development. Additionally, we provide an in-depth analysis of pivotal phase 2 and 3 clinical trials evaluating the performance and safety of FGFR inhibitors in oncology, thereby shedding light on the current state of clinical research in this field. Then, we highlight four drugs that have been approved for marketing by the FDA, offering insights into their molecular mechanisms and clinical achievements. Our discussion encompasses the intricate landscape of FGFR-driven tumorigenesis, current techniques for pinpointing FGFR anomalies, and clinical experiences with FGFR inhibitor regimens. Furthermore, we discuss the inherent challenges of targeting the FGFR pathway, encompassing resistance mechanisms such as activation by gatekeeper mutations, alternative pathways, and potential adverse reactions. By synthesizing the current evidence, we underscore the potential of FGFR-centric therapies to enhance patient prognosis, while emphasizing the imperative need for continued research to surmount resistance and optimize treatment modalities.
Collapse
Affiliation(s)
- Pei Zhang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Lin Yue
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - QingQing Leng
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Chen Chang
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Cailing Gan
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tinghong Ye
- Laboratory of Gastrointestinal Cancer and Liver Disease, Department of Gastroenterology and Hepatology, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Dan Cao
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, No. 37 Guoxue Alley, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
21
|
Sasaki M, Sato Y, Nakanuma Y. Expression of fibroblast growth factor receptor 2 (FGFR2) in combined hepatocellular-cholangiocarcinoma and intrahepatic cholangiocarcinoma: clinicopathological study. Virchows Arch 2024; 484:915-923. [PMID: 38532197 PMCID: PMC11186861 DOI: 10.1007/s00428-024-03792-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 03/12/2024] [Accepted: 03/19/2024] [Indexed: 03/28/2024]
Abstract
Genetic alterations including fusions in fibroblast growth factor receptor 2 (FGFR2) are detected in 10-20% of intrahepatic cholangiocarcinoma (iCCA), and FGFR2 inhibitors are effective for the treatment of iCCA. We examined a prevalence of FGFR2 genetic alterations and their clinicopathological significance in combined hepatocellular-cholangiocarcinoma (cHCC-CCA). FGFR2 expression, which is a surrogate marker for FGFR2 genetic alterations, was immunohistochemically assessed in the liver sections from 75 patients with cHCC-CCA, 35 with small duct-type iCCA, 30 with large duct-type iCCA, and 35 with hepatocellular carcinoma (HCC). FGFR2 genetic alterations were detected by reverse transcription-PCR and direct sequence. An association of FGFR2 expression with clinicopathological features was investigated in cHCC-CCAs. FGFR2 expression was detected in significantly more patients with cHCC-CCA (21.3%) and small duct-type iCCA (25.7%), compared to those with large duct-type iCCA (3.3%) and HCC (0%) (p < 0.05). FGFR2-positive cHCC-CCAs were significantly smaller size (p < 0.05), with more predominant cholangiolocarcinoma component (p < 0.01) and less nestin expression (p < 0.05). Genetic alterations of ARID1A and BAP1 and multiple genes were significantly more frequent in FGFR2-positive cHCC-CCAs (p < 0.05). 5'/3' imbalance in FGFR2 genes indicating exon18-truncated FGFR2 was significantly more frequently detected in FGFR2-positive cHCC-CCAs and small duct iCCAs, compared to FGFR2-negative ones (p < 0.05). FGFR2::BICC fusion was detected in a case of cHCC-CCAs. FGFR2 genetic alterations may be prevalent in cHCC-CCAs as well as small duct-type iCCAs, which suggest cHCC-CCAs may also be a possible therapeutic target of FGFR2 inhibitors.
Collapse
MESH Headings
- Humans
- Cholangiocarcinoma/pathology
- Cholangiocarcinoma/genetics
- Cholangiocarcinoma/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Female
- Male
- Bile Duct Neoplasms/pathology
- Bile Duct Neoplasms/genetics
- Bile Duct Neoplasms/metabolism
- Middle Aged
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Aged
- Adult
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
- Aged, 80 and over
- Immunohistochemistry
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Ubiquitin Thiolesterase
Collapse
Affiliation(s)
- Motoko Sasaki
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan.
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, 920-8640, Japan
| | - Yasuni Nakanuma
- Division of Pathology, Fukui Saiseikai Hospital, Fukui, Japan
| |
Collapse
|
22
|
Rodón J, Damian S, Furqan M, García-Donas J, Imai H, Italiano A, Spanggaard I, Ueno M, Yokota T, Veronese ML, Oliveira N, Li X, Gilmartin A, Schaffer M, Goyal L. Pemigatinib in previously treated solid tumors with activating FGFR1-FGFR3 alterations: phase 2 FIGHT-207 basket trial. Nat Med 2024; 30:1645-1654. [PMID: 38710951 PMCID: PMC11186762 DOI: 10.1038/s41591-024-02934-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/19/2024] [Indexed: 05/08/2024]
Abstract
Fibroblast growth factor receptor (FGFR) alterations drive oncogenesis in multiple tumor types. Here we studied pemigatinib, a selective, potent, oral FGFR1-FGFR3 inhibitor, in the phase 2 FIGHT-207 basket study of FGFR-altered advanced solid tumors. Primary end points were objective response rate (ORR) in cohorts A (fusions/rearrangements, n = 49) and B (activating non-kinase domain mutations, n = 32). Secondary end points were progression-free survival, duration of response and overall survival in cohorts A and B, and safety. Exploratory end points included ORR of cohort C (kinase domain mutations, potentially pathogenic variants of unknown significance, n = 26) and analysis of co-alterations associated with resistance and response. ORRs for cohorts A, B and C were 26.5% (13/49), 9.4% (3/32) and 3.8% (1/26), respectively. Tumors with no approved FGFR inhibitors or those with alterations not previously confirmed to be sensitive to FGFR inhibition had objective responses. In cohorts A and B, the median progression-free survival was 4.5 and 3.7 months, median duration of response was 7.8 and 6.9 months and median overall survival was 17.5 and 11.4 months, respectively. Safety was consistent with previous reports. The most common any-grade treatment-emergent adverse events were hyperphosphatemia (84%) and stomatitis (53%). TP53 co-mutations were associated with lack of response and BAP1 alterations with higher response rates. FGFR1-FGFR3 gatekeeper and molecular brake mutations led to acquired resistance. New therapeutic areas for FGFR inhibition and drug failure mechanisms were identified across tumor types. ClinicalTrials.gov identifier: NCT03822117 .
Collapse
MESH Headings
- Humans
- Receptor, Fibroblast Growth Factor, Type 3/genetics
- Receptor, Fibroblast Growth Factor, Type 3/antagonists & inhibitors
- Female
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors
- Pyrimidines/adverse effects
- Pyrimidines/therapeutic use
- Male
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/pathology
- Middle Aged
- Adult
- Aged
- Mutation
- Protein Kinase Inhibitors/adverse effects
- Protein Kinase Inhibitors/therapeutic use
- Progression-Free Survival
- Drug Resistance, Neoplasm/genetics
- Drug Resistance, Neoplasm/drug effects
- Morpholines
- Pyrroles
Collapse
Affiliation(s)
- Jordi Rodón
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Silvia Damian
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | | | - Hiroo Imai
- Tohoku University Hospital, Sendai-Shi, Japan
| | - Antoine Italiano
- Institut Bergonié, Bordeaux, France
- Faculty of Medicine, University of Bordeaux, Bordeaux, France
| | - Iben Spanggaard
- Rigshospitalet Copenhagen University Hospital, Copenhagen, Denmark
| | | | | | | | | | - Xin Li
- Incyte Corporation, Wilmington, DE, USA
| | | | | | - Lipika Goyal
- Mass General Cancer Center, Harvard Medical School, Boston, MA, USA.
- Stanford Cancer Center, Stanford School of Medicine, Stanford, CA, USA.
| |
Collapse
|
23
|
Berro A, Assi A, Farhat M, Hatoum L, Saad JP, Mohanna R, Bechara AMA, Prince G, Hachem MCR, Zalaquett Z, Kourie HR. Unlocking Hope: Anti-VEGFR inhibitors and their potential in glioblastoma treatment. Crit Rev Oncol Hematol 2024; 198:104365. [PMID: 38677355 DOI: 10.1016/j.critrevonc.2024.104365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/19/2024] [Indexed: 04/29/2024] Open
Abstract
PURPOSE This systematic review summarizes evidence of VEGFR gene mutations and VEGF/VEGFR protein expression in glioblastoma multiforme (GBM) patients, alongside the efficacy and safety of anti-VEGFR tyrosine kinase inhibitors (TKIs) for GBM treatment. METHODS A comprehensive literature review was conducted using PubMed up to August 2023. Boolean operators and MeSH term "glioma," along with specific VEGFR-related keywords, were utilized following thorough examination of existing literature. RESULTS VEGFR correlates with glioma grade and GBM progression, presenting a viable therapeutic target. Regorafenib and axitinib show promise among studied TKIs. Other multi-targeted TKIs (MTKI) and combination therapies exhibit potential, albeit limited by blood-brain barrier penetration and toxicity. Combining treatments like radiotherapy and enhancing BBB penetration may benefit patients. Further research is warranted in patient quality of life and biomarker-guided selection. CONCLUSION While certain therapies hold promise for GBM, future research should prioritize personalized medicine and innovative strategies for improved treatment outcomes.
