1
|
Lu S, Cao C, Zhang W, Li J, Yang J, Huang Z, Wu Z, Liu B, Huang H, Wang H, Wang Y, Liu D, Zhang Z, Liu K, Yang G, Gong X, Dai H, Li Y, Dong E, Zhang X, Zhang Y. Peficitinib suppresses diffuse-type tenosynovial giant cell tumor by targeting TYK2 and JAK/STAT signaling. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2790-7. [PMID: 39808223 DOI: 10.1007/s11427-024-2790-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/21/2024] [Indexed: 01/16/2025]
Abstract
Diffuse-type tenosynovial giant cell tumor (dTGCT) is a destructive but rare benign proliferative synovial neoplasm. Although surgery is currently the main treatment modality for dTGCT, the recurrence risk is up to 50%. Therefore, there is a great need for effective drugs against dTGCT with minor side effects. The Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling plays a central role in rheumatoid arthritis (RA), a disease with similar characteristics as dTGCT, but its function in dTGCT remains unknown. dTGCT fibroblast-like synoviocytes (FLS) and macrophages were isolated from 10 synovial tissue samples from dTGCT patients for the screening and validation of the five clinically approved JAK inhibitors to treat RA against dTGCT. Cell viability, cell death, inflammation and the activity of the JAK family members of cultured dTGCT FLS (both 2-D and 3-D) and macrophages were investigated for the efficacy of the JAK inhibitors. Here, we found that similar to RA, JAK/STAT signaling was markedly activated in the dTGCT synovium. Of the 5 JAK inhibitors, peficitinib was shown to have the most potency in addressing some of the pathological responses of dTGCT FLS and macrophages. The potency of peficitinib was much higher than pexidartinib, which is the only FDA-approved drug for dTGCT. Mechanistically, peficitinib inhibited tyrosine kinase 2 (TYK2), a JAK family member necessary for the pathological progression of dTGCT FLS and macrophages. In summary, we not only revealed JAK/STAT (especially TYK2) signaling as the major mechanism underlying dTGCT, but also identified peficitinib as a promising drug against dTGCT.
Collapse
Affiliation(s)
- Shan Lu
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Chenxi Cao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Wenjia Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Jiayi Li
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing, 100871, China
| | - Jingli Yang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Zisheng Huang
- Peking University Health Science Centre, Peking University, Beijing, 100871, China
| | - Zhijun Wu
- Peking University Health Science Centre, Peking University, Beijing, 100871, China
| | - Baitao Liu
- Peking University Health Science Centre, Peking University, Beijing, 100871, China
| | - Hongjie Huang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Haijun Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Yongjian Wang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Dingge Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Zhihua Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Kaiping Liu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Gang Yang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Xi Gong
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China
| | - Hui Dai
- Department of Immunology, School of Basic Medical Sciences, NHC Key Laboratory of Medical Immunology, Peking University, Beijing, 100871, China
| | - Yingjia Li
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
| | - Erdan Dong
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
- Research Unit of Medical Science Research Management/Basic and Clinical Research of Metabolic Cardiovascular Diseases, Chinese Academy of Medical Sciences, Haihe Laboratory of Cell Ecosystem, Beijing, 100191, China.
| | - Xin Zhang
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Sports Injuries, Beijing, 100191, China.
- Engineering Research Center of Sports Trauma Treatment Technology and Devices, Ministry of Education, Beijing, 100191, China.
