1
|
Seeling C, Dahlum S, Marienfeld R, Jan V, Rack B, Gerstenmaier U, Beer AJ, Mayer-Steinacker R, Thaiss W, Barth TFE, Seufferlein T, Gaisa NT, Stilgenbauer S, Janni W, Siebert R, Döhner H, Gaidzik VI. Exploiting somatic oncogenic driver alterations in a patient with Li-Fraumeni syndrome- paving the path towards precision medicine: a case report. J Cancer Res Clin Oncol 2025; 151:37. [PMID: 39820556 PMCID: PMC11739273 DOI: 10.1007/s00432-024-06077-7] [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: 08/30/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND Li-Fraumeni syndrome (LFS) is an autosomal dominant tumor predisposition syndrome characterized by a high familial incidence of various malignancies. It results from pathogenic/likely pathogenic heterozygous constitutional variants of the TP53 gene. Due to impaired DNA damage repair, conventional cytotoxic therapies or radiotherapy should be avoided whenever feasible to mitigate the high incidence of treatment-related secondary malignancies in these patients. However, there is limited evidence supporting the effectiveness of targeted therapy approaches in LFS patients. CASE PRESENTATION We present the case of a woman with breast cancer and subsequent osteosarcoma, both treated with surgery and chemotherapy. Constitutional genetic germline testing identified a pathogenic TP53 variant in line with the clinical features of Li-Fraumeni syndrome. Subsequent molecular analysis of the osteosarcoma tissue revealed homozygous loss of the CDKN2A gene locus, warranting treatment with CDK4/6 inhibitor palbociclib. Palbociclib therapy was discontinued after one year with no evidence of disease. One year later, ovarian cancer was diagnosed, with molecular analysis indicating interstitial heterozygous loss of the BRCA2 gene locus, providing a rationale for targeted therapy with the PARP inhibitor olaparib. CONCLUSIONS In the era of accessible and comprehensive genetic and phenotypic tumor profiling, this case study of a patient with Li-Fraumeni syndrome underscores the success of precision oncology in harnessing additional somatic oncogenic driver alterations. Furthermore, it emphasizes the indispensable role of an interdisciplinary molecular tumor board, enhancing the awareness of molecular profiling and targeted therapies in patients with rare cancer susceptibility disorders.
Collapse
Affiliation(s)
- Carolin Seeling
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Sonja Dahlum
- Institute of Human Genetics, University Hospital Ulm and University of Ulm, Ulm, Germany
| | - Ralf Marienfeld
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Vera Jan
- Institute of Human Genetics, University Hospital Ulm and University of Ulm, Ulm, Germany
| | - Brigitte Rack
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | | | - Ambros J Beer
- Department of Nuclear Medicine, University Hospital Ulm, Ulm, Germany
| | | | - Wolfgang Thaiss
- Department of Nuclear Medicine, University Hospital Ulm, Ulm, Germany
| | | | - Thomas Seufferlein
- Department of Internal Medicine I, University Hospital Ulm, Ulm, Germany
| | - Nadine T Gaisa
- Institute of Pathology, University Hospital Ulm, Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
- Comprehensive Cancer Center Ulm (CCCU), University Hospital Ulm, Ulm, Germany
| | - Wolfgang Janni
- Department of Gynecology and Obstetrics, University Hospital Ulm, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University Hospital Ulm and University of Ulm, Ulm, Germany
| | - Hartmut Döhner
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany
| | - Verena I Gaidzik
- Department of Internal Medicine III, University Hospital Ulm, Ulm, Germany.
