1
|
Amato F, Strotmann R, Castello R, Bruns R, Ghori V, Johne A, Berghoff K, Venkatakrishnan K, Terranova N. Explainable machine learning prediction of edema adverse events in patients treated with tepotinib. Clin Transl Sci 2024; 17:e70010. [PMID: 39222377 PMCID: PMC11368086 DOI: 10.1111/cts.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/05/2024] [Accepted: 08/04/2024] [Indexed: 09/04/2024] Open
Abstract
Tepotinib is approved for the treatment of patients with non-small-cell lung cancer harboring MET exon 14 skipping alterations. While edema is the most prevalent adverse event (AE) and a known class effect of MET inhibitors including tepotinib, there is still limited understanding about the factors contributing to its occurrence. Herein, we apply machine learning (ML)-based approaches to predict the likelihood of occurrence of edema in patients undergoing tepotinib treatment, and to identify factors influencing its development over time. Data from 612 patients receiving tepotinib in five Phase I/II studies were modeled with two ML algorithms, Random Forest, and Gradient Boosting Trees, to predict edema AE incidence and severity. Probability calibration was applied to give a realistic estimation of the likelihood of edema AE. Best model was tested on follow-up data and on data from clinical studies unused while training. Results showed high performances across all the tested settings, with F1 scores up to 0.961 when retraining the model with the most relevant covariates. The use of ML explainability methods identified serum albumin as the most informative longitudinal covariate, and higher age as associated with higher probabilities of more severe edema. The developed methodological framework enables the use of ML algorithms for analyzing clinical safety data and exploiting longitudinal information through various covariate engineering approaches. Probability calibration ensures the accurate estimation of the likelihood of the AE occurrence, while explainability tools can identify factors contributing to model predictions, hence supporting population and individual patient-level interpretation.
Collapse
Affiliation(s)
- Federico Amato
- Swiss Data Science Center (EPFL and ETH Zurich)LausanneSwitzerland
| | | | - Roberto Castello
- Swiss Data Science Center (EPFL and ETH Zurich)LausanneSwitzerland
| | - Rolf Bruns
- The healthcare business of Merck KGaADarmstadtGermany
| | - Vishal Ghori
- Ares Trading S.A., Eysins, Switzerland, an affiliate of Merck KGaA, DarmstadtGermany
| | - Andreas Johne
- The healthcare business of Merck KGaADarmstadtGermany
| | | | | | - Nadia Terranova
- Quantitative PharmacologyAres Trading S.A., Lausanne, Switzerland, an affiliate of Merck KGaADarmstadtGermany
| |
Collapse
|
2
|
Su P, Zhang M, Kang X. Targeting c-Met in the treatment of urologic neoplasms: Current status and challenges. Front Oncol 2023; 13:1071030. [PMID: 36959792 PMCID: PMC10028134 DOI: 10.3389/fonc.2023.1071030] [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: 10/15/2022] [Accepted: 02/23/2023] [Indexed: 03/09/2023] Open
Abstract
At present, studies have found that c-Met is mainly involved in epithelial-mesenchymal transition (EMT) of tumor tissues in urologic neoplasms. Hepatocyte growth factor (HGF) combined with c-Met promotes the mitosis of tumor cells, and then induces motility, angiogenesis, migration, invasion and drug resistance. Therefore, c-Met targeting therapy may have great potential in urologic neoplasms. Many strategies targeting c-Met have been widely used in the study of urologic neoplasms. Although the use of targeting c-Met therapy has a strong biological basis for the treatment of urologic neoplasms, the results of current clinical trials have not yielded significant results. To promote the application of c-Met targeting drugs in the clinical treatment of urologic neoplasms, it is very important to study the detailed mechanism of c-Met in urologic neoplasms and innovate c-Met targeted drugs. This paper firstly discussed the value of c-Met targeted therapy in urologic neoplasms, then summarized the related research progress, and finally explored the potential targets related to the HGF/c-Met signaling pathway. It may provide a new concept for the treatment of middle and late urologic neoplasms.