| | - Yan Zhang
- Institute of Cardiovascular Sciences, School of Basic Medical Sciences, Peking University Health Science Center; Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| |
Collapse
|
2
|
Gelderblom H, Bhadri V, Stacchiotti S, Bauer S, Wagner AJ, van de Sande M, Bernthal NM, López Pousa A, Razak AA, Italiano A, Ahmed M, Le Cesne A, Tinoco G, Boye K, Martín-Broto J, Palmerini E, Tafuto S, Pratap S, Powers BC, Reichardt P, Casado Herráez A, Rutkowski P, Tait C, Zarins F, Harrow B, Sharma MG, Ruiz-Soto R, Sherman ML, Blay JY, Tap WD. Vimseltinib versus placebo for tenosynovial giant cell tumour (MOTION): a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2024; 403:2709-2719. [PMID: 38843860 PMCID: PMC11740396 DOI: 10.1016/s0140-6736(24)00885-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND Tenosynovial giant cell tumour (TGCT) is a locally aggressive neoplasm for which few systemic treatment options exist. This study evaluated the efficacy and safety of vimseltinib, an oral, switch-control, CSF1R inhibitor, in patients with symptomatic TGCT not amenable to surgery. METHODS MOTION is a multicentre, randomised, double-blind, placebo-controlled, phase 3 trial done in 35 specialised hospitals in 13 countries. Eligible patients were adults (aged ≥18 years) with a histologically confirmed diagnosis of TGCT for which surgical resection could potentially worsen functional limitation or cause severe morbidity. Patients were randomly assigned (2:1) with interactive response technology to vimseltinib (30 mg orally twice weekly) or placebo, administrated in 28-day cycles for 24 weeks. Patients and site personnel were masked to treatment assignment until week 25, unless progressive disease was confirmed earlier. The primary endpoint was objective response rate by independent radiological review using Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST) at week 25 in the intention-to-treat population. Safety was assessed in all patients who received the study drug. The trial is registered with ClinicalTrials.gov, NCT05059262, and enrolment is complete. FINDINGS Between Jan 21, 2022, and Feb 21, 2023, 123 patients were randomly assigned (83 to vimseltinib and 40 to placebo). 73 (59%) patients were female and 50 (41%) were male. Nine (11%) of 83 patients assigned to vimseltinib and five (13%) of 40 patients assigned to placebo discontinued treatment before week 25; one patient in the placebo group did not receive any study drug. Objective response rate per RECIST was 40% (33 of 83 patients) in the vimseltinib group vs 0% (none of 40) in the placebo group (difference 40% [95% CI 29-51]; p<0·0001). Most treatment-emergent adverse events (TEAEs) were grade 1 or 2; the only grade 3 or 4 TEAE that occurred in more than 5% of patients receiving vimseltinib was increased blood creatine phosphokinase (eight [10%] of 83). One patient in the vimseltinib group had a treatment-related serious TEAE of subcutaneous abscess. No evidence of cholestatic hepatotoxicity or drug-induced liver injury was noted. INTERPRETATION Vimseltinib produced a significant objective response rate and clinically meaningful functional and symptomatic improvement in patients with TGCT, providing an effective treatment option for these patients. FUNDING Deciphera Pharmaceuticals.
Collapse
Affiliation(s)
- Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands.
| | - Vivek Bhadri
- Department of Medical Oncology, Chris O'Brien Lifehouse, Camperdown, NSW, Australia
| | | | - Sebastian Bauer
- Department of Medical Oncology and Sarcoma Center, West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany; German Cancer Consortium, Partner Site University Hospital Essen, Essen, Germany
| | - Andrew J Wagner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Michiel van de Sande
- Department of Medical Oncology, Leiden University Medical Center, Leiden, Netherlands
| | - Nicholas M Bernthal
- Department of Orthopaedic Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | | | | | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France; University of Bordeaux, Bordeaux, France
| | - Mahbubl Ahmed
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Axel Le Cesne
- Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| | - Gabriel Tinoco
- Department of Internal Medicine, Division of Medical Oncology, The Ohio State University, Columbus, OH, USA
| | - Kjetil Boye
- Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Javier Martín-Broto
- Fundación Jiménez Díaz University Hospital, University Hospital General de Villalba, Instituto de Investigactión Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | | | - Salvatore Tafuto