| |
Collapse
|
2
|
Liu CM, Lin FJ, Chhay C, Chen YC, Lin YK, Lu YY, Chan CS, Higa S, Chen SA, Chen YJ. Ibrutinib, a Bruton's tyrosine kinase inhibitor, regulates ventricular electromechanical activities and enhances arrhythmogenesis. Eur J Pharmacol 2024; 977:176675. [PMID: 38825303 DOI: 10.1016/j.ejphar.2024.176675] [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: 11/20/2023] [Revised: 05/12/2024] [Accepted: 05/27/2024] [Indexed: 06/04/2024]
Abstract
BACKGROUND Ibrutinib, a Bruton's tyrosine kinase inhibitor used in cancer therapy, exerts ventricular proarrhythmic effects; however, the underlying mechanisms remain unclear. Excitation-contraction coupling (E-C) disorders are pivotal for the genesis of ventricular arrhythmias (VAs), which arise mainly from the right ventricular outflow tract (RVOT). In this study, we aimed to comprehensively investigate whether ibrutinib regulates the electromechanical activities of the RVOT, leading to enhanced arrhythmogenesis, and explore the underlying mechanisms. METHODS We utilized conventional microelectrodes to synchronously record electrical and mechanical responses in rabbit RVOT tissue preparations before and after treatment with ibrutinib (10, 50, and 100 nM) and investigated their electromechanical interactions and arrhythmogenesis during programmed electrical stimulation. The fluorometric ratio technique was used to measure intracellular calcium concentration in isolated RVOT myocytes. RESULTS Ibrutinib (10-100 nM) shortened the action potential duration. Ibrutinib at 100 nM significantly increased pacing-induced ventricular tachycardia (VT) (from 0% to 62.5%, n = 8, p = 0.025). Comparisons between pacing-induced VT and non-VT episodes demonstrated that VT episodes had a greater increase in contractility than that of non-VT episodes (402.1 ± 41.4% vs. 232.4 ± 29.2%, p = 0.003). The pretreatment of ranolazine (10 μM, a late sodium current blocker) prevented the occurrence of ibrutinib-induced VAs. Ibrutinib (100 nM) increased late sodium current, reduced intracellular calcium transients, and enhanced calcium leakage in RVOT myocytes. CONCLUSION Ibrutinib increased the risk of VAs in the RVOT due to dysregulated electromechanical responses, which can be attenuated by ranolazine or apamin.
Collapse
Affiliation(s)
- Chih-Min Liu
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Fong-Jhih Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Chheng Chhay
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Cardiovascular Department, Faculty of Medicine, University of Health Sciences, Phnom Penh, Cambodia
| | - Yao-Chang Chen
- Department of Biomedical Engineering, National Defense Medical Center, Taipei, Taiwan
| | - Yung-Kuo Lin
- Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yen-Yu Lu
- Division of Cardiology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
| | - Chao-Shun Chan
- Division of Cardiology, Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Satoshi Higa
- Cardiac Electrophysiology and Pacing Laboratory, Division of Cardiovascular Medicine, Makiminato Central Hospital, Okinawa, Japan
| | - Shih-Ann Chen
- Heart Rhythm Center, Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan; Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan; Cardiovascular Center, Taichung Veterans General Hospital, Taichung, Taiwan; National Chung Hsing University, Taichung, Taiwan
| | - Yi-Jen Chen
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Cardiovascular Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Cardiovascular Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
3
|
Nikkarinen A, Glimelius I. Aplastic anemia triggered by the Bruton tyrosine kinase inhibitor acalabrutinib in two patients with mantle cell lymphoma - A case report. EJHAEM 2024; 5:820-824. [PMID: 39157591 PMCID: PMC11327736 DOI: 10.1002/jha2.929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 04/27/2024] [Accepted: 05/01/2024] [Indexed: 08/20/2024]
Abstract
The use of Bruton's tyrosine kinase inhibitors (BTKi) is rapidly increasing for patients with mantle cell lymphoma (MCL). Side effects reported so far are usually manageable. However, here we present two cases of life-threatening aplastic anemia (AA) upon treatment with the BTKi acalabrutinib for MCL. The first patient died of neutropenic infection secondary to AA. The second patient was successfully treated with immunosuppressive treatment but the MCL relapsed shortly thereafter. AA is a potentially fatal complication that should be considered when patients present with pancytopenia during treatment with BTKi.
Collapse
Affiliation(s)
- Anna Nikkarinen
- Department of ImmunologyGenetics and Pathology, Cancer Precision MedicineUppsala UniversityUppsalaSweden
| | - Ingrid Glimelius
- Department of ImmunologyGenetics and Pathology, Cancer Precision MedicineUppsala UniversityUppsalaSweden
| |
Collapse
|
4
|
Nath K, Gupta PK, Wasik MA. Emerging Role of Functional Magnetic Resonance Spectroscopy (MRS) to Monitor Response to Kinase Inhibitors in Cancer. ADVANCES IN CANCER RESEARCH & CLINICAL IMAGING : ACRCI 2024; 4:587. [PMID: 39161974 PMCID: PMC11332593 DOI: 10.33552/acrci.2024.04.000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Inhibitors of kinases involved in signaling and other intracellular pathways, have revolutionized cancer treatment by providing highly targeted and effective therapies. However, timely monitoring treatment response remains a considerable challenge since conventional methods such as assessing changes in tumor volume do not adequately capture early responses or resistance development, due to the predominantly cytostatic rather than cytotoxic effect of kinase inhibitors. Magnetic resonance spectroscopy (MRS) is a non-invasive imaging technique that can provide insights into cellular metabolism by detecting changes in metabolite concentrations. By measuring metabolite levels, MRS offers a means to assess treatment response in real-time, providing earlier indications of efficacy or resistance compared to conventional imaging modalities. Bruton's Tyrosine Kinase (BTK) is a critical enzyme involved in B-cell receptor signaling. BTK inhibitors have been approved for the treatment of Mantle Cell Lymphoma (MCL) and other B-cell malignancies. Recent studies involving genome-scale gene expression, metabolomic, and fluxomic analyses have demonstrated that ibrutinib, an index BTK inhibitor, profoundly affects the key metabolic pathways in MCL cells., including glycolysis, glutaminolysis, pentose shunt, TCA cycle and phospholipid metabolism. Importantly, the effects of ibrutinib on MCL cells directly and proportionately correlates with their sensitivity to the drug. Consequently, changes in specific metabolite concentrations detectable non-invasively by MRS such as lactate and alanine reflecting mostly the status of cellular glycolysis and glutaminolysis, respectively, have emerged as potential biomarkers for predicting response and resistance of MCL cells to BTK inhibition, both in vitro and in vivo. Preparations to validate the utility of these biomarkers in clinical setting are under way. These studies may pave the way to monitor therapeutic response to kinase inhibitors also in other types of cancer.