Collapse
Affiliation(s)
- Ali Berro
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Ahmad Assi
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Mohamad Farhat
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Lea Hatoum
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Jean-Pierre Saad
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Rami Mohanna
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Anna Maria Antoun Bechara
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Gilles Prince
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Maria Catherine Rita Hachem
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| | - Ziad Zalaquett
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon.
| | - Hampig-Raphael Kourie
- Hematology-Oncology Department, Hôtel-Dieu de France University Hospital, Saint Joseph University of Beirut, Beirut, Lebanon
| |
Collapse
|
24
|
Lau DK, Collin JP, Mariadason JM. Clinical Developments and Challenges in Treating FGFR2-Driven Gastric Cancer. Biomedicines 2024; 12:1117. [PMID: 38791079 PMCID: PMC11118914 DOI: 10.3390/biomedicines12051117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Recent advances in the treatment of gastric cancer (GC) with chemotherapy, immunotherapy, anti-angiogenic therapy and targeted therapies have yielded some improvement in survival outcomes; however, metastatic GC remains a lethal malignancy and amongst the leading causes of cancer-related mortality worldwide. Importantly, the ongoing molecular characterisation of GCs continues to uncover potentially actionable molecular targets. Among these, aberrant FGFR2-driven signalling, predominantly arising from FGFR2 amplification, occurs in approximately 3-11% of GCs. However, whilst several inhibitors of FGFR have been clinically tested to-date, there are currently no approved FGFR-directed therapies for GC. In this review, we summarise the significance of FGFR2 as an actionable therapeutic target in GC, examine the recent pre-clinical and clinical data supporting the use of small-molecule inhibitors, antibody-based therapies, as well as novel approaches such as proteolysis-targeting chimeras (PROTACs) for targeting FGFR2 in these tumours, and discuss the ongoing challenges and opportunities associated with their clinical development.
Collapse
Affiliation(s)
- David K. Lau
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Oncology, Monash Health, Clayton, VIC 3168, Australia
| | - Jack P. Collin
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| | - John M. Mariadason
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084, Australia;
- School of Cancer Medicine, La Trobe University, Heidelberg, VIC 3084, Australia
| |
Collapse
|
25
|
Heumann P, Albert A, Gülow K, Tümen D, Müller M, Kandulski A. Current and Future Therapeutic Targets for Directed Molecular Therapies in Cholangiocarcinoma. Cancers (Basel) 2024; 16:1690. [PMID: 38730642 PMCID: PMC11083102 DOI: 10.3390/cancers16091690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 04/17/2024] [Accepted: 04/21/2024] [Indexed: 05/13/2024] Open
Abstract
We conducted a comprehensive review of the current literature of published data, clinical trials (MEDLINE; ncbi.pubmed.com), congress contributions (asco.org; esmo.org), and active recruiting clinical trains (clinicaltrial.gov) on targeted therapies in cholangiocarcinoma. Palliative treatment regimens were analyzed as well as preoperative and perioperative treatment options. We summarized the current knowledge for each mutation and molecular pathway that is or has been under clinical evaluation and discussed the results on the background of current treatment guidelines. We established and recommended targeted treatment options that already exist for second-line settings, including IDH-, BRAF-, and NTRK-mutated tumors, as well as for FGFR2 fusion, HER2/neu-overexpression, and microsatellite instable tumors. Other options for targeted treatment include EGFR- or VEGF-dependent pathways, which are known to be overexpressed or dysregulated in this cancer type and are currently under clinical investigation. Targeted therapy in CCA is a hallmark of individualized medicine as these therapies aim to specifically block pathways that promote cancer cell growth and survival, leading to tumor shrinkage and improved patient outcomes based on the molecular profile of the tumor.
Collapse
Affiliation(s)
- Philipp Heumann
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases University Hospital Regensburg Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | | | | | | | | | - Arne Kandulski
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology, Rheumatology, and Infectious Diseases University Hospital Regensburg Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
26
|
Katoh M, Loriot Y, Brandi G, Tavolari S, Wainberg ZA, Katoh M. FGFR-targeted therapeutics: clinical activity, mechanisms of resistance and new directions. Nat Rev Clin Oncol 2024; 21:312-329. [PMID: 38424198 DOI: 10.1038/s41571-024-00869-z] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Fibroblast growth factor (FGF) signalling via FGF receptors (FGFR1-4) orchestrates fetal development and contributes to tissue and whole-body homeostasis, but can also promote tumorigenesis. Various agents, including pan-FGFR inhibitors (erdafitinib and futibatinib), FGFR1/2/3 inhibitors (infigratinib and pemigatinib), as well as a range of more-specific agents, have been developed and several have entered clinical use. Erdafitinib is approved for patients with urothelial carcinoma harbouring FGFR2/3 alterations, and futibatinib and pemigatinib are approved for patients with cholangiocarcinoma harbouring FGFR2 fusions and/or rearrangements. Clinical benefit from these agents is in part limited by hyperphosphataemia owing to off-target inhibition of FGFR1 as well as the emergence of resistance mutations in FGFR genes, activation of bypass signalling pathways, concurrent TP53 alterations and possibly epithelial-mesenchymal transition-related isoform switching. The next generation of small-molecule inhibitors, such as lirafugratinib and LOXO-435, and the FGFR2-specific antibody bemarituzumab are expected to have a reduced risk of hyperphosphataemia and the ability to overcome certain resistance mutations. In this Review, we describe the development and current clinical role of FGFR inhibitors and provide perspective on future research directions including expansion of the therapeutic indications for use of FGFR inhibitors, combination of these agents with immune-checkpoint inhibitors and the application of novel technologies, such as artificial intelligence.
Collapse
Affiliation(s)
| | - Yohann Loriot
- Drug Development Department (DITEP), Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
- INSERM U981, Institut Gustave Roussy, Université Paris-Saclay, Villejuif, France
| | - Giovanni Brandi
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simona Tavolari
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Zev A Wainberg
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Masaru Katoh
- M & M Precision Medicine, Tokyo, Japan.
- Department of Omics Network, National Cancer Center, Tokyo, Japan.
| |
Collapse
|
27
|
Jain NK, Tailang M, Thangavel N, Makeen HA, Albratty M, Najmi A, Alhazmi HA, Zoghebi K, Alagusundaram M, Jain HK, Chandrasekaran B. A comprehensive overview of selective and novel fibroblast growth factor receptor inhibitors as a potential anticancer modality. ACTA PHARMACEUTICA (ZAGREB, CROATIA) 2024; 74:1-36. [PMID: 38554385 DOI: 10.2478/acph-2024-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 11/09/2023] [Indexed: 04/01/2024]
Abstract
The arrival of comprehensive genome sequencing has accelerated the understanding of genetically aberrant advanced cancers and target identification for possible cancer treatment. Fibroblast growth factor receptor (FGFR) gene alterations are frequent findings in various rare and advanced cancers refractive to mainstay chemo-therapy or surgical interventions. Several FGFR inhibitors have been developed for addressing these genetically altered FGFR-harboring malignancies, and some have performed well in clinical trials. In contrast, others are still being investigated in different phases of clinical trials. FDA has approved four anticancer agents such as erdafitinib, pemigatinib, infigratinib, and futibatinib, for clinical use in oncogenic FGFR-driven malignancies. These include cholangiocarcinoma, urothelial carcinoma, and myeloid/lymphoid malignancies. Pemigatinib is the only FGFR inhibitor globally approved (USA, EU, and Japan) and available as a targeted therapy for two types of cancer, including FGFR2 fusion or other rearrangements harboring cholangiocarcinoma and relapsed/refractory myeloid/lymphoid neoplasms with FGFR1 rearrangements. Myeloid/lymphoid neoplasm is the latest area of application added to the therapeutic armamentarium of FGFR inhibitors. Furthermore, futibatinib is the first-in-class covalent or irreversible pan-FGFR inhibitor that has received FDA approval for locally advanced or metastatic intrahepatic cholangiocarcinoma harboring FGFR2 gene aberrations. This review highlights the current clinical progress concerning the safety and efficacy of all the approved FGFR-TKIs (tyrosine kinase inhibitors) and their ongoing investigations in clinical trials for other oncogenic FGFR-driven malignancies.