- Sarcomas and Rare Tumors Unit, Istituto Nazionale Tumori IRCCS Fondazione G Pascale, Naples, Italy
| | - Sarah Pratap
- Oxford Cancer and Haematology Centre, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Benjamin C Powers
- Department of Internal Medicine, Medical Oncology Division, University of Kansas Cancer Center, Overland Park, KS, USA
| | - Peter Reichardt
- Department of Interdisciplinary Oncology, HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | | | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | | | - Fiona Zarins
- Deciphera Pharmaceuticals, LLC, Waltham, MA, USA
| | | | | | | | | | - Jean-Yves Blay
- Department of Medical Oncology, Centre Léon Bérard, Lyon, France
| | - William D Tap
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
3
|
Hwang C, Agulnik M, Schulte B. Prices and Trends in FDA-Approved Medications for Sarcomas. Cancers (Basel) 2024; 16:1545. [PMID: 38672627 PMCID: PMC11048328 DOI: 10.3390/cancers16081545] [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: 03/10/2024] [Revised: 04/04/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Sarcomas represent a diverse set of both malignant and benign subtypes consisting of often rare and ultra-rare conditions. Over the course of the last decade, there have been numerous FDA approvals for agents treating various sarcoma subtypes. Given this burgeoning landscape of sarcoma treatments, we seek to review current FDA-approved agents with respect to their rates of incidence, approval rates, and financial costs. We gathered clinical trial data by searching FDA approval announcements from 2013 to 2023. We determined the 30 day and one year cost of therapy for patients of FDA-approved sarcoma treatments in the aforementioned timeframe. From 2013 to 2023, 14 medications have been FDA-approved for sarcoma subtypes. The 30-day dosing prices for these medications range from $11,162.86 to $46,926.00. Since 2013, the rates of approval for sarcoma medications have been higher than in prior decades. Nonetheless, there remains the potential for significant financial toxicity for patients living with sarcoma.
Collapse
Affiliation(s)
- Caleb Hwang
- School of Medicine, University of California, San Francisco, CA 94143, USA
| | - Mark Agulnik
- Division of Oncology, Keck School of Medicine of USC, Los Angeles, CA 90033, USA
| | - Brian Schulte
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA 94158, USA
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA 94158, USA
| |
Collapse
|
4
|
Hu Y, Nie W, Lyu L, Zhang X, Wang W, Zhang Y, He S, Guo A, Liu F, Wang B, Qian Z, Gao X. Tumor-Microenvironment-Activatable Nanoparticle Mediating Immunogene Therapy and M2 Macrophage-Targeted Inhibitor for Synergistic Cancer Immunotherapy. ACS NANO 2024; 18:3295-3312. [PMID: 38252684 DOI: 10.1021/acsnano.3c10037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Immunotherapy has achieved prominent clinical efficacy in combating cancer and has recently become a mainstream treatment strategy. However, achieving broad efficacy with a single modality is challenging, and the heterogeneity of the tumor microenvironment (TME) restricts the accuracy and effectiveness of immunotherapy strategies for tumors. Herein, a TME-responsive targeted nanoparticle to enhance antitumor immunity and reverse immune escape by codelivering interleukin-12 (IL-12) expressing gene and colony-stimulating factor-1 receptor (CSF-1R) inhibitor PLX3397 (PLX) is presented. The introduction of disulfide bonds and cyclo(Arg-Gly-Asp-d-Phe-Lys) (cRGD) peptides conferred reduction reactivity and tumor targeting to the nanoparticles, respectively. It is hypothesized that activating host immunity by the local expression of IL-12, while modulating the tumor-associated macrophages (TAM) function through blocking CSF-1/CSF-1R signaling, could constitute a feasible approach for cancer immunotherapy. The fabricated functional nanoparticle successfully ameliorated the TME by stimulating the proliferation and activation of T lymphocytes, promoting the repolarization of TAMs, reducing myeloid-derived suppressor cells (MDSCs), and promoting the maturation of dendritic cells (DC) as well as the secretion of antitumor cytokines, which efficiently suppressed tumor growth and metastasis. Finally, substantial changes in the TME were deciphered by single-cell analysis including infiltration of different cells, transcriptional states, secretory signaling and cell-cell communications. These findings provide a promising combinatorial immunotherapy strategy through immunomodulatory nanoparticles.