Collapse
Affiliation(s)
- Kavindra Nath
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Pradeep K. Gupta
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Mariusz A Wasik
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| |
Collapse
|
5
|
Nath K, Gupta PK, Basappa J, Wang S, Sen N, Lobello C, Tomar JS, Shestov AA, Orlovskiy S, Arias-Mendoza F, Rauert-Wunderlich H, Nelson DS, Glickson JD, Wasik MA. Impact of therapeutic inhibition of oncogenic cell signaling tyrosine kinase on cell metabolism: in vivo-detectable metabolic biomarkers of inhibition. J Transl Med 2024; 22:622. [PMID: 38965536 PMCID: PMC11225145 DOI: 10.1186/s12967-024-05371-9] [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: 02/20/2024] [Accepted: 06/02/2024] [Indexed: 07/06/2024] Open
Abstract
BACKGROUND Inhibition of kinases is the ever-expanding therapeutic approach to various types of cancer. Typically, assessment of the treatment response is accomplished by standard, volumetric imaging procedures, performed weeks to months after the onset of treatment, given the predominantly cytostatic nature of the kinase inhibitors, at least when used as single agents. Therefore, there is a great clinical need to develop new monitoring approaches to detect the response to kinase inhibition much more promptly. Noninvasive 1H magnetic resonance spectroscopy (MRS) can measure in vitro and in vivo concentration of key metabolites which may potentially serve as biomarkers of response to kinase inhibition. METHODS We employed mantle cell lymphoma (MCL) cell lines demonstrating markedly diverse sensitivity of inhibition of Bruton's tyrosine kinase (BTK) regarding their growth and studied in-depth effects of the inhibition on various aspects of cell metabolism including metabolite synthesis using metabolomics, glucose and oxidative metabolism by Seahorse XF technology, and concentration of index metabolites lactate, alanine, total choline and taurine by 1H MRS. RESULTS Effective BTK inhibition profoundly suppressed key cell metabolic pathways, foremost pyrimidine and purine synthesis, the citrate (TCA) cycle, glycolysis, and pyruvate and glutamine/alanine metabolism. It also inhibited glycolysis and amino acid-related oxidative metabolism. Finally, it profoundly and quickly decreased concentration of lactate (a product of mainly glycolysis) and alanine (an indicator of amino acid metabolism) and, less universally total choline both in vitro and in vivo, in the MCL xenotransplant model. The decrease correlated directly with the degree of inhibition of lymphoma cell expansion and tumor growth. CONCLUSIONS Our results indicate that BTK inhibition exerts a broad and profound suppressive effect on cell metabolism and that the affected index metabolites such as lactate, alanine may serve as early, sensitive, and reliable biomarkers of inhibition in lymphoma patients detectable by noninvasive MRS-based imaging method. This kind of imaging-based detection may also be applicable to other kinase inhibitors, as well as diverse lymphoid and non-lymphoid malignancies.
Collapse
Affiliation(s)
- Kavindra Nath
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA.
| | - Pradeep K Gupta
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Johnvesly Basappa
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111-2497, USA
| | - Shengchun Wang
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111-2497, USA
| | - Neil Sen
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111-2497, USA
| | - Cosimo Lobello
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111-2497, USA
| | - Jyoti S Tomar
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Alexander A Shestov
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Stepan Orlovskiy
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Fernando Arias-Mendoza
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
- Advanced Imaging Research, Inc., Cleveland, OH, USA
| | | | - David S Nelson
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Jerry D Glickson
- Department of Radiology, University of Pennsylvania, 423 Curie Blvd, Philadelphia, PA, 19104-6069, USA
| | - Mariusz A Wasik
- Department of Pathology, Fox Chase Cancer Center, 333 Cottman Ave, Philadelphia, PA, 19111-2497, USA.