Collapse
Affiliation(s)
- Nem Kumar Jain
- School of Pharmacy, ITM University Gwalior 474001, Madhya Pradesh, India
- School of Studies in Pharmaceutical Sciences, Jiwaji University Gwalior 474001, Madhya Pradesh, India
| | - Mukul Tailang
- School of Studies in Pharmaceutical Sciences, Jiwaji University Gwalior 474001, Madhya Pradesh, India
| | - Neelaveni Thangavel
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit Department of Clinical Pharmacy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | - Asim Najmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | - Hassan Ahmad Alhazmi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | - Khalid Zoghebi
- Department of Pharmaceutical Chemistry and Pharmacognosy, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 82912, Saudi Arabia
| | | | - Hemant Kumar Jain
- Department of General Medicine Government Medical College Datia 475661, Madhya Pradesh, India
| | | |
Collapse
|
28
|
Ding W, Yan L, Sheng L, Chen S, Li Y, Cheng S, Luo L, Huang H, Shao H, Zhang D. Identification of Piperazinyl-Difluoro-indene Derivatives Containing Pyridyl Groups as Potent FGFR Inhibitors against FGFR Mutant Tumor: Design, Synthesis, and Biological Evaluation. J Med Chem 2024; 67:2941-2962. [PMID: 38294952 DOI: 10.1021/acs.jmedchem.3c02040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
The fibroblast growth factor receptor (FGFR) signaling pathway plays important roles in cellular processes such as proliferation, differentiation, and migration. In this study, we highlighted the potential of FGFR inhibitors bearing the (S)-3,3-difluoro-1-(4-methylpiperazin-1-yl)-2,3-dihydro-1H-indene scaffold containing a crucial 3-pyridyl group for the treatment of FGFR mutant cancers. The representative compound (S)-23, which was identified through comprehensive evaluation, exhibited potent antiproliferative activity with GI50 in the range of 6.4-10.4 nM against FGFR1 fusion protein-carrying, FGFR2-amplified, and FGFR2 mutant cancer cell lines and good antiproliferative activity against FGFR3 translocation and mutant FGFR4 cancer cell lines, as well as potency assessment against FGFR1-4 kinases. Moreover, compound (S)-23 exhibited favorable pharmacokinetic properties, low potential for drug-drug interactions, and very potent antitumor activity in MFE-296 xenograft mouse models with a TGI of 99.1% at the dose of 10 mg/kg. These findings demonstrate that compound (S)-23 is a potential therapeutic agent for FGFR mutant tumors.
Collapse
Affiliation(s)
- Wei Ding
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Liting Yan
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Li Sheng
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Shuting Chen
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Ying Li
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Shihao Cheng
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Lijun Luo
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Haihong Huang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| | - Huanjie Shao
- College of Life Sciences, Shaanxi Normal University, Xi'an 710119, P. R. China
| | - Dongfeng Zhang
- Beijing Key Laboratory of Active Substance Discovery and Druggability Evaluation, Institute of Materia Medica, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100050, P. R. China
| |
Collapse
|
29
|
Necchi A, Pouessel D, Leibowitz R, Gupta S, Fléchon A, García-Donas J, Bilen MA, Debruyne PR, Milowsky MI, Friedlander T, Maio M, Gilmartin A, Li X, Veronese ML, Loriot Y. Pemigatinib for metastatic or surgically unresectable urothelial carcinoma with FGF/FGFR genomic alterations: final results from FIGHT-201. Ann Oncol 2024; 35:200-210. [PMID: 37956738 DOI: 10.1016/j.annonc.2023.10.794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/17/2023] [Accepted: 10/26/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND Fibroblast growth factor receptor 3 (FGFR3) alterations are oncogenic drivers of urothelial carcinoma (UC). Pemigatinib is a selective, oral inhibitor of FGFR1-3 with antitumor activity. We report the efficacy and safety of pemigatinib in the open-label, single-arm, phase II study of previously treated, unresectable or metastatic UC with FGFR3 alterations (FIGHT-201; NCT02872714). PATIENTS AND METHODS Patients ≥18 years old with FGFR3 mutations or fusions/rearrangements (cohort A) and other FGF/FGFR alterations (cohort B) were included. Patients received pemigatinib 13.5 mg once daily continuously (CD) or intermittently (ID) until disease progression or unacceptable toxicity. The primary endpoint was centrally confirmed objective response rate (ORR) as per RECIST v1.1 in cohort A-CD. Secondary endpoints included ORR in cohorts A-ID and B, duration of response (DOR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS Overall, 260 patients were enrolled and treated (A-CD, n = 101; A-ID, n = 103; B, n = 44; unconfirmed FGF/FGFR status, n = 12). All discontinued treatment, most commonly due to progressive disease (68.5%). ORR [95% confidence interval (CI)] in cohorts A-CD and A-ID was 17.8% (10.9% to 26.7%) and 23.3% (15.5% to 32.7%), respectively. Among patients with the most common FGFR3 mutation (S249C; n = 107), ORR was similar between cohorts (A-CD, 23.9%; A-ID, 24.6%). In cohorts A-CD/A-ID, median (95% CI) DOR was 6.2 (4.1-8.3)/6.2 (4.6-8.0) months, PFS was 4.0 (3.5-4.2)/4.3 (3.9-6.1) months, and OS was 6.8 (5.3-9.1)/8.9 (7.5-15.2) months. Pemigatinib had limited clinical activity among patients in cohort B. Of 36 patients with samples available at progression, 6 patients had 8 acquired FGFR3 secondary resistance mutations (V555M/L, n = 3; V553M, n = 1; N540K/S, n = 2; M528I, n = 2). The most common treatment-emergent adverse events overall were diarrhea (44.6%) and alopecia, stomatitis, and hyperphosphatemia (42.7% each). CONCLUSIONS Pemigatinib was generally well tolerated and demonstrated clinical activity in previously treated, unresectable or metastatic UC with FGFR3 mutations or fusions/rearrangements.
Collapse
Affiliation(s)
- A Necchi
- Vita-Salute San Raffaele University, Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy.
| | - D Pouessel
- Institut Claudius Regaud-IUCT Oncopole, Toulouse, France
| | - R Leibowitz
- Chaim Sheba Medical Center, Ramat Gan; Shamir Medical Center, Zerifin, Israel
| | - S Gupta
- Huntsman Cancer Institute, Salt Lake City, USA
| | | | | | - M A Bilen
- Winship Cancer Institute of Emory University, Atlanta, USA
| | - P R Debruyne
- Kortrijk Cancer Centre, General Hospital Groeninge, Kortrijk, Belgium; Medical Technology Research Centre (MTRC), School of Life Sciences, Anglia Ruskin University, Cambridge; School of Nursing and Midwifery, University of Plymouth, Plymouth, UK
| | - M I Milowsky
- University of North Carolina Lineberger Comprehensive Cancer Center, Chapel Hill
| | - T Friedlander
- Helen Diller Family Comprehensive Cancer Center, University of California San Francisco, San Francisco, USA
| | - M Maio
- University of Siena and Center for Immuno-Oncology, Department of Oncology, University Hospital, Siena, Italy
| | | | - X Li
- Incyte Corporation, Wilmington, USA
| | - M L Veronese
- Incyte International Biosciences Sàrl, Morges, Switzerland
| | - Y Loriot
- Gustave Roussy, DITEP, Université Paris-Saclay, INSERM 981, Villejuif, France.
| |
Collapse
|
30
|
Fan Z, Zou X, Wang G, Liu Y, Jiang Y, Wang H, Zhang P, Wei F, Du X, Wang M, Sun X, Ji B, Hu X, Chen L, Zhou P, Wang D, Bai J, Xiao X, Zuo L, Xia X, Yi X, Lv G. A transcriptome based molecular classification scheme for cholangiocarcinoma and subtype-derived prognostic biomarker. Nat Commun 2024; 15:484. [PMID: 38212331 PMCID: PMC10784309 DOI: 10.1038/s41467-024-44748-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
Previous studies on the molecular classification of cholangiocarcinoma (CCA) focused on certain anatomical sites, and disregarded tissue contamination biases in transcriptomic profiles. We aim to provide universal molecular classification scheme and prognostic biomarker of CCAs across anatomical locations. Comprehensive bioinformatics analysis is performed on transcriptomic data from 438 CCA cases across various anatomical locations. After excluding CCA tumors showing normal tissue expression patterns, we identify two universal molecular subtypes across anatomical subtypes, explore the molecular, clinical, and microenvironmental features of each class. Subsequently, a 30-gene classifier and a biomarker (called "CORE-37") are developed to predict the molecular subtype of CCA and prognosis, respectively. Two subtypes display distinct molecular characteristics and survival outcomes. Key findings are validated in external cohorts regardless of the stage and anatomical location. Our study provides a CCA classification scheme that complements the conventional anatomy-based classification and presents a promising prognostic biomarker for clinical application.