Collapse
Affiliation(s)
- Yuzhu Hu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Wen Nie
- Department of Radiation Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Liang Lyu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Xifeng Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Wanyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Yunchu Zhang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Shi He
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Anjie Guo
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Fei Liu
- Department of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bilan Wang
- Department of Pharmacy, West China Second University Hospital of Sichuan University, Chengdu 610041, P. R. China
| | - Zhiyong Qian
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy, West China Hospital, West China Medical School, Sichuan University, Chengdu, 610041, PR China
| |
Collapse
|
5
|
Alkubaisi BO, Aljobowry R, Ali SM, Sultan S, Zaraei SO, Ravi A, Al-Tel TH, El-Gamal MI. The latest perspectives of small molecules FMS kinase inhibitors. Eur J Med Chem 2023; 261:115796. [PMID: 37708796 DOI: 10.1016/j.ejmech.2023.115796] [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: 07/03/2023] [Revised: 09/03/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
FMS kinase is a type III tyrosine kinase receptor that plays a central role in the pathophysiology and management of several diseases, including a range of cancer types, inflammatory disorders, neurodegenerative disorders, and bone disorders among others. In this review, the pathophysiological pathways of FMS kinase in different diseases and the recent developments of its monoclonal antibodies and inhibitors during the last five years are discussed. The biological and biochemical features of these inhibitors, including binding interactions, structure-activity relationships (SAR), selectivity, and potencies are discussed. The focus of this article is on the compounds that are promising leads and undergoing advanced clinical investigations, as well as on those that received FDA approval. In this article, we attempt to classify the reviewed FMS inhibitors according to their core chemical structure including pyridine, pyrrolopyridine, pyrazolopyridine, quinoline, and pyrimidine derivatives.
Collapse
Affiliation(s)
- Bilal O Alkubaisi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Raya Aljobowry
- College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Salma M Ali
- College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Sara Sultan
- College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Seyed-Omar Zaraei
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Anil Ravi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Taleb H Al-Tel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| | - Mohammed I El-Gamal
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; College of Pharmacy, University of Sharjah, Sharjah, 27272, United Arab Emirates; Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| |
Collapse
|
6
|
Wang Q, Wang YY, Pu WJ, Ma X, Ni RJ. Dynamic changes in microglia in the mouse hippocampus during administration and withdrawal of the CSF1R inhibitor PLX3397. J Anat 2023; 243:394-403. [PMID: 37038887 PMCID: PMC10439370 DOI: 10.1111/joa.13874] [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: 07/21/2022] [Revised: 02/01/2023] [Accepted: 03/28/2023] [Indexed: 04/12/2023] Open
Abstract
Pexidartinib (PLX3397), a colony-stimulating factor-1 receptor (CSF1R) inhibitor, is currently in phase 1-3 clinical trials as a treatment for a variety of tumours. CSF1R signalling regulates the development, survival and maintenance of microglia, the resident brain innate immune cells. In this study, we examined the effects of PLX3397 in the drinking water of mice on microglia in the hippocampus using ionized calcium-binding adapter molecule 1 (Iba1, a microglial marker) immunocytochemistry. A high concentration of PLX3397 (1 mg/mL) significantly decreased the density of Iba1-immunoreactive cells after 7 days of exposure, but a low concentration of PLX3397 (0.5 mg/mL) did not. In addition, both low and high concentrations of PLX3397 significantly increased the intersection number, total length and maximum length of microglial processes in male mice. PLX3397 administered for 21 days eliminated microglia with 78% efficiency in males and 84% efficiency in females. Significant increases in microglial processes were found after both seven and 21 days of PLX3397 exposure in males, whereas decreases in microglial processes were observed after both 14 and 21 days of exposure in females. After PLX3397 withdrawal following its administration for 14 days in males, the soma size quickly returned to normal levels within a week. However, the microglial density, intersection number and total length of microglial processes after 3 days of recovery stabilized to untreated levels. In summary, these findings provide detailed insight into the dynamic changes in microglial number and morphology in the hippocampus in a dose- and time-dependent manner after PLX3397 treatment and withdrawal.