Collapse
|
3
|
Zhang Z, Li D, Yun H, Tong J, Liu W, Chai K, Zeng T, Gao Z, Xie Y. Opportunities and challenges of targeting c-Met in the treatment of digestive tumors. Front Oncol 2022; 12:923260. [PMID: 35978812 PMCID: PMC9376446 DOI: 10.3389/fonc.2022.923260] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/27/2022] [Indexed: 11/13/2022] Open
Abstract
At present, a large number of studies have demonstrated that c-Met generally exerts a crucial function of promoting tumor cells proliferation and differentiation in digestive system tumors. c-Met also mediates tumor progression and drug resistance by signaling interactions with other oncogenic molecules and then activating downstream pathways. Therefore, c-Met is a promising target for the treatment of digestive system tumors. Many anti-tumor therapies targeting c-Met (tyrosine kinase inhibitors, monoclonal antibodies, and adoptive immunotherapy) have been developed in treating digestive system tumors. Some drugs have been successfully applied to clinic, but most of them are defective due to their efficacy and complications. In order to promote the clinical application of targeting c-Met drugs in digestive system tumors, it is necessary to further explore the mechanism of c-Met action in digestive system tumors and optimize the anti-tumor treatment of targeting c-Met drugs. Through reading a large number of literatures, the author systematically reviewed the biological functions and molecular mechanisms of c-Met associated with tumor and summarized the current status of targeting c-Met in the treatment of digestive system tumors so as to provide new ideas for the treatment of digestive system tumors.
Collapse
Affiliation(s)
- Zhengchao Zhang
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
- Department of General Surgery, Second Hospital of Lanzhou University, Lanzhou, China
| | - Dong Li
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Heng Yun
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Jie Tong
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Wei Liu
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Keqiang Chai
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Tongwei Zeng
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
| | - Zhenghua Gao
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
- *Correspondence: Yongqiang Xie, ; Zhenghua Gao,
| | - Yongqiang Xie
- Department of General Surgery, The Third Affiliated Hospital of Gansu University of Traditional Chinese Medicine, Baiyin, China
- *Correspondence: Yongqiang Xie, ; Zhenghua Gao,
| |
Collapse
|
4
|
Xiong W, Hietala SF, Nyberg J, Papasouliotis O, Johne A, Berghoff K, Goteti K, Dong J, Girard P, Venkatakrishnan K, Strotmann R. Exposure-response analyses for the MET inhibitor tepotinib including patients in the pivotal VISION trial: support for dosage recommendations. Cancer Chemother Pharmacol 2022; 90:53-69. [PMID: 35771259 PMCID: PMC9300558 DOI: 10.1007/s00280-022-04441-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 05/10/2022] [Indexed: 11/24/2022]
Abstract
Purpose Tepotinib is a highly selective MET inhibitor approved for treatment of non-small cell lung cancer (NSCLC) harboring METex14 skipping alterations. Analyses presented herein evaluated the relationship between tepotinib exposure, and efficacy and safety outcomes. Methods Exposure–efficacy analyses included data from an ongoing phase 2 study (VISION) investigating 500 mg/day tepotinib in NSCLC harboring METex14 skipping alterations. Efficacy endpoints included objective response, duration of response, and progression-free survival. Exposure–safety analyses included data from VISION, plus four completed studies in advanced solid tumors/hepatocellular carcinoma (30–1400 mg). Safety endpoints included edema, serum albumin, creatinine, amylase, lipase, alanine aminotransferase, aspartate aminotransferase, and QT interval corrected using Fridericia’s method (QTcF). Results Tepotinib exhibited flat exposure–efficacy relationships for all endpoints within the exposure range observed with 500 mg/day. Tepotinib also exhibited flat exposure–safety relationships for all endpoints within the exposure range observed with 30–1400 mg doses. Edema is the most frequently reported adverse event and the most frequent cause of tepotinib dose reductions and interruptions; however, the effect plateaued at low exposures. Concentration-QTc analyses using data from 30 to 1400 mg tepotinib resulted in the upper bounds of the 90% confidence interval being less than 10 ms for the mean exposures at the therapeutic (500 mg) and supratherapeutic (1000 mg) doses. Conclusions These analyses provide important quantitative pharmacologic support for benefit/risk assessment of the 500 mg/day dosage of tepotinib as being appropriate for the treatment of NSCLC harboring METex14 skipping alterations. Registration Numbers NCT01014936, NCT01832506, NCT01988493, NCT02115373, NCT02864992. Supplementary Information The online version contains supplementary material available at 10.1007/s00280-022-04441-3.
