1
|
Bauer S, Larkin J, Hodi FS, Stephen F, Kapiteijn EHW, Schwartz GK, Calvo E, Yerramilli-Rao P, Piperno-Neumann S, Carvajal RD. A phase Ib trial of combined PKC and MEK inhibition with sotrastaurin and binimetinib in patients with metastatic uveal melanoma. Front Oncol 2023; 12:975642. [PMID: 37359242 PMCID: PMC10288853 DOI: 10.3389/fonc.2022.975642] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 11/28/2022] [Indexed: 06/28/2023] Open
Abstract
Background Uveal melanoma is a disease characterized by constitutive activation of the G alpha pathway and downstream signaling of protein kinase C (PKC) and the mitogen-activated protein kinase (MAPK) pathway. While limited clinical activity has been observed in patients with metastatic disease with inhibition of PKC or MEK alone, preclinical data has demonstrated synergistic antitumor effects with concurrent inhibition of PKC and MEK. Method We conducted a phase Ib study of the PKC inhibitor sotrastaurin in combination with the MEK inhibitor binimetinib in patients with metastatic uveal melanoma using a Bayesian logistic regression model guided by the escalation with overdose control principle (NCT01801358). Serial blood samples and paired tumor samples were collected for pharmacokinetic (PK) and pharmacodynamic analysis. Results Thirty-eight patients were treated across six dose levels. Eleven patients experienced DLTs across the five highest dose levels tested, most commonly including vomiting (n=3), diarrhea (n=3), nausea (n=2), fatigue (n=2) and rash (n=2). Common treatment related adverse events included diarrhea (94.7%), nausea (78.9%), vomiting (71.1%), fatigue (52.6%), rash (39.5%), and elevated blood creating phosphokinase (36.8%). Two dose combinations satisfying criteria for the maximum tolerated dose (MTD) were identified: (1) sotrastaurin 300 mg and binimetinib 30 mg; and, (2) sotrastaurin 200 mg and binimetinib 45 mg. Exposure to both drugs in combination was consistent with single-agent data for either drug, indicating no PK interaction between sotrastaurin and binimetinib. Stable disease was observed in 60.5% of patients treated. No patient achieved a radiographic response per RECIST v1.1. Conclusions Concurrent administration of sotrastaurin and binimetinib is feasible but associated with substantial gastrointestinal toxicity. Given the limited clinical activity achieved with this regimen, accrual to the phase II portion of the trial was not initiated.
Collapse
Affiliation(s)
- Sebastian Bauer
- Department of Medical Oncology, Sarcoma Center, West German Cancer Center, University Duisburg-Essen, Medical School, Essen, Germany
| | - James Larkin
- Department of Medical Oncology and Hematology, The Royal Marsden Hospital, London, United Kingdom
| | - F. Stephen Hodi
- Melanoma Center and Center for Immuno-Oncology, Dana−Farber Cancer Institute, Boston, MD, United States
| | - Frank Stephen
- Hebrew University Hadassah Medical School, The Sharett Institute of Oncology, Jerusalem, Israel
- Jacob Schachter, Sheba Medical Center at Tel Hashomer, Tel-Aviv University Medical School, Tel Aviv, Israel
| | - Ellen H. W. Kapiteijn
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, Netherlands
| | - Gary K. Schwartz
- Division of Hematology and Oncology, Columbia University Irving Medical Center, New York, NY, United States
| | - Emilano Calvo
- Early Phase Clinical Drug Development in Oncology, START Madrid-CIOCC, Centro Integral Oncológico Clara Campa, Madrid, Spain
| | - Padmaja Yerramilli-Rao
- Translational Clinical Oncology, Novartis Institutes for BioMedical Research, Cambridge, MA, United States
| | | | - Richard D. Carvajal
- Department of Medical Oncology, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
2
|
Mohamed SMA, Wohlmann A, Schofield P, Sia KCS, McCalmont H, Savvides SN, Verstraete K, Kavallaris M, Christ D, Friedrich KH, Bayat N, Lock RB. A recombinant antibody fragment directed to the thymic stromal lymphopoietin receptor (CRLF2) efficiently targets pediatric Philadelphia chromosome-like acute lymphoblastic leukemia. Int J Biol Macromol 2021; 190:214-223. [PMID: 34481852 DOI: 10.1016/j.ijbiomac.2021.08.194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/21/2021] [Accepted: 08/26/2021] [Indexed: 10/20/2022]
Abstract