Collapse
Affiliation(s)
- Qirun Wang
- Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Yi-Yan Wang
- Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Wen-Jun Pu
- Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Xiaohong Ma
- Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| | - Rong-Jun Ni
- Psychiatric Laboratory and Mental Health Center, West China Hospital, Sichuan University, Chengdu, China
- Sichuan Clinical Medical Research Center for Mental Disorders, Chengdu, China
| |
Collapse
|
7
|
Ebrahimi N, Fardi E, Ghaderi H, Palizdar S, Khorram R, Vafadar R, Ghanaatian M, Rezaei-Tazangi F, Baziyar P, Ahmadi A, Hamblin MR, Aref AR. Receptor tyrosine kinase inhibitors in cancer. Cell Mol Life Sci 2023; 80:104. [PMID: 36947256 PMCID: PMC11073124 DOI: 10.1007/s00018-023-04729-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 01/31/2023] [Accepted: 02/13/2023] [Indexed: 03/23/2023]
Abstract
Targeted therapy is a new cancer treatment approach, involving drugs that particularly target specific proteins in cancer cells, such as receptor tyrosine kinases (RTKs) which are involved in promoting growth and proliferation, Therefore inhibiting these proteins could impede cancer progression. An understanding of RTKs and the relevant signaling cascades, has enabled the development of many targeted drug therapies employing RTK inhibitors (RTKIs) some of which have entered clinical application. Here we discuss RTK structures, activation mechanisms and functions. Moreover, we cover the potential effects of combination drug therapy (including chemotherapy or immunotherapy agents with one RTKI or multiple RTKIs) especially for drug resistant cancers.
Collapse
Affiliation(s)
- Nasim Ebrahimi
- Genetics Division, Department of Cell and Molecular Biology and Microbiology, Faculty of Science and Technology, University of Isfahan, Isfahan, Iran
| | - Elmira Fardi
- Medical Branch, Islamic Azad University of Tehran, Tehran, Iran
| | - Hajarossadat Ghaderi
- Laboratory of Regenerative and Medical Innovation, Pasteur Institute of Iran, Tehran, Iran
| | - Sahar Palizdar
- Division of Microbiology, Faculty of Basic Sciences, Islamic Azad University of Tehran East Branch, Tehran, Iran
| | - Roya Khorram
- Bone and Joint Diseases Research Center, Department of Orthopedic Surgery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Reza Vafadar
- Department of Orthopeadic Surgery, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Ghanaatian
- Master 1 Bio-Santé-Parcours Toulouse Graduate School of Cancer, Ageing and Rejuvenation (CARe), Université Toulouse III-Paul Sabatier, Toulouse, France
| | - Fatemeh Rezaei-Tazangi
- Department of Anatomy, School of Medicine, Fasa University of Medical Sciences, Fasa, Iran
| | - Payam Baziyar
- Department of Molecular and Cell Biology, Faculty of Basic Science, Uinversity of Mazandaran, Babolsar, Iran
| | - Amirhossein Ahmadi
- Department of Biological Science and Technology, Faculty of Nano and Bio Science and Technology, Persian Gulf University, Bushehr, 75169, Iran.
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa.
| | - Amir Reza Aref
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, 02115, USA.
- Translational Medicine Group, Xsphera Biosciences, 6 Tide Street, Boston, MA, 02210, USA.
| |
Collapse
|
8
|
Stacchiotti S, Dürr HR, Schaefer IM, Woertler K, Haas R, Trama A, Caraceni A, Bajpai J, Baldi GG, Bernthal N, Blay JY, Boye K, Broto JM, Chen WWT, Dei Tos PA, Desai J, Emhofer S, Eriksson M, Gronchi A, Gelderblom H, Hardes J, Hartmann W, Healey J, Italiano A, Jones RL, Kawai A, Leithner A, Loong H, Mascard E, Morosi C, Otten N, Palmerini E, Patel SR, Reichardt P, Rubin B, Rutkowski P, Sangalli C, Schuster K, Seddon BM, Shkodra M, Staals EL, Tap W, van de Rijn M, van Langevelde K, Vanhoenacker FMM, Wagner A, Wiltink L, Stern S, Van de Sande VM, Bauer S. Best clinical management of tenosynovial giant cell tumour (TGCT): A consensus paper from the community of experts. Cancer Treat Rev 2023; 112:102491. [PMID: 36502615 DOI: 10.1016/j.ctrv.2022.102491] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022]
Abstract
Tenosynovial giant cell tumour (TGCT) is a rare, locally aggressive, mesenchymal tumor arising from the joints, bursa and tendon sheaths. TGCT comprises a nodular- and a diffuse-type, with the former exhibiting mostly indolent course and the latter a locally aggressive behavior. Although usually not life-threatening, TGCT may cause chronic pain and adversely impact function and quality of life (QoL). CSFR1 inhibitors are effective with benefit on symptoms and QoL but are not available in most countries. The degree of uncertainty in selecting the most appropriate therapy and the lack of guidelines on the clinical management of TGCT make the adoption of new treatments inconsistent across the world, with suboptimal outcomes for patients. A global consensus meeting was organized in June 2022, involving experts from several disciplines and patient representatives from SPAGN to define the best evidence-based practice for the optimal approach to TGCT and generate the recommendations presented herein.