Collapse
Affiliation(s)
- Zhongqi Fan
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xinchen Zou
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
| | - Guangyi Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Yanfang Jiang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Haoyan Wang
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
| | - Ping Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Feng Wei
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaohong Du
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Meng Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Xintong Hu
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Liguo Chen
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Peiwen Zhou
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Duo Wang
- Genetic Diagnosis Center, The First Hospital of Jilin University, Changchun, China
| | - Jing Bai
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
| | - Xiao Xiao
- Geneplus-Shenzhen, No.14 Zhongxing Road, Pingshan District, Shenzhen, China
| | - Lijiao Zuo
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
| | - Xuefeng Xia
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
| | - Xin Yi
- Geneplus-Beijing Institute, 9th Floor, No.6 Building, Peking University Medical Industrial Park, Zhongguancun Life Science Park, Beijing, China
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
31
|
Frampton JE. Pemigatinib: A Review in Advanced Cholangiocarcinoma. Target Oncol 2024; 19:107-114. [PMID: 38206555 DOI: 10.1007/s11523-023-01024-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2023] [Indexed: 01/12/2024]
Abstract
Pemigatinib (Pemazyre®), a selective, potent, reversible, oral inhibitor of fibroblast growth factor receptor (FGFR) 1-3, has received conditional (in the EU) or accelerated (in the USA) approval for the treatment of adults with previously treated, unresectable locally-advanced or metastatic cholangiocarcinoma (CCA) with an FGFR2 gene fusion or rearrangement. Over the course of a single-arm, phase 2 study (FIGHT-202), just over a third of patients with pretreated, advanced CCA [almost exclusively intrahepatic CCA (iCCA)] harbouring an FGFR2 fusion or rearrangement who received pemigatinib once daily (2 weeks on, 1 week off) had an objective response; nearly half had stable disease. Median progression-free survival and overall survival at the time of the final analysis were 7.0 months and 17.5 months, respectively. Pemigatinib was generally well tolerated and had a manageable safety profile. The most common treatment-related adverse event, hyperphosphataemia, was exclusively grade 1-2 in severity and, similarly, observed ocular and nail toxicities were rarely grade ≥ 3 in severity. Pending confirmation of its clinical benefits in an ongoing cisplatin plus gemcitabine-controlled, phase 3 study (FIGHT-302), pemigatinib provides a valuable targeted therapy for pretreated patients with advanced (i)CCA harbouring a FGFR2 fusion or rearrangement.
Collapse
Affiliation(s)
- James E Frampton
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
32
|
Deng T, Zhang L, Shi Y, Bai G, Pan Y, Shen A, Han X, Yang Z, Chen M, Zhou H, Luo Y, Zheng S, Ba Y. Pharmacokinetics, pharmacodynamics and efficacy of pemigatinib (a selective inhibitor of fibroblast growth factor receptor 1-3) monotherapy in Chinese patients with advanced solid tumors: a phase i clinical trial. Invest New Drugs 2023; 41:808-815. [PMID: 37889382 PMCID: PMC10663244 DOI: 10.1007/s10637-023-01396-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023]
Abstract
Pemigatinib is a selective fibroblast growth factor receptor (FGFR)1-3 inhibitor and has demonstrated acceptable tolerability and clinical activity in advanced solid tumors in Western population. This phase I trial evaluated pharmacokinetics/pharmacodynamics (PK/PD) characteristics, preliminary safety and efficacy of pemigatinib in Chinese patients with advanced, solid tumors. Patients with unresectable advanced or metastatic solid tumors bearing FGF/FGFR1-3 alterations received oral pemigatinib at 13.5 mg once daily (QD) on a 2-weeks-on/1-week-off schedule. The primary endpoint was PK/PD characteristics; secondary endpoints were safety and efficacy. Twelve patients were enrolled (median age: 61 years, 58.3% males). PK data demonstrated pemigatinib (13.5 mg QD) was rapidly absorbed with a geometric mean elimination half-life of 11.3 h. The geometric mean values of maximum serum concentration and area under the plasma concentration-time curve from 0 to 24 h at steady state were 215.1 nmol/L and 2636.9 h·nmol/L, respectively. The mean clearance adjusted by bioavailability at steady state was low (11.8 L/h), and the apparent oral volume of distribution was moderate (170.5 L). The PD marker, serum phosphate level, increased on days 8 and 15 of cycle 1 (mean: 2.25 mg/dL, CV% [percent coefficient of variation]: 31.3%) and decreased to baseline post 1 week off. Three (25.0%) patients experienced grade ≥ 3 treatment-emergent adverse events. Partial response was confirmed in one patient with FGFR1-mutant esophageal carcinoma and one with FGFR2-mutant cholagiocarcinoma. Pemigatinib had similar PK/PD characteristics to Western population and demonstrated an acceptable safety profile and potential anti-cancer benefit in Chinese patients with FGF/FGFR1-3 altered, advanced, solid tumor. (ClinicalTrials.gov: NCT04258527 [prospectively registered February 6, 2020]).
Collapse
Affiliation(s)
- Ting Deng
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, and Tianjin's Clinical Research Center for Cancer, and Tianjin's Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Le Zhang
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, and Tianjin's Clinical Research Center for Cancer, and Tianjin's Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yehui Shi
- Phase I Clinical Trial Ward, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, & Tianjin's Clinical Research Center for Cancer, & Tianjin's Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Guiying Bai
- Phase I Clinical Trial Ward, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, & Tianjin's Clinical Research Center for Cancer, & Tianjin's Key Laboratory of Cancer Prevention and Therapy, Tianjin, China
| | - Yueyin Pan
- Oncology Department, Anhui Provincial Hospital, Hefei, China
| | - Aizong Shen
- Pharmacy Department, Anhui Provincial Hospital, Hefei, China
| | - Xinghua Han
- Oncology Department, Anhui Provincial Hospital, Hefei, China
| | - Zhaoyi Yang
- Pharmacy Department, Anhui Provincial Hospital, Hefei, China
| | - Mingxia Chen
- Department of Biostatistics and Information, Innovent Biologics, Inc, Suzhou, China
| | - Hui Zhou
- Department of Medical Science and Oncology, Innovent Biologics, Inc, Suzhou, China
| | - Yang Luo
- Department of Medical Science and Oncology, Innovent Biologics, Inc, Suzhou, China
| | - Shirui Zheng
- Department of Clinical Pharmacology, Innovent Biologics, Inc, Suzhou, China
| | - Yi Ba
- Department of GI Medical Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, and Tianjin's Clinical Research Center for Cancer, and Tianjin's Key Laboratory of Cancer Prevention and Therapy, Tianjin, China.
- Department of Cancer Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
33
|
Subbiah V, Verstovsek S. Clinical development and management of adverse events associated with FGFR inhibitors. Cell Rep Med 2023; 4:101204. [PMID: 37757826 PMCID: PMC10591034 DOI: 10.1016/j.xcrm.2023.101204] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 06/02/2023] [Accepted: 08/31/2023] [Indexed: 09/29/2023]
Abstract
Approved fibroblast growth factor receptor (FGFR) inhibitors include erdafitinib, pemigatinib, and futibatinib. We review the most common toxicities associated with FGFR inhibitors and provide practical advice regarding their management. Hyperphosphatemia can be managed with careful monitoring, dose reduction or interruption, a prophylactic low-phosphate diet, and phosphate-lowering therapy. Ocular adverse events (AEs) are managed by withholding or adjusting the dose of the FGFR inhibitor. Dermatologic AEs include alopecia, which can be managed with minoxidil, and dry skin, which can be treated with moisturizers. Hand-foot syndrome can be prevented by lifestyle changes and managed with moisturizing creams, urea, or salicylic acid. Among gastrointestinal AEs, diarrhea may be managed with loperamide; stomatitis can be managed with baking soda rinses, mucosa-coating agents, and topical anesthetics; and dry mouth may be alleviated with salivary stimulants. Most FGFR inhibitor-associated toxicities are manageable with prophylactic measures and treatments; proactive monitoring is key to ensuring optimal clinical benefits.