Collapse
Affiliation(s)
- Wenyuan Xiong
- Merck Institute of Pharmacometrics, Lausanne, Switzerland
| | | | | | | | | | | | - Kosalaram Goteti
- EMD Serono Research and Development Institute Inc., Billerica, MA, USA
| | - Jennifer Dong
- EMD Serono Research and Development Institute Inc., Billerica, MA, USA
| | - Pascal Girard
- Merck Institute of Pharmacometrics, Lausanne, Switzerland
| | | | | |
Collapse
|
5
|
Fogli S, Tabbò F, Capuano A, Re MD, Passiglia F, Cucchiara F, Scavone C, Gori V, Novello S, Schmidinger M, Danesi R. The expanding family of c-Met inhibitors in solid tumors: a comparative analysis of their pharmacologic and clinical differences. Crit Rev Oncol Hematol 2022; 172:103602. [DOI: 10.1016/j.critrevonc.2022.103602] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 01/15/2022] [Accepted: 01/17/2022] [Indexed: 12/16/2022] Open
|
6
|
Rashid MH. Full-length recombinant antibodies from Escherichia coli: production, characterization, effector function (Fc) engineering, and clinical evaluation. MAbs 2022; 14:2111748. [PMID: 36018829 PMCID: PMC9423848 DOI: 10.1080/19420862.2022.2111748] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Although several antibody fragments and antibody fragment-fusion proteins produced in Escherichia coli (E. coli) are approved as therapeutics for various human diseases, a full-length monoclonal or a bispecific antibody produced in E. coli has not yet been approved. The past decade witnessed substantial progress in expression of full-length antibodies in the E. coli cytoplasm and periplasm, as well as in cell-free expression systems. The equivalency of E. coli-produced aglycosylated antibodies and their mammalian cell-produced counterparts, with respect to biochemical and biophysical properties, including antigen binding, in vitro and in vivo serum stability, pharmacokinetics, and in vivo serum half-life, has been demonstrated. Extensive engineering of the Fc domain of aglycosylated antibodies enables recruitment of various effector functions, despite the lack of N-linked glycans. This review summarizes recent research, preclinical advancements, and clinical development of E. coli-produced aglycosylated therapeutic antibodies as monoclonal, bispecific, and antibody-drug conjugates for use in autoimmune, oncology, and immuno-oncology areas.Abbreviations: ADA Anti-drug antibody; ADCC Antibody-dependent cellular cytotoxicity; ADCP Antibody-dependent cellular phagocytosis; ADC Antibody-drug conjugate; aFc Aglycosylated Fc; AMD Age-related macular degeneration aTTP Acquired thrombotic thrombocytopenic purpura; BCMA B-cell maturation antigen; BLA Biologics license application; BsAb Bispecific antibody; C1q Complement protein C1q; CDC Complement-dependent cytotoxicity; CDCC Complement-dependent cellular cytotoxicity; CDCP Complement-dependent cellular phagocytosis; CEX Cation exchange chromatography; CFPS Cell-free protein expression; CHO Chinese Hamster Ovary; CH1-3 Constant heavy chain 1-3; CL Constant light chain; DLBCL Diffuse large B-cell lymphoma; DAR Drug antibody ratio; DC Dendritic cell; dsFv Disulfide-stabilized Fv; EU European Union; EGFR Epidermal growth factor receptor; E. coli Escherichia coli; EpCAM Epithelial cell adhesion molecule; Fab Fragment antigen binding; FACS Fluorescence activated cell sorting; Fc Fragment crystallizable; FcRn Neonatal Fc receptor; FcɣRs Fc gamma receptors; FDA Food and Drug Administration; FL-IgG Full-length immunoglobulin; Fv Fragment variable; FolRαa Folate receptor alpha; gFc Glycosylated Fc; GM-CSF Granulocyte macrophage-colony stimulating factor; GPx7 Human peroxidase 7; HCL Hairy cell leukemia; HIV Human immunodeficiency