Collapse
Affiliation(s)
- Silvia Stacchiotti
- Department of cancer medicine, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy.
| | - Hans Roland Dürr
- Department of Orthopaedics and Trauma Surgery, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Inga-Marie Schaefer
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, USA
| | - Klaus Woertler
- Department of Radiology, Technische Universität München, Munich, Germany
| | - Rick Haas
- Department of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Annalisa Trama
- Evaluative Epidemiology Unit, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Augusto Caraceni
- High-Complexity Unit of Palliative Care, Pain Therapy and Rehabilitation, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Jyoti Bajpai
- Department of Medical Oncology, Homi Bhabha National Institute, Mumbai, India
| | | | | | - Jean-Yves Blay
- Department of Medical Oncology, Université Centre Léon Bérard, Lyon, France
| | - Kjetil Boye
- Department of Medical Oncology, Oslo University Hospital, Oslo, Norway
| | - Javier-Martin Broto
- Oncology Department, Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Wei-Wu Tom Chen
- Department of Medical Oncology, National Taiwan University Hospital and Cancer Center, Taiwan
| | | | - Jayesh Desai
- Peter MacCallum Cancer Centre/Royal Melbourne Hospital, Melbourne, Australia
| | | | - Mikael Eriksson
- Department of Medical Oncology, LUCC - Lund University Cancer Centre, Lund, Sweden
| | - Alessandro Gronchi
- Department of Surgery, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Hans Gelderblom
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Jendrik Hardes
- Department of Orthopaedic Oncology, Uniklinik Essen, Essen, Germany
| | - Wolfgang Hartmann
- Gerhard-Domagk-Institute for Pathology, Uniklinik Münster, Münster, Germany
| | - John Healey
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York City, USA
| | - Antoine Italiano
- Department of Medical Oncology, Institut Bergonié, Bordeaux, France
| | - Robin L Jones
- Sarcoma Unit, The Royal Marsden, London, United Kingdom
| | - Akira Kawai
- Department of Muscoloskeletal Oncology, National Cancer Center Hospital (NCCH), Tokyo, Japan
| | - Andreas Leithner
- Department of Orthopaedics and Trauma, Medizinische Universität Graz, Graz, Austria
| | - Herbert Loong
- Department of Clinical Oncology, The Chinese University of Hong Kong, Hong Kong
| | - Eric Mascard
- Department of Paediatric Orthopaedic Surgery, Clinique Arago, Paris, France
| | - Carlo Morosi
- Department of Radiology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | | | - Emanuela Palmerini
- Department of Osteoncology, Bone and Soft Tissue Sarcomas and Innovative Therapies, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | | | - Peter Reichardt
- Department of Medical Oncology, Helios Klinikum Berlin-Buch, Berlin, Germany
| | - Brian Rubin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute and Department of Cancer Biology, Cleveland Clinic, Cleveland, USA
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie Memorial Cancer Center and Institute of Oncology, Warsaw, Poland
| | - Claudia Sangalli
- Department of Radiation Oncology, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | | | - Beatrice M Seddon
- Department of Oncology, University College Hospital London, London, United Kingdom
| | - Morena Shkodra
- High-Complexity Unit of Palliative Care, Pain Therapy and Rehabilitation, Fondazione IRCCS Istituto Nazionale Tumori, Milan, Italy
| | - Eric L Staals
- Department of Orthopaedic Surgery, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - William Tap
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York City, USA
| | | | | | | | - Andrew Wagner
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, USA
| | - Lisette Wiltink
- Department of Radiotherapy, The Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Sydney Stern
- Patient Representative, Life Raft Group, and Pharmacokinetics, University of Maryland Baltimore, USA
| | | | - Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, Uniklinik Essen, Essen, Germany
| |
Collapse
|
9
|