Collapse
Affiliation(s)
- Vivek Subbiah
- Sarah Cannon Research Institute, Nashville, TN, USA.
| | - Srdan Verstovsek
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
34
|
Storandt MH, Kurniali PC, Mahipal A, Jin Z. Targeted Therapies in Advanced Cholangiocarcinoma. Life (Basel) 2023; 13:2066. [PMID: 37895447 PMCID: PMC10608206 DOI: 10.3390/life13102066] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/02/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Primary tumor resection and liver transplantation are the only curative treatment options for the management of cholangiocarcinoma (CCA). However, for patients with advanced or metastatic disease, palliative systemic therapy remains the only treatment option. The development of targeted therapeutics has begun to shift the treatment paradigm in CCA. Targets of interest in CCA include mutated isocitrate dehydrogenase-1 (mIDH-1), human epidermal growth factor receptor 2 (HER2) overexpression/amplification, and fibroblast growth factor receptor 2 (FGFR2) fusion, in addition to less frequently observed targets such as BRAF V600E, deficient mismatch repair/high microsatellite instability (dMMR/MSI-H), and high tumor mutation burden (TMB-H). These targets are observed in varying frequency among patients with intrahepatic CCA and extrahepatic CCA. Multiple novel therapies have been developed to exploit each of these targets, with some having received United States Food and Drug Administration approval for use in the second-line setting. In the current review, we discuss targets of interest in CCA and summarize current evidence evaluating available therapies directed at these targets.
Collapse
Affiliation(s)
| | - Peter C. Kurniali
- Department of Hematology/Medical Oncology, Sanford Cancer Center, Bismarck, ND 58501, USA;
- Department of Internal Medicine, Division of Hematology/Oncology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND 58203, USA
| | - Amit Mahipal
- Department of Oncology, University Hospitals Seidman Cancer Center and Case Western Reserve University, Cleveland, OH 44106, USA;
| | - Zhaohui Jin
- Department of Medical Oncology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
35
|
Gandhy SU, Casak SJ, Mushti SL, Cheng J, Subramaniam S, Zhao H, Zhao M, Bi Y, Liu G, Fan J, Adeniyi O, Charlab R, Kufrin D, Thompson MD, Jarrell K, Auth D, Lemery SJ, Pazdur R, Kluetz PG, Fashoyin-Aje LA. FDA Approval Summary: Futibatinib for Unresectable Advanced or Metastatic, Chemotherapy Refractory Intrahepatic Cholangiocarcinoma with FGFR2 Fusions or Other Rearrangements. Clin Cancer Res 2023; 29:4027-4031. [PMID: 37289037 PMCID: PMC10592512 DOI: 10.1158/1078-0432.ccr-23-1042] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/19/2023] [Accepted: 06/06/2023] [Indexed: 06/09/2023]
Abstract
On September 30, 2022, the FDA granted accelerated approval to futibatinib for the treatment of adult patients with previously treated, unresectable, locally advanced or metastatic intrahepatic cholangiocarcinoma (iCCA) with FGFR2 fusions or other rearrangements. Approval was based on Study TAS-120-101, a multicenter open-label, single-arm trial. Patients received futibatinib 20-mg orally once daily. The major efficacy outcome measures were overall response rate (ORR) and duration of response (DoR) as determined by an independent review committee (IRC) according to RECIST v1.1. ORR was 42% (95% confidence interval, 32%-52%). Median DoR was 9.7 months. Adverse reactions occurring in ≥30% patients were nail toxicity, musculoskeletal pain, constipation, diarrhea, fatigue, dry mouth, alopecia, stomatitis, and abdominal pain. The most common laboratory abnormalities (≥50%) were increased phosphate, increased creatinine, decreased hemoglobin, and increased glucose. Ocular toxicity (including dry eye, keratitis, and retinal epithelial detachment) and hyperphosphatemia are important risks of futibatinib, which are listed under Warnings and Precautions. This article summarizes the FDA's thought process and data supporting the approval of futibatinib.
Collapse
Affiliation(s)
- Shruti U. Gandhy
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Sandra J. Casak
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Sirisha L Mushti
- Office of Biostatistics, Office of Translational Sciences
(OTS), CDER, U.S. FDA
| | - Joyce Cheng
- Office of Biostatistics, Office of Translational Sciences
(OTS), CDER, U.S. FDA
| | | | - Hong Zhao
- Office of Clinical Pharmacology, OTS, CDER, U.S. FDA
| | - Miao Zhao
- Office of Clinical Pharmacology, OTS, CDER, U.S. FDA
| | - Youwei Bi
- Office of Clinical Pharmacology, OTS, CDER, U.S. FDA
| | - Guansheng Liu
- Office of Clinical Pharmacology, OTS, CDER, U.S. FDA
| | - Jianghong Fan
- Office of Clinical Pharmacology, OTS, CDER, U.S. FDA
| | | | | | - Dubravka Kufrin
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Matthew D. Thompson
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Kristin Jarrell
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Doris Auth
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Steven J. Lemery
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Richard Pazdur
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Paul G. Kluetz
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| | - Lola A. Fashoyin-Aje
- Office of Oncologic Diseases, Center for Drug Evaluation
and Research (CDER), U.S. Food and Drug Administration (FDA)
| |
Collapse
|
36
|
Al-Qargholi B, Al-Dolaimy F, Altalbawy FMA, Kadhim AJ, Alsaalamy AH, Suliman M, Abbas AHR. Surface modification of a screen-printed electrode with a flower-like nanostructure to fabricate a guanine DNA-based electrochemical biosensor to determine the anticancer drug pemigatinib. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2023; 15:5146-5156. [PMID: 37753580 DOI: 10.1039/d3ay01103h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/28/2023]
Abstract
The present study developed a DNA biosensor to determine pemigatinib for the first time. Three-dimensional carnation flower-like Eu3+:β-MnO2 nanostructures (3D CF-L Eu3+:β-MnO2 NSs) and a screen-printed electrode (SPE) modified with polyaniline (PA) were employed. The double-stranded DNA was also immobilized completely on the PA/3D CF-L Eu3+:β-MnO2 NSs/SPE. Then, electrochemical techniques were used for characterizing the modified electrode. After that, the interaction between pemigatinib and DNA was shown by a reduction in the oxidation current of guanine using differential pulse voltammetry (DPV). According to the analysis, the dynamic range of pemigatinib was between 0.001 and 180.0 μM, indicating the new electrode has a low limit of detection (LOD = 0.23 nM) for pemigatinib. Afterwards, pemigatinib in real samples was measured using the PA/3D CF-L Eu3+:β-MnO2 NSs/SPE loaded with ds-DNA. The proposed DNA biosensor showed good selectivity toward pemigatinib in the presence of other interference analytes, such as other ions, structurally related pharmaceuticals, and plasma proteins. In addition, the interaction site of pemigatinib with DNA was predicted by molecular docking, which showed the interaction of pemigatinib with the guanine bases of DNA through a groove binding mode. Finally, we employed the t-test to verify the capability of the ds-DNA/PA/3D CF-L Eu3+:β-MnO2 NSs/SPE for analyzing pemigatinib in real samples.
Collapse
Affiliation(s)
- Basim Al-Qargholi
- Biomedical Engineering Department, Al-Mustaqbal University College, 51001 Hilla, Iraq
| | | | - Farag M A Altalbawy
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza 12613, Egypt
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - Abed J Kadhim
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq
| | - Ali Hashiem Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna 66002, Iraq
| | - Muath Suliman
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia.
| | - Ahmed Hussien R Abbas
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Iraq
| |
Collapse
|
37
|
Kikuchi Y, Yamaguchi K, Shimizu R, Matsumoto Y, Kurose Y, Okano N, Otsuka Y, Shibuya K, Matsuda T, Shimada H. Intrahepatic cholangiocarcinoma with FGFR2 fusion gene positive that responded to pemigatinib and caused hypophosphatemia. Int Cancer Conf J 2023; 12:285-290. [PMID: 37577338 PMCID: PMC10421826 DOI: 10.1007/s13691-023-00619-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 06/15/2023] [Indexed: 08/15/2023] Open
Abstract
Intrahepatic cholangiocarcinoma is a condition with a poor prognosis. Traditionally, there was no cure unless important drugs such as gemcitabine, cisplatin, and tegafur/gimeracil/uracil potassium showed efficacy. Pemigatinib has recently become accessible for the treatment of intrahepatic cholangiocarcinoma with FGFR2 fusion or rearrangement gene abnormalities. Hyperphosphatemia is typically linked to pemigatinib. In the current case, pemigatinib was used to effectively treat a 48-year-old woman, and hypophosphatemia was observed. Patients with intrahepatic cholangiocarcinoma should undergo aggressive cancer multigene panel testing as well as careful monitoring of serum phosphorus levels.