Antibody fragments are promising building blocks for developing targeted therapeutics, thus improving treatment efficacy while minimising off-target toxicity. Despite recent advances in targeted therapeutics, patients with Philadelphia-like acute lymphoblastic leukemia (Ph-like ALL), a high-risk malignancy, lack specific and effective targeted treatments. Cytokine receptor-like factor 2 (CRLF2) is overexpressed in 50% of Ph-like ALL cases, conferring the survival of leukemia blasts through activation of the JAK/STAT signalling pathway. Targeting such a vital cell-surface protein could result in potent anti-leukaemic efficacy and reduce the likelihood of relapse associated with antigen loss. Herein, we developed a novel single-chain variable fragment (scFv) against CRLF2 based on a monoclonal antibody raised against the recombinant extracellular domain of human TSLPRα chain. The scFv fragment demonstrated excellent binding affinity with CRLF2 protein in the nanomolar range. Cellular association studies in vitro using an inducible CRLF2 knockdown cell line and ex vivo using patient-derived xenografts revealed the selective association of the scFv with CRLF2. The fragment exhibited significant receptor antagonistic effects on STAT5 signalling, suggesting possible therapeutic implications in vivo. This study is the first to describe the potential use of a novel scFv for targeting Ph-like ALL.
Collapse
Affiliation(s)
- Sara M A Mohamed
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Andreas Wohlmann
- Institute of Biochemistry II, Jena University Hospital, Jena, Germany
| | - Peter Schofield
- Garvan Institute of Medical Research, Sydney, NSW, Australia; St.Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | - Keith C S Sia
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Hannah McCalmont
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | | | | | - Maria Kavallaris
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia; Australian Centre for Nanomedicine, ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, UNSW Sydney, Sydney, NSW, Australia
| | - Daniel Christ
- Garvan Institute of Medical Research, Sydney, NSW, Australia; St.Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, NSW, Australia
| | | | - Narges Bayat
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia
| | - Richard B Lock
- Children's Cancer Institute, Lowy Cancer Centre, UNSW Sydney, Kensington, NSW, Australia; School of Women's and Children's Health, UNSW Sydney, Kensington, NSW, Australia.; University of New South Wales Centre for Childhood Cancer Research, UNSW Sydney, Kensington, NSW, Australia.
| |
Collapse
|
3
|
FAN M, DONG S, ZOU X, ZHENG B, HUANG Y, WANG J, ZENG L. [Correlation of phosphorylated S6 protein expression in blood and brain tissue in mice and rats with kainic acid-induced seizure]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2019; 48:303-309. [PMID: 31496163 PMCID: PMC8800752 DOI: 10.3785/j.issn.1008-9292.2019.06.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/15/2019] [Indexed: 06/10/2023]
Abstract
OBJECTIVE To determine the correlation of phosphorylated ribosomal S6 protein (P-S6) content in blood and brain tissue in mice and rats with seizure. METHODS Seizure models were induced by intraperitoric injection of kainic acid (KA) in C57BL/mice and SD rats. Flow cytometry was used to detect the content of P-S6 in blood; Western blot was used to detect the expression of P-S6 in brain tissues. The correlation between P-S6 expression in blood and in brain tissue was examine by Pearson analysis, and the correlation between P-S6 expression in blood and the severity of seizure was also observed. RESULTS Western blotting analysis showed that the expression of P-S6 was significantly increased in peripheral blood and brain tissue in mice 1 h after KA-induced seizure,and the expression levels increased to (1.49±0.45) times (P<0.05) and (2.55±0.66) times (P <0.01) of the control group, respectively. Flow cytometry showed that the positive percentage and average fluorescence intensity of P-S6 in the blood of mice increased significantly 1 h after KA-induced seizures (P<0.01), which was consistent with the expression of P-S6 in brain tissue (r=0.8474, P<0.01). Flow cytometry showed that the average fluorescence intensity of P-S6 in blood increased from 14.89±9.75 to 52.35±21.72 (P<0.01) in rats with seizure, which was consistent with the change of P-S6 in brain tissue (r=0.9385, P<0.01). Rats with higher levels of seizure were of higher levels of P-S6 in peripheral blood. CONCLUSIONS Consistent correlation of P-S6 expression is demonstrated in peripheral blood and in brain tissue after KA-induced seizure, suggesting that the expression of P-S6 in blood can accurately reflect the changes of mTOR signaling pathway in brain tissue.