| |
Collapse
|
6
|
Lins FV, Bispo ECI, Rodrigues NS, Silva MVS, Carvalho JL, Gelfuso GM, Saldanha-Araujo F. Ibrutinib Modulates Proliferation, Migration, Mitochondrial Homeostasis, and Apoptosis in Melanoma Cells. Biomedicines 2024; 12:1012. [PMID: 38790974 PMCID: PMC11117653 DOI: 10.3390/biomedicines12051012] [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: 04/07/2024] [Revised: 04/23/2024] [Accepted: 05/01/2024] [Indexed: 05/26/2024] Open
Abstract
Ibrutinib, a tyrosine kinase inhibitor with a broad spectrum of action, has been successfully explored to treat hematological and solid cancers. Herein, we investigated the anti-cancer effect of Ibrutinib on melanoma cell lines. Cytotoxicity was evaluated using the MTT assay. Apoptosis, mitochondrial membrane potential, reactive oxygen species (ROS) production, cell proliferation, and cell cycle stages were determined by flow cytometry. LDH release and Caspase 3/7 activity were determined by colorimetric and luminescent assays, respectively. Cell migration was evaluated by wound scratch assay. Gene expression was determined by real-time PCR. Gene Ontology (GO) enrichment analysis of melanoma clinical samples was performed using the Database for Annotation, Visualization and Integrated Discovery (DAVID). MTT assays showed that Ibrutinib is toxic for MeWo, SK-MEL-28, and WM164 cells. The annexin V/PI staining, Caspase 3/7 activity, and LDH release in MeWo cells revealed that apoptosis is the primary mechanism of death caused by Ibrutinib. Corroborating such observation, we identified that Ibrutinib treatment impairs the mitochondrial membrane potential of such cells and significantly increases the transcriptional levels of the pro-apoptotic factors ATM, HRK, BAX, BAK, CASP3, and CASP8. Furthermore, Ibrutinib showed antimetastatic potential by inhibiting the migration of MeWo cells. Finally, we performed a functional enrichment analysis and identified that the differential expression of Ibrutinib-target molecules is associated with enrichment of apoptosis and necrosis pathways in melanoma samples. Taken together, our results clearly suggest that Ibrutinib can be successfully explored as an effective therapeutic approach for melanomas.
Collapse
Affiliation(s)
- Fernanda Vitelli Lins
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
- Children’s Medical Center Research Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Elizabete Cristina Iseke Bispo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Naomí Souza Rodrigues
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| | - Maria Victória Souto Silva
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Juliana Lott Carvalho
- Laboratório Interdisciplinar de Biociências, Faculdade de Medicina, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (M.V.S.S.); (J.L.C.)
| | - Guilherme Martins Gelfuso
- Laboratório de Medicamentos, Alimentos e Cosméticos, Universidade de Brasília, Brasília 70910-900, DF, Brazil;
| | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília 70910-900, DF, Brazil; (F.V.L.); (E.C.I.B.); (N.S.R.)
| |
Collapse
|
7
|
Mar N, Zakharia Y, Falcon A, Morales-Barrera R, Mellado B, Duran I, Oh DY, Williamson SK, Gajate P, Arkenau HT, Jones RJ, Teo MY, Turan T, McLaughlin RT, Peltier HM, Chong E, Atluri H, Dean JP, Castellano D. Results from a Phase 1b/2 Study of Ibrutinib Combination Therapy in Advanced Urothelial Carcinoma. Cancers (Basel) 2023; 15:2978. [PMID: 37296940 PMCID: PMC10251876 DOI: 10.3390/cancers15112978] [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: 03/30/2023] [Revised: 05/11/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Ibrutinib is a first-in-class Bruton's tyrosine kinase inhibitor approved for the treatment of various B-cell malignancies and chronic graft-versus-host disease. We evaluated the safety and efficacy of ibrutinib, alone or combined with standard-of-care regimens, in adults with advanced urothelial carcinoma (UC). Once-daily ibrutinib was administered orally at 840 mg (single-agent or with paclitaxel) or at 560 mg (with pembrolizumab). Phase 1b determined the recommended phase 2 dose (RP2D) of ibrutinib, and phase 2 assessed progression-free survival (PFS), overall response rate (ORR), and safety. Thirty-five, eighteen, and fifty-nine patients received ibrutinib, ibrutinib plus pembrolizumab, and ibrutinib plus paclitaxel at the RP2D, respectively. Safety profiles were consistent with those of the individual agents. The best-confirmed ORRs were 7% (two partial responses) with single-agent ibrutinib and 36% (five partial responses) with ibrutinib plus pembrolizumab. Median PFS was 4.1 months (range, 1.0-37.4+) with ibrutinib plus paclitaxel. The best-confirmed ORR was 26% (two complete responses). In previously treated patients with UC, ORR was higher with ibrutinib plus pembrolizumab than with either agent alone (historical data in the intent-to-treat population). ORR with ibrutinib plus paclitaxel was greater than historical values for single-agent paclitaxel or ibrutinib. These data warrant further evaluation of ibrutinib combinations in UC.