Collapse
Affiliation(s)
- Yoshinori Kikuchi
- Department of Clinical Oncology, Toho University, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541 Japan
| | - Kazuhisa Yamaguchi
- Division of Gastroenterology and Hepatology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Ryo Shimizu
- Division of Gastroenterology and Hepatology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Yuu Matsumoto
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| | - Yasuko Kurose
- Department of Surgical Pathology (Omori), Toho University, Tokyo, Japan
| | - Naoki Okano
- Division of Gastroenterology and Hepatology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Yuichirou Otsuka
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| | - Kazutoshi Shibuya
- Department of Surgical Pathology (Omori), Toho University, Tokyo, Japan
| | - Takahisa Matsuda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine (Omori), Toho University, Tokyo, Japan
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, 6-11-1 Omori-nishi, Ota-ku, Tokyo, 143-8541 Japan
| |
Collapse
|
38
|
Veniaminova NA, Jia YY, Hartigan AM, Huyge TJ, Tsai SY, Grachtchouk M, Nakagawa S, Dlugosz AA, Atwood SX, Wong SY. Distinct mechanisms for sebaceous gland self-renewal and regeneration provide durability in response to injury. Cell Rep 2023; 42:113121. [PMID: 37715952 PMCID: PMC10591672 DOI: 10.1016/j.celrep.2023.113121] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/01/2023] [Accepted: 08/25/2023] [Indexed: 09/18/2023] Open
Abstract
Sebaceous glands (SGs) release oils that protect our skin, but how these glands respond to injury has not been previously examined. Here, we report that SGs are largely self-renewed by dedicated stem cell pools during homeostasis. Using targeted single-cell RNA sequencing, we uncovered both direct and indirect paths by which resident SG progenitors ordinarily differentiate into sebocytes, including transit through a Krt5+PPARγ+ transitional basal cell state. Upon skin injury, however, SG progenitors depart their niche, reepithelialize the wound, and are replaced by hair-follicle-derived stem cells. Furthermore, following targeted genetic ablation of >99% of SGs from dorsal skin, these glands unexpectedly regenerate within weeks. This regenerative process is mediated by alternative stem cells originating from the hair follicle bulge, is dependent upon FGFR2 signaling, and can be accelerated by inducing hair growth. Altogether, our studies demonstrate that stem cell plasticity promotes SG durability following injury.
Collapse
Affiliation(s)
- Natalia A Veniaminova
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yunlong Y Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Adrien M Hartigan
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas J Huyge
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shih-Ying Tsai
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marina Grachtchouk
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seitaro Nakagawa
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA; Department of Dermatology, Department of Cutaneous Immunology and Microbiology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Andrzej A Dlugosz
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA.
| | - Sunny Y Wong
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA.
| |
Collapse
|
39
|
Pace A, Scirocchi F, Napoletano C, Zizzari IG, Po A, Megiorni F, Asquino A, Pontecorvi P, Rahimi H, Marchese C, Ferretti E, Nuti M, Rughetti A. Targeting FGFRs by pemigatinib induces G1 phase cell cycle arrest, cellular stress and upregulation of tumor suppressor microRNAs. J Transl Med 2023; 21:626. [PMID: 37715207 PMCID: PMC10504800 DOI: 10.1186/s12967-023-04450-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 08/18/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Fibroblast growth factor receptor (FGFR) gene family alterations are found in several cancers, indicating their importance as potential therapeutic targets. The FGFR-tyrosine kinase inhibitor (TKI) pemigatinib has been introduced in the treatment of advanced cholangiocarcinoma and more recently for relapsed or refractory myeloid/lymphoid neoplasms with FGFR2 and FGFR1 rearrangements, respectively. Several clinical trials are currently investigating the possible combination of pemigatinib with immunotherapy. In this study, we analyzed the biological and molecular effects of pemigatinib on different cancer cell models (lung, bladder, and gastric), which are currently objective of clinical trial investigations. METHODS NCI-H1581 lung, KATO III gastric and RT-112 bladder cancer cell lines were evaluated for FGFR expression by qRT-PCR and Western blot. Cell lines were treated with Pem and then characterized for cell proliferation, apoptosis, production of intracellular reactive oxygen species (ROS), and induction of senescence. The expression of microRNAs with tumor suppressor functions was analyzed by qRT-PCR, while modulation of the proteins coded by their target genes was evaluated by Western blot and mRNA. Descriptive statistics was used to analyze the various data and student's t test to compare the analysis of two groups. RESULTS Pemigatinib exposure triggered distinct signaling pathways and reduced the proliferative ability of all cancer cells, inducing G1 phase cell cycle arrest and strong intracellular stress resulting in ROS production, senescence and apoptosis. Pemigatinib treatment also caused the upregulation of microRNAs (miR-133b, miR-139, miR-186, miR-195) with tumor suppressor functions, along with the downregulation of validated protein targets with oncogenic roles (c-Myc, c-MET, CDK6, EGFR). CONCLUSIONS These results contribute to clarifying the biological effects and molecular mechanisms mediated by the anti-FGFR TKI pemigatinib in distinct tumor settings and support its exploitation for combined therapies.
Collapse
Affiliation(s)
- Angelica Pace
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Fabio Scirocchi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Chiara Napoletano
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy.
| | | | - Agnese Po
- Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Francesca Megiorni
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Angela Asquino
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Paola Pontecorvi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Hassan Rahimi
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Cinzia Marchese
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Elisabetta Ferretti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Marianna Nuti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| | - Aurelia Rughetti
- Department of Experimental Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
40
|
Wekking D, Pretta A, Martella S, D'Agata AP, Joeun Choe J, Denaro N, Solinas C, Scartozzi M. Fibroblast growth factor receptors as targets for anticancer therapy in cholangiocarcinomas and urothelial carcinomas. Heliyon 2023; 9:e19541. [PMID: 37681152 PMCID: PMC10481293 DOI: 10.1016/j.heliyon.2023.e19541] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/16/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023] Open
Abstract
Cholangiocarcinomas and urothelial carcinomas are lethal tumors worldwide and only a minority of patients are eligible for surgery at diagnosis. Moreover, patients are poorly responsive to current therapeutic strategies, including chemotherapy, radiotherapy, immunotherapy, and multimodality treatments. Recently, several advances have been made in precision medicine and these results are modifying the treatment paradigm for patients diagnosed with cholangiocarcinomas and urothelial carcinoma. These histotypes exhibit a high rate of multiple fibroblast growth factor receptor (FGFR) genetic alterations and numerous preclinical and clinical studies support FGFR as a highly attractive novel therapeutic target. Moreover, identifying specific genetic alterations may predict the tumor's response to conventional and novel FGFR-targeted drugs. Recent clinical studies showed promising data for FGFR-targeted therapy in reducing tumor volume and led to the United States Food and Drug Administration (FDA) approval of, e.g., pemigatinib, infigratinib, futibatinib, and erdafitinib. Moreover, FGFR inhibitors show promising results in the first-line setting of cholangiocarcinomas and urothelial carcinomas. Pemigatinib (FIGHT-302) and futibatinib (FOENIX-CAA3) are being evaluated in phase III trials that compare these agents to current first-line gemcitabine and cisplatin in FGFR2-rearranged cholangiocarcinoma. However, complexity in targeting the FGFR signaling pathway is observed. Herein, we describe the characteristics of the FDA-approved and other investigational FGFR-targeted therapeutics, evaluate the most recent preclinical and clinical studies focusing on targeting FGFR genomic alterations in the treatment of cholangiocarcinomas and urothelial cancer, and provide insight into factors involved in response and (acquired) resistance to FGFR inhibition.
Collapse
Affiliation(s)
- Demi Wekking
- Amsterdam UMC, Location Academic Medical Centre, University of Amsterdam, Amsterdam, the Netherlands
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Italy
| | - Serafina Martella
- Medical Oncology, University Hospital Policlinico G.Rodolico-San Marco, 95123, Catania, Italy
| | | | - Joanna Joeun Choe
- Cancer Outcomes Research and Education, Massachusetts General Hospital, Boston, MA, USA
| | | | - Cinzia Solinas
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato, CA, Italy
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Italy
- Medical Oncology AOU Cagliari Policlinico Duilio Casula, Monserrato, CA, Italy
| |
Collapse
|
41
|
Wadsley J, Christie A, Gillmore R, Trinh A, Greig R. Clinical experience with pemigatinib for previously treated metastatic cholangiocarcinoma: practical considerations from clinical cases. Drugs Context 2023; 12:2023-5-5. [PMID: 37602357 PMCID: PMC10435267 DOI: 10.7573/dic.2023-5-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/13/2023] [Indexed: 08/22/2023] Open
Abstract
The management of advanced cholangiocarcinoma (CCA) is challenging. In patients with advanced CCA, gemcitabine/cisplatin combination is the standard frontline chemotherapy, with 5-fluorouracil-based regimens preserved for subsequent lines; however, the expected survival is poor. Pemigatinib was approved for locally advanced or metastatic CCA with FGFR2 fusions or rearrangement. Pemigatinib has a manageable safety profile and achieves a durable response. Nearly 50 patients with CCA have been treated with pemigatinib in the United Kingdom. However, clinical experience with pemigatinib is lacking. We present our experience with three clinical cases to illustrate the position of pemigatinib in the management of CCA and related toxicities.