Collapse
Affiliation(s)
| | | | | | | | | | - Jianda WANG
- 王健达(1988-), 男, 硕士, 住院医师, 主要从事神经内科学研究; E-mail:
;
https://orcid.org/0000-0001-7148-3183
| | - Linghui ZENG
- 曾玲晖(1972-), 女, 博士, 教授, 博士生导师, 主要从事神经药理学研究; E-mail:
;
https://orcid.org/0000-0002-5924-4419
| |
Collapse
|
4
|
Zhang L, Beasley S, Prigozhina NL, Higgins R, Ikeda S, Lee FY, Marrinucci D, Jia S. Detection and Characterization of Circulating Tumour Cells in Multiple Myeloma. J Circ Biomark 2016; 5:10. [PMID: 28936258 PMCID: PMC5548310 DOI: 10.5772/64124] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 05/06/2016] [Indexed: 12/15/2022] Open
Abstract
Multiple myeloma (MM) remains an incurable disease despite recent therapeutic improvements. The ability to detect and characterize MM circulating tumour cells (CTCs) in peripheral blood provides an alternative to replace or augment invasive bone marrow (BM) biopsies with a simple blood draw, providing real-time, clinically relevant information leading to improved disease management and therapy selection. Here we have developed and qualified an enrichment-free, cell-based immunofluorescence MM CTC assay that utilizes an automated digital pathology algorithm to distinguish MM CTCs from white blood cells (WBCs) on the basis of CD138 and CD45 expression levels, as well as a number of morphological parameters. These MM CTCs were further characterized for expression of phospho-ribosomal protein S6 (pS6) as a readout for PI3K/AKT pathway activation. Clinical feasibility of the assay was established by testing blood samples from a small cohort of patients, where we detected populations of both CD138pos and CD138neg MM CTCs. In this study, we developed an immunofluorescent cell-based assay to detect and characterize CTCs in MM.
Collapse
Affiliation(s)
- Liangxuan Zhang
- Departments of Oncology Biomarker Development, Genentech Inc, South San Francisco, CA, USA
| | | | | | | | - Shoji Ikeda
- Departments of Oncology Biomarker Development, Genentech Inc, South San Francisco, CA, USA
| | | | | | - Shidong Jia
- Departments of Oncology Biomarker Development, Genentech Inc, South San Francisco, CA, USA
- Predicine Inc, Hayward, CA, USA
| |
Collapse
|
5
|
Hojjat-Farsangi M. Targeting non-receptor tyrosine kinases using small molecule inhibitors: an overview of recent advances. J Drug Target 2015. [DOI: 10.3109/1061186x.2015.1068319] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Mohammad Hojjat-Farsangi
- Department of Oncology-Pathology, Immune and Gene Therapy Lab, Cancer Center Karolinska (CCK), Karolinska University Hospital Solna and Karolinska Institute, Stockholm, Sweden and
- Department of Immunology, School of Medicine, Bushehr University of Medical Sciences, Bushehr, Iran
| |
Collapse
|
6
|
Shapiro GI, Rodon J, Bedell C, Kwak EL, Baselga J, Braña I, Pandya SS, Scheffold C, Laird AD, Nguyen LT, Xu Y, Egile C, Edelman G. Phase I safety, pharmacokinetic, and pharmacodynamic study of SAR245408 (XL147), an oral pan-class I PI3K inhibitor, in patients with advanced solid tumors. Clin Cancer Res 2013; 20:233-45. [PMID: 24166903 DOI: 10.1158/1078-0432.ccr-13-1777] [Citation(s) in RCA: 126] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE SAR245408 is a pan-class I phosphoinositide 3-kinase (PI3K) inhibitor. This phase I study determined the maximum tolerated dose (MTD) of two dosing schedules [first 21 days of a 28-day period (21/7) and continuous once-daily dosing (CDD)], pharmacokinetic and pharmacodynamic profiles, and preliminary efficacy. EXPERIMENTAL DESIGN Patients with refractory advanced solid malignancies were treated with SAR245408 using a 3 + 3 