Collapse
Affiliation(s)
- Nataliya Mar
- Division of Hematology/Oncology, University of California Irvine, Orange, CA 92868, USA
| | - Yousef Zakharia
- Division of Hematology/Oncology, University of Iowa Hospitals and Clinics, Iowa City, IA 52242, USA
| | | | - Rafael Morales-Barrera
- Vall d’Hebron Institute of Oncology, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Begona Mellado
- Medical Oncology Department, Hospital Clínic i Provincial de Barcelona, Institut D’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), University of Barcelona, 08036 Barcelona, Spain
| | - Ignacio Duran
- Hospital Universitario Marqués de Valdecilla, Instituto de Investigación Valdecilla (IDIVAL), 39011 Santander, Spain
| | - Do-Youn Oh
- Seoul National University Hospital, Cancer Research Institute, Seoul National University College of Medicine, Seoul National University Graduate School, Seoul 03080, Republic of Korea
| | | | - Pablo Gajate
- Hospital Universitario Ramón y Cajal, 28034 Madrid, Spain
| | - Hendrik-Tobias Arkenau
- Sarah Cannon Research Institute United Kingdom (SCRI-UK) and University College London Cancer Institute, London W1G 6AD, UK
| | - Robert J. Jones
- Beatson West of Scotland Cancer Centre, University of Glasgow, Glasgow G12 0YN, UK
| | - Min Yuen Teo
- Genitourinary Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tolga Turan
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA 94080, USA
| | | | - Hillary M. Peltier
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA 94080, USA
| | - Elizabeth Chong
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA 94080, USA
| | - Harisha Atluri
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA 94080, USA
| | - James P. Dean
- Pharmacyclics LLC, an AbbVie Company, South San Francisco, CA 94080, USA
| | | |
Collapse
|
8
|
Patel H, Palekar S, Patel A, Patel K. Ibrutinib amorphous solid dispersions with enhanced dissolution at colonic pH for the localized treatment of colorectal cancer. Int J Pharm 2023; 641:123056. [PMID: 37207861 DOI: 10.1016/j.ijpharm.2023.123056] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 04/23/2023] [Accepted: 05/11/2023] [Indexed: 05/21/2023]
Abstract
Colorectal cancer (CRC) is the second most leading cause of cancer-related deaths worldwide. Ibrutinib (IBR), the first in class bruton tyrosine kinase (BTK) inhibitor has promising anticancer activity. In this study, we aimed to develop a hot melt extrusion based amorphous solid dispersions (ASD) of IBR with enhanced dissolution at colonic pH and assess the anticancer activity against colon cancer cell lines. Since colonic pH is higher in CRC patients compared to healthy individuals, Eudragit® FS100 was used as pH dependent polymeric matrix for colon enabled release of IBR. Poloxamer 407, TPGS and poly(2-ethyl-2-oxazoline) were screened as plasticizer and solubilizer to improve the processability and solubility. Solid state characterization and filament appearance confirmed that IBR was molecularly dispersed within FS100+TPGS matrix. In-vitro drug release of ASD showed >96% drug release within 6 h at colonic pH with no precipitation for 12 h. Contrary, crystalline IBR showed negligible release. ASD with TPGS showed significantly higher anticancer activity in 2D and multicellular 3D spheroids of colon carcinoma cell lines (HT-29 and HT-116). The outcomes of this research suggested that ASD with a pH dependent polymer is a promising strategy to improve solubility and an effective approach in colorectal cancer targeting.
Collapse
Affiliation(s)
- Henis Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Siddhant Palekar
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Akanksha Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA
| | - Ketan Patel
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, USA.