Collapse
Affiliation(s)
| | - Alan Christie
- Edinburgh Cancer Centre, Western General Hospital, Edinburgh, UK
| | | | - Amy Trinh
- Department of Oncology, Royal Free Hospital, London, UK
| | | |
Collapse
|
42
|
Bader A, Begemann M, Al-Obaidi A, Habib MH, Anwer F, Raza S. Ocular complications of antineoplastic therapies. Future Sci OA 2023; 9:FSO871. [PMID: 37485446 PMCID: PMC10357395 DOI: 10.2144/fsoa-2022-0081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/15/2023] [Indexed: 07/25/2023] Open
Abstract
Ocular complications of antineoplastic agents can have a profound effect on the quality of life of cancer patients. New oncologic treatments like monoclonal antibodies, immunotherapies, antibody-drug conjugates, checkpoint inhibitors and growth factor receptors have resulted in increased ocular complications. These ocular complications differs in respect to distinct mechanisms of actions and lead to significant challenges in the management of cancer patients. In this review, we reviewed literature, clinical studies and cases detailing ocular complications due to administration of antineoplastic agents and emphasized the need for communication between oncologists and ophthalmologists toward early detection and management of ocular complications.
Collapse
Affiliation(s)
- Abbas Bader
- University of Missouri Kansas City School of Medicine, 5000 Holmes St, Kansas City, MO 64110, USA
| | - Madeline Begemann
- Saint Luke's Hospital of Kansas City, 4401 Wornall Rd, Kansas City, MO 64111, USA
| | - Ammar Al-Obaidi
- Saint Luke's Hospital of Kansas City, 4401 Wornall Rd, Kansas City, MO 64111, USA
| | - Muhammad Hamza Habib
- Rutgers Cancer Institute of New Jersey, 195 Little Albany St, New Brunswick, NJ 08901, USA
| | - Faiz Anwer
- Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Shahzad Raza
- Cleveland Clinic Taussig Cancer Institute, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| |
Collapse
|
43
|
Hung KL, Jones MG, Wong ITL, Lange JT, Luebeck J, Scanu E, He BJ, Brückner L, Li R, González RC, Schmargon R, Dörr JR, Belk JA, Bafna V, Werner B, Huang W, Henssen AG, Mischel PS, Chang HY. Coordinated inheritance of extrachromosomal DNA species in human cancer cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.18.549597. [PMID: 37503111 PMCID: PMC10371175 DOI: 10.1101/2023.07.18.549597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The chromosomal theory of inheritance has dominated human genetics, including cancer genetics. Genes on the same chromosome segregate together while genes on different chromosomes assort independently, providing a fundamental tenet of Mendelian inheritance. Extrachromosomal DNA (ecDNA) is a frequent event in cancer that drives oncogene amplification, dysregulated gene expression and intratumoral heterogeneity, including through random segregation during cell division. Distinct ecDNA sequences, herein termed ecDNA species, can co-exist to facilitate intermolecular cooperation in cancer cells. However, how multiple ecDNA species within a tumor cell are assorted and maintained across somatic cell generations to drive cancer cell evolution is not known. Here we show that cooperative ecDNA species can be coordinately inherited through mitotic co-segregation. Imaging and single-cell analyses show that multiple ecDNAs encoding distinct oncogenes co-occur and are correlated in copy number in human cancer cells. EcDNA species are coordinately segregated asymmetrically during mitosis, resulting in daughter cells with simultaneous copy number gains in multiple ecDNA species prior to any selection. Computational modeling reveals the quantitative principles of ecDNA co-segregation and co-selection, predicting their observed distributions in cancer cells. Finally, we show that coordinated inheritance of ecDNAs enables co-amplification of specialized ecDNAs containing only enhancer elements and guides therapeutic strategies to jointly deplete cooperating ecDNA oncogenes. Coordinated inheritance of ecDNAs confers stability to oncogene cooperation and novel gene regulatory circuits, allowing winning combinations of epigenetic states to be transmitted across cell generations.
Collapse
Affiliation(s)
- King L. Hung
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Matthew G. Jones
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Ivy Tsz-Lo Wong
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Joshua T. Lange
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Elisa Scanu
- Department of Mathematics, Queen Mary University of London, London, UK
| | - Britney Jiayu He
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Lotte Brückner
- Max-Delbrück-Centrum für Molekulare Medizin (BIMSB/BIH), Berlin, Germany
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
| | - Rui Li
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Rocío Chamorro González
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Rachel Schmargon
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Jan R. Dörr
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Julia A. Belk
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California at San Diego, La Jolla, CA, 92093, USA
| | - Benjamin Werner
- Evolutionary Dynamics Group, Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Weini Huang
- Department of Mathematics, Queen Mary University of London, London, UK
- Group of Theoretical Biology, The State Key Laboratory of Biocontrol, School of Life Science, Sun Yat-sen University, Guangzhou, China
| | - Anton G. Henssen
- Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine and Charité—Universitätsmedizin Berlin, Lindenberger Weg 80, 13125, Berlin, Germany
- Department of Pediatric Oncology/Hematology, Charité—Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, and German Cancer Research Center DKFZ, Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
- Berlin Institute of Health, Anna-Louisa-Karsch-Str. 2, 10178, Berlin, Germany
| | - Paul S. Mischel
- Sarafan ChEM-H, Stanford University, Stanford, CA, USA
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
44
|
Lu T, Li T, Wu MK, Zheng CC, He XM, Zhu HL, Li L, Man RJ. Molecular simulations required to target novel and potent inhibitors of cancer invasion. Expert Opin Drug Discov 2023; 18:1367-1377. [PMID: 37676052 DOI: 10.1080/17460441.2023.2254695] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 08/30/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Computer-aided drug design (CADD) is a computational approach used to discover, develop, and analyze drugs and active molecules with similar biochemical properties. Molecular simulation technology has significantly accelerated drug research and reduced manufacturing costs. It is an optimized drug discovery method that greatly improves the efficiency of novel drug development processes. AREASCOVERED This review discusses the development of molecular simulations of effective cancer inhibitors and traces the main outcomes of in silico studies by introducing representative categories of six important anticancer targets. The authors provide views on this topic from the perspective of both medicinal chemistry and artificial intelligence, indicating the major challenges and predicting trends. EXPERT OPINION The goal of introducing CADD into cancer treatment is to realize a highly efficient, accurate, and desired approach with a high success rate for identifying potent drug candidates. However, the major challenge is the lack of a sophisticated data-filtering mechanism to verify bottom data from mixed-quality references. Consequently, despite the continuous development of algorithms, computer power, and interface optimization, specific data filtering mechanisms will become an urgent and crucial issue in the future.
Collapse
Affiliation(s)
| | - Tong Li
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Meng-Ke Wu
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Chi-Chong Zheng
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| | - Xue-Mei He
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Science, Nanning, China
| | - Hai-Liang Zhu
- School of Life Sciences, Nanjing University, Nanjing, China
| | - Li Li
- Agro-food Science and Technology Research Institute, Guangxi Academy of Agricultural Science, Nanning, China
| | - Ruo-Jun Man
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, Guangxi University for Nationalities, Nanning, China
| |
Collapse
|
45
|
Uson PLS, Bearss J, Babiker HM, Borad M. A drug safety evaluation of pemigatinib for advanced cholangiocarcinoma. Expert Opin Drug Saf 2023; 22:637-641. [PMID: 37363820 DOI: 10.1080/14740338.2023.2227561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
INTRODUCTION Pemigatinib is a selective small-molecule inhibitor of the fibroblast growth factor receptor (FGFR) 1-3. FGFR is associated with increased cell division, proliferation, and survival. Inhibition of this receptor is an effective treatment against tumors driven by activated fusions in FGFR2. AREAS COVERED The drug was first evaluated in patients with advanced solid tumors and demonstrated a manageable safety profile, with the most common adverse events being oscillations in blood phosphate levels, fatigue, gastrointestinal symptoms, and skin and ocular toxicities. Pemigatinib was further evaluated in a phase II cohort study of patients with previously treated locally advanced or metastatic cholangiocarcinoma harboring FGFR2 genomic alterations. After a median follow-up of 17.8 months, the objective response rate in patients with tumors harboring FGFR2 fusions or rearrangements was 35.5% (95% CI, 26.5-45.4). Based on these results, the FDA granted accelerated approval on 17 April 2020, to pemigatinib, for the treatment of adults with previously treated, unresectable locally advanced or metastatic cholangiocarcinoma with an FGFR2 fusion or another rearrangement. Articles selected for this review were based on reported studies indexed in PubMed (2010-2023). EXPERT OPINION Future perspectives in the treatment of FGFR2 fused cholangiocarcinoma include the evaluation of pemigatinib in previously untreated patients and possible active combinations or sequencing strategies with other drugs.