design. Pharmacokinetic parameters were determined after single and repeated doses. Pharmacodynamic effects were evaluated in plasma, hair sheath cells, and skin and tumor biopsies. RESULTS Sixty-nine patients were enrolled. The MTD of both schedules was 600 mg; dose-limiting toxicities were maculopapular rash and hypersensitivity reaction. The most frequent drug-related adverse events included dermatologic toxicities, diarrhea, nausea, and decreased appetite. Plasma pharmacokinetics showed a median time to maximum concentration of 8 to 22 hours, mean terminal elimination half-life of 70 to 88 hours, and 5- to 13-fold accumulation after daily dosing (first cycle). Steady-state concentration was reached between days 15 and 21, and exposure was dose-proportional with doses up to 400 mg. SAR245408 inhibited the PI3K pathway (∼40%-80% reduction in phosphorylation of AKT, PRAS40, 4EBP1, and S6 in tumor and surrogate tissues) and, unexpectedly, also inhibited the MEK/ERK pathway. A partial response was seen in one patient with advanced non-small cell lung cancer. Eight patients were progression-free at 6 months. Pharmacodynamic and clinical activity were observed irrespective of tumor PI3K pathway molecular alterations. CONCLUSIONS SAR245408 was tolerable at doses associated with PI3K pathway inhibition. The recommended phase II dose of the capsule formulation is 600 mg administered orally with CDD.
Collapse
Affiliation(s)
- Geoffrey I Shapiro
- Authors' Affiliations: Dana-Farber Cancer Institute; Massachusetts General Hospital; Beth Israel Deaconess Medical Center, Boston, Massachusetts; Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain; Mary Crowley Cancer Research Centers, Dallas, Texas; Exelixis Inc., South San Francisco, California; and Sanofi, Cambridge, Massachusetts, and Vitry-sur-Seine, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Lu Y, Wollak KN, Cross VA, Westrick E, Wheeler LW, Stinnette TW, Vaughn JF, Hahn SJ, Xu LC, Vlahov IR, Leamon CP. Folate receptor-targeted aminopterin therapy is highly effective and specific in experimental models of autoimmune uveitis and autoimmune encephalomyelitis. Clin Immunol 2013; 150:64-77. [PMID: 24333534 DOI: 10.1016/j.clim.2013.10.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/01/2013] [Accepted: 10/16/2013] [Indexed: 01/01/2023]
Abstract
EC0746 is a rationally designed anti-inflammatory drug conjugate consisting of a modified folic acid-based ligand linked to a γ-hydrazide analog of aminopterin. In this report, EC0746's effectiveness was evaluated against experimental retinal S-antigen (PDSAg) induced autoimmune uveitis (EAU) and myelin-basic-protein induced autoimmune encephalomyelitis (EAE). In both models, functional FR-β was detected on activated macrophages in local (retinal or central-nervous-system, respectively) and systemic (peritoneal cavity) sites of inflammation. In myelin-rich regions of EAE rats, an increased uptake of (99m)Tc-EC20 (etarfolatide; a FR-specific radioimaging agent) was also observed. EC0746 treatment at disease onset suppressed the clinical severity of both EAU and EAE, and it strongly attenuated progressive histopathological changes in the affected organs. In all parameters assessed, EC0746 activity was completely blocked by a benign folate competitor, suggesting that these therapeutic outcomes were specifically FR-β mediated. EC0746 may emerge as a useful macrophage-modulating agent for treating inflammatory episodes of organ-specific autoimmunity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Le-Cun Xu
- Endocyte, Inc., West Lafayette, IN, USA
| | | | | |
Collapse
|