| |
Collapse
|
9
|
Deng G, He J, Huang Q, Li T, Huang Z, Gao S, Xu J, Wang T, Di J. Ibrutinib Inhibits BTK Signaling in Tumor-Infiltrated B Cells and Amplifies Antitumor Immunity by PD-1 Checkpoint Blockade for Metastatic Prostate Cancer. Cancers (Basel) 2023; 15:cancers15082356. [PMID: 37190284 DOI: 10.3390/cancers15082356] [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: 02/17/2023] [Revised: 04/09/2023] [Accepted: 04/13/2023] [Indexed: 05/17/2023] Open
Abstract
Metastatic prostate cancer (PCa) remains incurable and causes considerably diminished overall survival. Despite significant progress in pharmacotherapy, the disease prognosis remains unchanged. Immune checkpoint inhibitors (ICIs) have demonstrated effectiveness in treating various advanced malignancies, but their efficacy in metastatic PCa is relatively limited. Previous studies have confirmed the immunosuppressive role of tumor-infiltrating B cells (TIL-Bs) in the PCa microenvironment, which accounts for their poor immunogenic potency. In this study, we demonstrated that an oral kinase agent, ibrutinib, strongly potentiated anti-PD-1 checkpoint blockade efficacy and successfully controlled tumor growth in a murine orthotopic PCa model constructed using a metastatic and hormone-independent cell line (RM-1). We identified close relationships between TIL-Bs, Bruton's tyrosine kinase (BTK), and immunosuppressive molecules by bioinformatics and histological analysis. An in vitro study showed that a low dose of ibrutinib significantly inhibited B cell proliferation and activation as well as IL-10 production through the BTK pathway. Moreover, ibrutinib-treated B cells promoted CD8+ T cell proliferation and inhibitory receptor (IR) expression. However, the same dose of ibrutinib was insufficient to induce apoptosis in cancer cells. An in vivo study showed that ibrutinib monotherapy failed to achieve tumor regression in murine models but decreased B cell infiltration and inhibited activation and IL-10 production. More importantly, CD8+ T cell infiltration increased with high IR expression. Ibrutinib synergized with anti-PD-1 checkpoint blockade enormously improved antitumor immunity, thereby reducing tumor volume in the same scenario. These data set the scene for the clinical development of ibrutinib as an immunogenic trigger to potentiate anti-PD-1 checkpoint blockade for metastatic PCa immunotherapy.
Collapse
Affiliation(s)
- Gengguo Deng
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jiannan He
- Department of Urology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China
| | - Qunxiong Huang
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tengcheng Li
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Zhansen Huang
- Department of Infertility and Sexual Medicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Shuntian Gao
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jinbin Xu
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Tiantian Wang
- Department of Medical Oncology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Jinming Di
- Department of Urology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| |
Collapse
|
10
|
Meena J, Hasija Y. Rare deleterious mutations in Bruton's tyrosine kinase as biomarkers for ibrutinib-based therapy: an in silico insight. J Mol Model 2023; 29:120. [PMID: 36991253 DOI: 10.1007/s00894-023-05515-6] [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: 11/21/2022] [Accepted: 03/14/2023] [Indexed: 03/31/2023]
Abstract
CONTEXT Squamous cell carcinoma (SCC) is the second most common type of skin cancer caused by malignant keratinocytes. Multiple studies have shown that protein mutations have a significant impact on the development and progression of cancer, including SCC. We attempted to decode the effect of single amino acid mutations in the Bruton's tyrosine kinase (BTK) protein in this study. Molecular dynamic (MD) simulations were performed on selected deleterious mutations of the BTK protein, revealing that the variants adversely affect the protein, indicating that they may contribute to the prognosis of SCC by making the protein unstable. Then, we investigated the interaction between the protein and its mutants with ibrutinib, a drug designed to treat SCC. Even though the mutations have deleterious effects on protein structure, they bind to ibrutinib similarly to their wild type counterpart. This study demonstrates that the effect of detected missense mutations is unfavorable and can result in function loss, which is severe for SCC, but that ibrutinib-based therapy can still be effective on them, and the mutations can be used as biomarkers for Ibrutinib-based treatment. METHODS Seven different computational techniques were used to compute the effect of SAVs in accordance with the experimental requirements of this study. To understand the differences in protein and mutant dynamics, MD simulation and trajectory analysis, including RMSD, RMSF, PCA, and contact analysis, were performed. The free binding energy and its decomposition for each protein-drug complex were determined using docking, MM-GBSA, MM-PBSA, and interaction analysis (wild and mutants).
Collapse
Affiliation(s)
- Jaishree Meena
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India
| | - Yasha Hasija
- Department of Biotechnology, Delhi Technological University, Delhi, 110042, India.