virusl; HER2 Human epidermal growth factor receptor 2; HGF Hepatocyte growth factor; HIC Hydrophobic interaction chromatography; HLA Human leukocyte antigen; IBs Inclusion bodies; IgG1-4 Immunoglobulin 1-4; IP Intraperitoneal; ITC Isothermal titration calorimetry; ITP Immune thrombocytopenia; IV Intravenous; kDa Kilodalton; KiH Knob-into-Hole; mAb Monoclonal antibody; MAC Membrane-attack complex; mCRC Metastatic colorectal cancer; MM Multipl myeloma; MOA Mechanism of action; MS Mass spectrometry; MUC1 Mucin 1; MG Myasthenia gravis; NB Nanobody; NK Natural killer; nsAA Nonstandard amino acid; NSCLC Non-small cell lung cancer; P. aeruginosa Pseudomonas aeruginosa; PD-1 Programmed cell death 1; PD-L1 Programmed cell death-ligand 1; PDI Protein disulfide isomerase; PECS Periplasmic expression cytometric screening; PK Pharmacokinetics; P. pastoris Pichia pastoris; PTM Post-translational modification; Rg Radius of gyration; RA Rheumatoid arthritis; RT-PCR Reverse transcription polymerase chain reaction; SAXS Small angle X-ray scattering; scF Single chain Fv; SCLC Small cell lung cancer; SDS-PAGE Sodium dodecyl sulfate-polyacrylamide gel electrophoresis; SEC Size exclusion chromatography; SEED Strand-exchange engineered domain; sRNA Small regulatory RNA; SRP Signal recognition particle; T1/2 Half-life; Tagg Aggregation temperature; TCR T cell receptor; TDB T cell-dependent bispecific; TF Tissue factor; TIR Translation initiation region; Tm Melting temperature; TNBC Triple-negative breast cancer; TNF Tumor necrosis factor; TPO Thrombopoietin; VEGF Vascular endothelial growth factor; vH Variable heavy chain; vL Variable light chain; vWF von Willebrand factor; WT Wild type.
Collapse
|
7
|
Aftimos P, Rolfo C, Rottey S, Barthélémy P, Borg C, Park K, Oh DY, Kim SW, De Jonge N, Hanssens V, Zwanenpoel K, Molthoff C, Vugts D, Dreier T, Verheesen P, van Dongen GA, Jacobs J, Van Rompaey L, Hultberg A, Michieli P, Pauwels P, Fung S, Thibault A, de Haard H, Leupin N, Awada A. The NHance ® Mutation-Equipped Anti-MET Antibody ARGX-111 Displays Increased Tissue Penetration and Anti-Tumor Activity in Advanced Cancer Patients. Biomedicines 2021; 9:biomedicines9060665. [PMID: 34200749 PMCID: PMC8229762 DOI: 10.3390/biomedicines9060665] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/27/2021] [Accepted: 06/08/2021] [Indexed: 11/25/2022] Open
Abstract
Dysregulation of MET signaling has been implicated in tumorigenesis and metastasis. ARGX-111 combines complete blockade of this pathway with enhanced tumor cell killing and was investigated in 24 patients with MET-positive advanced cancers in a phase 1b study at four dose levels (0.3–10 mg/kg). ARGX-111 was well tolerated up to 3 mg/kg (MTD). Anti-tumor activity was observed in nearly half of the patients (46%) with a mean duration of treatment of 12 weeks. NHance® mutations in the Fc of ARGX-111 increased affinity for the neonatal Fc receptor (FcRn) at acidic pH, stimulating transcytosis across FcRn-expressing cells and radiolabeled ARGX-111 accumulated in lymphoid tissues, bone and liver, organs expressing FcRn at high levels in a biodistribution study using human FcRn transgenic mice. In line with this, we observed, in a patient with MET-amplified (>10 copies) gastric cancer, diminished metabolic activity in multiple metastatic lesions in lymphoid and bone tissues by 18F-FDG-PET/CT after two infusions with 0.3 mg/kg ARGX-111. When escalated to 1 mg/kg, a partial response was reached. Furthermore, decreased numbers of CTC (75%) possibly by the enhanced tumor cell killing witnessed the modes of action of the drug, warranting further clinical investigation of ARGX-111.