van IJzendoorn DG, Matusiak M, Charville GW, Spierenburg G, Varma S, Colburg DR, van de Sande MA, van Langevelde K, Mohler DG, Ganjoo KN, Bui NQ, Avedian RS, Bovée JV, Steffner R, West RB, van de Rijn M. Interactions in CSF1-Driven Tenosynovial Giant Cell Tumors. Clin Cancer Res 2022; 28:4934-4946. [PMID: 36007098 PMCID: PMC9660542 DOI: 10.1158/1078-0432.ccr-22-1898] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/25/2022] [Accepted: 08/23/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE A major component of cells in tenosynovial giant cell tumor (TGCT) consists of bystander macrophages responding to CSF1 that is overproduced by a small number of neoplastic cells with a chromosomal translocation involving the CSF1 gene. An autocrine loop was postulated where the neoplastic cells would be stimulated through CSF1R expressed on their surface. Here, we use single-cell RNA sequencing (scRNA-seq) to investigate cellular interactions in TGCT. EXPERIMENTAL DESIGN A total of 18,788 single cells from three TGCT and two giant cell tumor of bone (GCTB) samples underwent scRNA-seq. The three TGCTs were additionally analyzed using long-read RNA sequencing. Immunofluorescence and IHC for a range of markers were used to validate and extend the scRNA-seq findings. RESULTS Two recurrent neoplastic cell populations were identified in TGCT that are highly similar to nonneoplastic synoviocytes. We identified GFPT2 as a marker that highlights the neoplastic cells in TCGT. We show that the neoplastic cells themselves do not express CSF1R. We identified overlapping MAB features between the giant cells in TGCT and GCTB. CONCLUSIONS The neoplastic cells in TGCT are highly similar to nonneoplastic synoviocytes. The lack of CSF1R on the neoplastic cells indicates they may be unaffected by current therapies. High expression of GFPT2 in the neoplastic cells is associated with activation of the YAP1/TAZ pathway. In addition, we identified expression of the platelet-derived growth factor receptor in the neoplastic cells. These findings suggest two additional pathways to target in this tumor.
Collapse
Affiliation(s)
| | - Magdalena Matusiak
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Gregory W. Charville
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Geert Spierenburg
- Department of Orthopedic Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Deana R.C. Colburg
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | | | | | - David G. Mohler
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Kristen N. Ganjoo
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Nam Q. Bui
- Division of Oncology, Department of Medicine, Stanford University, Stanford, California
| | - Raffi S. Avedian
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Judith V.M.G. Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Robert Steffner
- Department of Orthopedic Surgery, Stanford University, Stanford, California
| | - Robert B. West
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
10
|
Peterfy C, Chen Y, Countryman P, Chmielowski B, Anthony SP, Healey JH, Wainberg ZA, Cohn AL, Shapiro GI, Keedy VL, Singh A, Puzanov I, Wagner AJ, Qian M, Sterba M, Hsu HH, Tong-Starksen S, Tap WD. CSF1 receptor inhibition of tenosynovial giant cell tumor using novel disease-specific MRI measures of tumor burden. Future Oncol 2022; 18:1449-1459. [PMID: 35040698 PMCID: PMC11197039 DOI: 10.2217/fon-2021-1437] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 01/05/2022] [Indexed: 11/21/2022] Open
Abstract
Aim: Monitoring treatment of tenosynovial giant cell tumor (TGCT) is complicated by the irregular shape and asymmetrical growth of the tumor. We compared responses to pexidartinib by Response Evaluation Criteria in Solid Tumors (RECIST) 1.1 with those by tumor volume score (TVS) and modified RECIST (m-RECIST). Materials & methods: MRIs acquired every two cycles were assessed centrally using RECIST 1.1, m-RECIST and TVS and tissue damage score (TDS). Results: Thirty-one evaluable TGCT patients were treated with pexidartinib. From baseline to last visit, 94% of patients (29/31) showed a decrease in tumor size (median change: -60% [RECIST], -66% [m-RECIST], -79% [TVS]). All methods showed 100% disease control rate. For TDS, improvements were seen in bone erosion (32%), bone marrow edema (58%) and knee effusion (46%). Conclusion: TVS and m-RECIST offer potentially superior alternatives to conventional RECIST for monitoring disease progression and treatment response in TGCT. TDS adds important information about joint damage associated with TGCT.