| |
Collapse
|
11
|
Rozkiewicz D, Hermanowicz JM, Kwiatkowska I, Krupa A, Pawlak D. Bruton's Tyrosine Kinase Inhibitors (BTKIs): Review of Preclinical Studies and Evaluation of Clinical Trials. Molecules 2023; 28:2400. [PMID: 36903645 PMCID: PMC10005125 DOI: 10.3390/molecules28052400] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
In the last few decades, there has been a growing interest in Bruton's tyrosine kinase (BTK) and the compounds that target it. BTK is a downstream mediator of the B-cell receptor (BCR) signaling pathway and affects B-cell proliferation and differentiation. Evidence demonstrating the expression of BTK on the majority of hematological cells has led to the hypothesis that BTK inhibitors (BTKIs) such as ibrutinib can be an effective treatment for leukemias and lymphomas. However, a growing body of experimental and clinical data has demonstrated the significance of BTK, not just in B-cell malignancies, but also in solid tumors, such as breast, ovarian, colorectal, and prostate cancers. In addition, enhanced BTK activity is correlated with autoimmune disease. This gave rise to the hypothesis that BTK inhibitors can be beneficial in the therapy of rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), multiple sclerosis (MS), Sjögren's syndrome (SS), allergies, and asthma. In this review article, we summarize the most recent findings regarding this kinase as well as the most advanced BTK inhibitors that have been developed to date and their clinical applications mainly in cancer and chronic inflammatory disease patients.
Collapse
Affiliation(s)
- Dariusz Rozkiewicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Justyna Magdalena Hermanowicz
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
- Department of Clinical Pharmacy, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Iwona Kwiatkowska
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| | - Anna Krupa
- Department of Internal Medicine and Metabolic, Medical University of Bialystok, M. Sklodowskiej-Curie 24a, 15-276 Bialystok, Poland
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2c, 15-222 Bialystok, Poland
| |
Collapse
|
12
|
Mete C, Pınar PT. Using a Boron‐Doped Diamond Electrode in Anionic Surfactant Media as an Improved Electrochemical Sensor for the Anticancer Drug Ibrutinib. ChemistrySelect 2023. [DOI: 10.1002/slct.202204492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Cihat Mete
- Department of Analytical Chemistry Institution of Van Yuzuncu Yil University Van Yuzuncu Yil University Faculty of Pharmacy
| | - Pınar Talay Pınar
- Department of Analytical Chemistry Institution of Van Yuzuncu Yil University Van Yuzuncu Yil University Faculty of Pharmacy
| |
Collapse
|
13
|
Cai Z, Liu R, Chan C, Lu Y, Winnik MA, Cescon DW, Reilly RM. 90Y-Labeled Gold Nanoparticle Depot (NPD) Combined with Anti-PD-L1 Antibodies Strongly Inhibits the Growth of 4T1 Tumors in Immunocompetent Mice and Induces an Abscopal Effect on a Distant Non-Irradiated Tumor. Mol Pharm 2022; 19:4199-4211. [PMID: 36287201 DOI: 10.1021/acs.molpharmaceut.2c00572] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The effectiveness and normal tissue toxicity of a novel nanoparticle depot (NPD) brachytherapy seed incorporating gold nanoparticles (AuNPs) labeled with β-particle emitting, 90Y (termed a "radiation nanomedicine"), were studied for the treatment of 4T1 triple-negative murine mammary carcinoma tumors in Balb/c mice and for inducing an abscopal effect on a distant non-irradiated tumor alone or combined with anti-PD-L1 immune checkpoint antibodies. Balb/c mice with two subcutaneous 4T1 tumors─a primary tumor and a distant secondary tumor were implanted intratumorally (i.t.) in the primary tumor with NPD incorporating 3.5 MBq of 90Y-AuNPs (1 × 1014 AuNPs) or unlabeled AuNPs, alone or combined with systemically administered anti-PD-L1 antibodies (200 μg i.p. three times/week for 2 weeks) or received anti-PD-L1 antibodies alone or no treatment. The primary tumor was strongly growth-inhibited over 14 d by NPD incorporating 90Y-AuNPs but only very modestly inhibited by NPD incorporating unlabeled AuNPs. Anti-PD-L1 antibodies alone were ineffective, and combining anti-PD-L1 antibodies with NPD incorporating 90Y-AuNPs did not further inhibit the growth of the primary tumor. Secondary tumor growth was inhibited by treatment of the primary tumor with NPD incorporating 90Y-AuNPs, and growth inhibition was enhanced by anti-PD-L1 antibodies. Treatment of the primary tumor with NPD incorporating unlabeled AuNPs or anti-PD-L1 antibodies alone had no effect on secondary tumor growth. Biodistribution studies showed high uptake of 90Y in the primary tumor [516-810% implanted dose/g (%ID/g)] but very low uptake in the secondary tumor (0.033-0.16% ID/g) and in normal tissues (<0.5% ID/g) except for kidneys (5-8% ID/g). Very high radiation absorbed doses were estimated for the primary tumor (472 Gy) but very low doses in the secondary tumor (0.13 Gy). There was highdose-heterogeneity in the primary tumor with doses as high as 9964 Gy in close proximity to the NPD, decreasing rapidly with distance from the NPD. Normal organ doses were low (<1 Gy) except for kidneys (4 Gy). No normal tissue toxicity was observed, but white blood cell counts (WBC) decreased in tumor-bearing mice treated with NPD incorporating 90Y-AuNPs. Decreased WBC counts were interpreted as tumor response and not toxicity since these were higher than that in healthy non-tumor-bearing mice, and there was a direct association between WBC counts and 4T1 tumor burden. We conclude that implantation of NPD incorporating 90Y-AuNPs into a primary 4T1 tumor in Balb/c mice strongly inhibited tumor growth and combined with anti-PD-L1 antibodies induced an abscopal effect on a distant secondary tumor. This radiation nanomedicine is promising for the local treatment of triple-negative breast cancer tumors in patients, and these therapeutic effects may extend to non-irradiated lesions, especially when combined with checkpoint immunotherapy.