Collapse
Affiliation(s)
- Philippe Aftimos
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| | - Christian Rolfo
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | | | - Philippe Barthélémy
- Medical Oncology Unit, Hôpitaux Universitaires de Strasbourg, 67000 Strasbourg, France;
| | - Christophe Borg
- Medical Oncology Department, University Hospital of Besançon, CEDEX, 25000 Besançon, France;
| | - Keunchil Park
- Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea;
| | - Do-Youn Oh
- Seoul National University Hospital, Seoul 03080, Korea;
| | - Sang-We Kim
- Asan Medical Center, Department of Oncology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Natalie De Jonge
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Valérie Hanssens
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Karen Zwanenpoel
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Carla Molthoff
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Daniëlle Vugts
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Torsten Dreier
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
| | - Peter Verheesen
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Guus A.M.S. van Dongen
- Department of Radiology & Nuclear Medicine, VU University Medical Center Amsterdam, 1012 Amsterdam, The Netherlands; (C.M.); (D.V.); (G.A.M.S.v.D.)
| | - Julie Jacobs
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Luc Van Rompaey
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Anna Hultberg
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Paolo Michieli
- AgomAb Therapeutics NV, 9000 Ghent, Belgium;
- Department of Oncology, University of Torino Medical School, 10124 Turin, Italy
| | - Patrick Pauwels
- University Hospital Antwerp, 2650 Edegem, Belgium; (C.R.); (K.Z.); (P.P.)
| | - Samson Fung
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Alain Thibault
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Hans de Haard
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
| | - Nicolas Leupin
- Argenx BV, Industriepark Zwijnaarde 7, 9052 Ghent, Belgium; (N.D.J.); (V.H.); (T.D.); (P.V.); (J.J.); (L.V.R.); (A.H.); (S.F.); (A.T.); (H.d.H.)
- Correspondence: ; Tel.: +41-79-293-18-14
| | - Ahmad Awada
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, 1000 Brussels, Belgium; (P.A.); (A.A.)
| |
Collapse
|
8
|
Sandomenico A, Sivaccumar JP, Ruvo M. Evolution of Escherichia coli Expression System in Producing Antibody Recombinant Fragments. Int J Mol Sci 2020; 21:ijms21176324. [PMID: 32878291 PMCID: PMC7504322 DOI: 10.3390/ijms21176324] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/12/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023] Open
Abstract
Antibodies and antibody-derived molecules are continuously developed as both therapeutic agents and key reagents for advanced diagnostic investigations. Their application in these fields has indeed greatly expanded the demand of these molecules and the need for their production in high yield and purity. While full-length antibodies require mammalian expression systems due to the occurrence of functionally and structurally important glycosylations, most antibody fragments and antibody-like molecules are non-glycosylated and can be more conveniently prepared in E. coli-based expression platforms. We propose here an updated survey of the most effective and appropriate methods of preparation of antibody fragments that exploit E. coli as an expression background and review the pros and cons of the different platforms available today. Around 250 references accompany and complete the review together with some lists of the most important new antibody-like molecules that are on the market or are being developed as new biotherapeutics or diagnostic agents.
Collapse
|
9
|
Strickler JH, LoRusso P, Salgia R, Kang YK, Yen CJ, Lin CC, Ansell P, Motwani M, Wong S, Yue H, Wang L, Reilly E, Afar D, Naumovski L, Ramanathan RK. Phase I Dose-Escalation and -Expansion Study of Telisotuzumab (ABT-700), an Anti-c-Met Antibody, in Patients with Advanced Solid Tumors. Mol Cancer Ther 2020; 19:1210-1217. [PMID: 32127466 DOI: 10.1158/1535-7163.mct-19-0529] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/25/2019] [Accepted: 02/28/2020] [Indexed: 11/16/2022]
Abstract
This first-in-human phase I study evaluated the pharmacokinetics, safety, and preliminary efficacy of telisotuzumab, formerly called ABT-700, an antagonistic antibody directed against c-Met. For dose escalation (3+3 design), 3 to 6 patients with advanced solid tumors were enrolled into four dose cohorts (5-25 mg/kg). In the dose-expansion phase, a subset of patients was prospectively selected for MET amplification (FISH screening). Patients received telisotuzumab intravenously on day 1 every 21 days. For dose expansion, 15 mg/kg was chosen as the dose on the basis of safety, pharmacokinetics, and other data from the escalation cohorts. Forty-five patients were enrolled and received at least one dose of telisotuzumab (dose escalation, n = 15; dose expansion, n = 30). Telisotuzumab showed a linear pharmacokinetics profile; peak plasma concentration was proportional to dose level. There were no acute infusion reactions and no dose-limiting toxicities were observed. The most common treatment-related adverse events included hypoalbuminemia (n = 9, 20.0%) and fatigue (n = 5, 11.1%). By Response Evaluation Criteria In Solid Tumors (RECIST), 4 of 10 (40.0%) patients with MET-amplified tumors had confirmed partial response in target lesions (one ovarian, two gastric, and one esophageal), two (20.0%) had stable disease, three (30.0%) had progressive disease; one patient was unable to be evaluated. Among patients with nonamplified tumors (n = 35), no objective responses were observed; however, 11 patients had stable disease per RECIST criteria. In conclusion, telisotuzumab has an acceptable safety profile with clinical activity observed in patients with MET-amplified advanced solid tumors.