Collapse
Affiliation(s)
| | - Yan Chen
- Spire Sciences, Inc., Boca Raton,
FL, USA
| | | | - Bartosz Chmielowski
- University of California Los Angeles, Jonsson Comprehensive Cancer Center,
Los Angeles, CA90095, USA
| | | | - John H Healey
- Memorial Sloan Kettering Cancer Center & Weill Cornell Medical College,
New York, NY10065, USA
| | | | - Allen L Cohn
- Rocky Mountain Cancer Centers,
Denver, CO80216, USA
| | - Geoffrey I Shapiro
- Dana–Farber Cancer Institute & Harvard Medical School,
Boston, MA02215, USA
| | - Vicki L Keedy
- Vanderbilt University Medical Center,
Nashville, TN37235, USA
| | - Arun Singh
- UCLA Medical Center,
Santa Monica, CA90404, USA
| | - Igor Puzanov
- Roswell Park Comprehensive Cancer Center,
Buffalo, NY14203, USA
| | - Andrew J Wagner
- Dana–Farber Cancer Institute & Harvard Medical School,
Boston, MA02215, USA
| | - Meng Qian
- Daiichi Sankyo, Inc.,
Basking Ridge, NJ07920, USA
| | - Mike Sterba
- Plexxikon Inc.,
South San Francisco,
CA94080, USA
| | - Henry H Hsu
- Plexxikon Inc.,
South San Francisco,
CA94080, USA
| | | | - William D Tap
- Memorial Sloan Kettering Cancer Center & Weill Cornell Medical College,
New York, NY10065, USA
| |
Collapse
|
11
|
Tap WD, Healey JH. Role of colony-stimulating factor 1 in the neoplastic process of tenosynovial giant cell tumor. Tumour Biol 2022; 44:239-248. [PMID: 36502356 PMCID: PMC11167812 DOI: 10.3233/tub-220005] [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] [Indexed: 12/13/2022] Open
Abstract
Tenosynovial giant cell tumors (TGCTs) are rare, locally aggressive, mesenchymal neoplasms, most often arising from the synovium of joints, bursae, or tendon sheaths. Surgical resection is the first-line treatment, but recurrence is common, with resulting impairments in patients' mobility and quality of life. Developing and optimizing the role of systemic pharmacologic therapies in TGCT management requires an understanding of the underlying disease mechanisms. The colony-stimulating factor 1 receptor (CSF1R) has emerged as having an important role in the neoplastic processes underlying TGCT. Lesions appear to contain CSF1-expressing neoplastic cells derived from the synovial lining surrounded by non-neoplastic macrophages that express the CSF1R, with lesion growth stimulated by both autocrine effects causing proliferation of the neoplastic cells themselves and by paracrine effects resulting in recruitment of CSF1 R-bearing macrophages. Other signaling pathways with evidence for involvement in TGCT pathogenesis include programmed death ligand-1, matrix metalloproteinases, and the Casitas B-cell lymphoma family of ubiquitin ligases. While growing understanding of the pathways leading to TGCT has resulted in the development of both regulatory approved and investigational therapies, more detail on underlying disease mechanisms still needs to be elucidated in order to improve the choice of individualized therapies and to enhance treatment outcomes.
Collapse
Affiliation(s)
- William D. Tap
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| | - John H. Healey
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|