Collapse
Affiliation(s)
- Zhongli Cai
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, OntarioM5S 3M2, Canada
| | - Rella Liu
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, OntarioM5S 3M2, Canada
| | - Conrad Chan
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, OntarioM5S 3M2, Canada
| | - Yijie Lu
- Department of Chemistry, University of Toronto, Toronto, OntarioM5S 3H6, Canada
| | - Mitchell A. Winnik
- Department of Chemistry, University of Toronto, Toronto, OntarioM5S 3H6, Canada
| | - David W. Cescon
- Department of Medical Oncology, Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 2C1, Canada
| | - Raymond M. Reilly
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College St., Toronto, OntarioM5S 3M2, Canada
- Department of Medical Imaging, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada
- Joint Department of Medical Imaging and Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, M5G 2C1, Canada
| |
Collapse
|
14
|
Bruton’s Tyrosine Kinase Inhibitor Zanubrutinib Effectively Modulates Cancer Resistance by Inhibiting Anthracycline Metabolism and Efflux. Pharmaceutics 2022; 14:pharmaceutics14101994. [PMID: 36297430 PMCID: PMC9611657 DOI: 10.3390/pharmaceutics14101994] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/29/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Zanubrutinib (ZAN) is a Bruton’s tyrosine kinase inhibitor recently approved for the treatment of some non-Hodgkin lymphomas. In clinical trials, ZAN is often combined with standard anthracycline (ANT) chemotherapy. Although ANTs are generally effective, drug resistance is a crucial obstacle that leads to treatment discontinuation. This study showed that ZAN counteracts ANT resistance by targeting aldo-keto reductase 1C3 (AKR1C3) and ATP-binding cassette (ABC) transporters. AKR1C3 catalyses the transformation of ANTs to less potent hydroxy-metabolites, whereas transporters decrease the ANT-effective concentrations by pumping them out of the cancer cells. In our experiments, ZAN inhibited the AKR1C3-mediated inactivation of daunorubicin (DAUN) at both the recombinant and cellular levels. In the drug combination experiments, ZAN synergistically sensitised AKR1C3-expressing HCT116 and A549 cells to DAUN treatment. Gene induction studies further confirmed that ZAN did not increase the intracellular level of AKR1C3 mRNA; thus, the drug combination effect is not abolished by enzyme induction. Finally, in accumulation assays, ZAN was found to interfere with the DAUN efflux mediated by the ABCB1, ABCG2, and ABCC1 transporters, which might further contribute to the reversal of ANT resistance. In summary, our data provide the rationale for ZAN inclusion in ANT-based therapy and suggest its potential for the treatment of tumours expressing AKR1C3 and/or the above-mentioned ABC transporters.
Collapse
|
15
|
Kozyra P, Krasowska D, Pitucha M. New Potential Agents for Malignant Melanoma Treatment-Most Recent Studies 2020-2022. Int J Mol Sci 2022; 23:6084. [PMID: 35682764 PMCID: PMC9180979 DOI: 10.3390/ijms23116084] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/25/2022] [Accepted: 05/25/2022] [Indexed: 02/05/2023] Open
Abstract
Malignant melanoma (MM) is the most lethal skin cancer. Despite a 4% reduction in mortality over the past few years, an increasing number of new diagnosed cases appear each year. Long-term therapy and the development of resistance to the drugs used drive the search for more and more new agents with anti-melanoma activity. This review focuses on the most recent synthesized anti-melanoma agents from 2020-2022. For selected agents, apart from the analysis of biological activity, the structure-activity relationship (SAR) is also discussed. To the best of our knowledge, the following literature review delivers the latest achievements in the field of new anti-melanoma agents.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| | - Danuta Krasowska
- Department of Dermatology, Venerology and Pediatric Dermatology, Medical University of Lublin, 20-081 Lublin, Poland;
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|