Collapse
Affiliation(s)
| | - Patricia LoRusso
- Yale University School of Medicine, Yale Cancer Center, New Haven, Connecticut
| | | | - Yoon-Koo Kang
- Asan Medical Center, University of Ulsan, Seoul, South Korea
| | - Chia Jui Yen
- National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Chia-Chi Lin
- National Taiwan University Hospital, Taipei, Taiwan
| | | | | | - Shekman Wong
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - Huibin Yue
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - Lan Wang
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | | | - Daniel Afar
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | - Louie Naumovski
- Oncology Early Development, AbbVie Inc., Redwood City, California
| | | |
Collapse
|
10
|
Decoding and targeting the molecular basis of MACC1-driven metastatic spread: Lessons from big data mining and clinical-experimental approaches. Semin Cancer Biol 2019; 60:365-379. [PMID: 31430556 DOI: 10.1016/j.semcancer.2019.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 08/09/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Metastasis remains the key issue impacting cancer patient survival and failure or success of cancer therapies. Metastatic spread is a complex process including dissemination of single cells or collective cell migration, penetration of the blood or lymphatic vessels and seeding at a distant organ site. Hundreds of genes involved in metastasis have been identified in studies across numerous cancer types. Here, we analyzed how the metastasis-associated gene MACC1 cooperates with other genes in metastatic spread and how these coactions could be exploited by combination therapies: We performed (i) a MACC1 correlation analysis across 33 cancer types in the mRNA expression data of TCGA and (ii) a comprehensive literature search on reported MACC1 combinations and regulation mechanisms. The key genes MET, HGF and MMP7 reported together with MACC1 showed significant positive correlations with MACC1 in more than half of the cancer types included in the big data analysis. However, ten other genes also reported together with MACC1 in the literature showed significant positive correlations with MACC1 in only a minority of 5 to 15 cancer types. To uncover transcriptional regulation mechanisms that are activated simultaneously with MACC1, we isolated pan-cancer consensus lists of 1306 positively and 590 negatively MACC1-correlating genes from the TCGA data and analyzed each of these lists for sharing transcription factor binding motifs in the promotor region. In these lists, binding sites for the transcription factors TELF1, ETS2, ETV4, TEAD1, FOXO4, NFE2L1, ELK1, SP1 and NFE2L2 were significantly enriched, but none of them except SP1 was reported in combination with MACC1 in the literature. Thus, while some of the results of the big data analysis were in line with the reported experimental results, hypotheses on new genes involved in MACC1-driven metastasis formation could be generated and warrant experimental validation. Furthermore, the results of the big data analysis can help to prioritize cancer types for experimental studies and testing of combination therapies.