Collapse
Affiliation(s)
- Pedro Luiz Serrano Uson
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
- Center for Personalized Medicine, Hospital Israelita Albert Einstein, Sao Paulo, SP, Brazil
| | | | - Hani M Babiker
- Department of Oncology, Mayo Clinic Jacksonville, Florida, USA
| | - Mitesh Borad
- Mayo Clinic Cancer Center, Mayo Clinic, Phoenix, AZ, USA
| |
Collapse
|
46
|
Zhang L, Zheng H, Xu L, You S, Shen Y, Han Y, Anderson S. A Robust FISH Assay to Detect FGFR2 Translocations in Intrahepatic Cholangiocarcinoma Patients. Diagnostics (Basel) 2023; 13:2088. [PMID: 37370984 DOI: 10.3390/diagnostics13122088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
FGFR fusions retaining the FGFR kinase domain are active kinases that are either overexpressed or constitutively activated throughout diverse cancer types. The presence of FGFR translocations enhances tumor cell proliferation and contributes to significant sensitivity to FGFR kinase inhibitors. FGFR2 as an actionable target in intrahepatic cholangiocarcinoma (iCCA) has been tested in many clinical trials. FISH (fluorescence in situ hybridization) and NGS (next-generation sequence) are well-known tools to investigate the translocations of FGFR with multiple or unknown translocation partners. A rapid and robust FISH assay was developed and validated to detect FGFR2 translocations from FFPE specimens in iCCA. The analytical performance of the FISH assay was evaluated for probe localization, probe sensitivity and specificity, and assay precision. Twenty-five archival FFPE specimens from local iCCA patients were tested for FGFR2 translocations. FISH results were correlated with that of NGS on some samples. Biallelic translocations and a novel FGFR2 translocation involving the partner gene, SHROOM3, t(4;10) (q21;q26), were identified in a local iCCA patient.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Hao Zheng
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Linyu Xu
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Si You
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Yuanyuan Shen
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Yang Han
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| | - Steve Anderson
- Department of Anatomic Pathology and Histology, Central Laboratory Service, Labcorp Drug Development, 8211 Scicor Dr, Indianapolis, IN 46214, USA
| |
Collapse
|
47
|
Veniaminova NA, Jia Y, Hartigan AM, Huyge TJ, Tsai SY, Grachtchouk M, Nakagawa S, Dlugosz AA, Atwood SX, Wong SY. Distinct mechanisms for sebaceous gland self-renewal and regeneration provide durability in response to injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.05.539454. [PMID: 37205445 PMCID: PMC10187279 DOI: 10.1101/2023.05.05.539454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Sebaceous glands (SGs) release oils that protect our skin, but how these glands respond to injury has not been previously examined. Here, we report that SGs are largely self-renewed by dedicated stem cell pools during homeostasis. Using targeted single cell RNA-sequencing, we uncovered both direct and indirect paths by which these resident SG progenitors ordinarily differentiate into sebocytes, including transit through a PPARγ+Krt5+ transitional cell state. Upon skin injury, however, SG progenitors depart their niche, reepithelialize the wound, and are replaced by hair follicle-derived stem cells. Furthermore, following targeted genetic ablation of >99% of SGs from dorsal skin, these glands unexpectedly regenerate within weeks. This regenerative process is mediated by alternative stem cells originating from the hair follicle bulge, is dependent upon FGFR signaling, and can be accelerated by inducing hair growth. Altogether, our studies demonstrate that stem cell plasticity promotes SG durability following injury.
Collapse
Affiliation(s)
- Natalia A. Veniaminova
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yunlong Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Adrien M. Hartigan
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Thomas J. Huyge
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Shih-Ying Tsai
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marina Grachtchouk
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Seitaro Nakagawa
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrzej A. Dlugosz
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Scott X. Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Sunny Y. Wong
- Department of Dermatology, Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
- Lead Contact:
| |
Collapse
|
48
|
Shomali W, Colucci P, George TI, Kiladjian JJ, Langford C, Patel JL, Reiter A, Vannucchi AM, Gotlib J. Comprehensive response criteria for myeloid/lymphoid neoplasms with eosinophilia and tyrosine kinase gene fusions: a proposal from the MLN International Working Group. Leukemia 2023; 37:981-987. [PMID: 37076693 PMCID: PMC10169632 DOI: 10.1038/s41375-023-01859-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/20/2022] [Accepted: 02/21/2023] [Indexed: 04/21/2023]
Affiliation(s)
- William Shomali
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | | | - Tracy I George
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | - Cheryl Langford
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA
| | - Jay L Patel
- ARUP Laboratories and University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | - Jason Gotlib
- Division of Hematology, Stanford Cancer Institute/Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
49
|
McSheehy PMJ, Forster-Gross N, El Shemerly M, Bachmann F, Roceri M, Hermann N, Spickermann J, Kellenberger L, Lane HA. The fibroblast growth factor receptor inhibitor, derazantinib, has strong efficacy in human gastric tumor models and synergizes with paclitaxel in vivo. Anticancer Drugs 2023; 34:532-543. [PMID: 36729959 DOI: 10.1097/cad.0000000000001469] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Derazantinib (DZB) is an inhibitor of fibroblast growth factor receptors 1-3 (FGFR1-3), with additional activity against colony-stimulating-factor-1 receptor (CSF1R). We have profiled the activity of DZB in gastric cancer (GC) as monotherapy and combined with paclitaxel, and explored means of stratifying patients for treatment. The antiproliferative potency of DZB in vitro was quantified in 90 tumor cell lines and shown to correlate significantly with FGFR expression (<0.01) but not with FGFR DNA copy-number (CN) or FGFR mutations. In four GC cell lines in vitro , little or no synergy was observed with paclitaxel. In athymic nude mice, bearing cell-line derived xenografts (CDX) or patient-derived xenograft (PDX) GC models, DZB efficacy correlated highly significantly with FGFR gene expression ( r2 = 0.58; P = 0.0003; n = 18), but not FGFR mutations or DNA-CN. In FGFR-driven GC models, DZB had comparable efficacy to three other FGFR inhibitors and was more efficacious than paclitaxel. DZB had dose-dependent plasma pharmacokinetics but showed low brain penetration at all doses. GC models (one CDX and six PDX) were tested for sensitivity to the combination of DZB and paclitaxel and characterized by immunohistochemistry. The combination showed synergy (5) or additivity (2), and no antagonism, with synergy significantly associated ( P < 0.05) with higher levels of M2-type macrophages. The association of strong efficacy of the combination in vivo with M2 macrophages, which are known to express CSF1R, and the absence of synergy in vitro is consistent with the tumor microenvironment also being a factor in DZB efficacy and suggests additional means by which DZB could be stratified for cancer treatment in the clinic.
Collapse
|
50
|
Mahapatra S, Jonniya NA, Koirala S, Ursal KD, Kar P. The FGF/FGFR signalling mediated anti-cancer drug resistance and therapeutic intervention. J Biomol Struct Dyn 2023; 41:13509-13533. [PMID: 36995019 DOI: 10.1080/07391102.2023.2191721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/26/2023] [Indexed: 03/31/2023]
Abstract
Fibroblast Growth Factor (FGF) ligands and their receptors are crucial factors driving chemoresistance in several malignancies, challenging the efficacy of currently available anti-cancer drugs. The Fibroblast growth factor/receptor (FGF/FGFR) signalling malfunctions in tumor cells, resulting in a range of molecular pathways that may impact its drug effectiveness. Deregulation of cell signalling is critical since it can enhance tumor growth and metastasis. Overexpression and mutation of FGF/FGFR induce regulatory changes in the signalling pathways. Chromosomal translocation facilitating FGFR fusion production aggravates drug resistance. Apoptosis is inhibited by FGFR-activated signalling pathways, reducing multiple anti-cancer medications' destructive impacts. Angiogenesis and epithelial-mesenchymal transition (EMT) are facilitated by FGFRs-dependent signalling, which correlates with drug resistance and enhances metastasis. Further, lysosome-mediated drug sequestration is another prominent method of resistance. Inhibition of FGF/FGFR by following a plethora of therapeutic approaches such as covalent and multitarget inhibitors, ligand traps, monoclonal antibodies, recombinant FGFs, combination therapy, and targeting lysosomes and micro RNAs would be helpful. As a result, FGF/FGFR suppression treatment options are evolving nowadays. To increase positive impacts, the processes underpinning the FGF/FGFR axis' role in developing drug resistance need to be clarified, emphasizing the need for more studies to develop novel therapeutic options to address this significant problem. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Subhasmita Mahapatra
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Nisha Amarnath Jonniya
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Suman Koirala
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Kapil Dattatray Ursal
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| | - Parimal Kar
- Department of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Indore, Madhya Pradesh, India
| |
Collapse
|