Collapse
|
11
|
Casaletto JB, Geddie ML, Abu-Yousif AO, Masson K, Fulgham A, Boudot A, Maiwald T, Kearns JD, Kohli N, Su S, Razlog M, Raue A, Kalra A, Håkansson M, Logan DT, Welin M, Chattopadhyay S, Harms BD, Nielsen UB, Schoeberl B, Lugovskoy AA, MacBeath G. MM-131, a bispecific anti-Met/EpCAM mAb, inhibits HGF-dependent and HGF-independent Met signaling through concurrent binding to EpCAM. Proc Natl Acad Sci U S A 2019; 116:7533-7542. [PMID: 30898885 PMCID: PMC6462049 DOI: 10.1073/pnas.1819085116] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Activation of the Met receptor tyrosine kinase, either by its ligand, hepatocyte growth factor (HGF), or via ligand-independent mechanisms, such as MET amplification or receptor overexpression, has been implicated in driving tumor proliferation, metastasis, and resistance to therapy. Clinical development of Met-targeted antibodies has been challenging, however, as bivalent antibodies exhibit agonistic properties, whereas monovalent antibodies lack potency and the capacity to down-regulate Met. Through computational modeling, we found that the potency of a monovalent antibody targeting Met could be dramatically improved by introducing a second binding site that recognizes an unrelated, highly expressed antigen on the tumor cell surface. Guided by this prediction, we engineered MM-131, a bispecific antibody that is monovalent for both Met and epithelial cell adhesion molecule (EpCAM). MM-131 is a purely antagonistic antibody that blocks ligand-dependent and ligand-independent Met signaling by inhibiting HGF binding to Met and inducing receptor down-regulation. Together, these mechanisms lead to inhibition of proliferation in Met-driven cancer cells, inhibition of HGF-mediated cancer cell migration, and inhibition of tumor growth in HGF-dependent and -independent mouse xenograft models. Consistent with its design, MM-131 is more potent in EpCAM-high cells than in EpCAM-low cells, and its potency decreases when EpCAM levels are reduced by RNAi. Evaluation of Met, EpCAM, and HGF levels in human tumor samples reveals that EpCAM is expressed at high levels in a wide range of Met-positive tumor types, suggesting a broad opportunity for clinical development of MM-131.
Collapse
Affiliation(s)
| | - Melissa L Geddie
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Adnan O Abu-Yousif
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Kristina Masson
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Aaron Fulgham
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Antoine Boudot
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Tim Maiwald
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Jeffrey D Kearns
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Neeraj Kohli
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Stephen Su
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Maja Razlog
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Andreas Raue
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139;
| | - Ashish Kalra
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Maria Håkansson
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | - Derek T Logan
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | - Martin Welin
- SARomics Biostructures AB, Medicon Village, SE-223 81 Lund, Sweden
| | | | - Brian D Harms
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Ulrik B Nielsen
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Birgit Schoeberl
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Alexey A Lugovskoy
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139
| | - Gavin MacBeath
- Discovery Division, Merrimack Pharmaceuticals, Inc., Cambridge, MA 02139;
| |
Collapse
|
12
|
Cruickshanks N, Zhang Y, Hine S, Gibert M, Yuan F, Oxford M, Grello C, Pahuski M, Dube C, Guessous F, Wang B, Deveau C, Saoud K, Gallagher I, Wulfkuhle J, Schiff D, Phan S, Petricoin E, Abounader R. Discovery and Therapeutic Exploitation of Mechanisms of Resistance to MET Inhibitors in Glioblastoma. Clin Cancer Res 2018; 25:663-673. [PMID: 30201763 DOI: 10.1158/1078-0432.ccr-18-0926] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/13/2018] [Accepted: 09/05/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE Glioblastoma (GBM) is the most common and most lethal primary malignant brain tumor. The receptor tyrosine kinase MET is frequently upregulated or overactivated in GBM. Although clinically applicable MET inhibitors have been developed, resistance to single modality anti-MET drugs frequently occurs, rendering these agents ineffective. We aimed to determine the mechanisms of MET inhibitor resistance in GBM and use the acquired information to develop novel therapeutic approaches to overcome resistance.Experimental Design: We investigated two clinically applicable MET inhibitors: crizotinib, an ATP-competitive small molecule inhibitor of MET, and onartuzumab, a monovalent monoclonal antibody that binds to the extracellular domain of the MET receptor. We developed new MET inhibitor-resistant cells lines and animal models and used reverse phase protein arrays (RPPA) and functional assays to uncover the compensatory pathways in MET inhibitor-resistant GBM. RESULTS We identified critical proteins that were altered in MET inhibitor-resistant GBM including mTOR, FGFR1, EGFR, STAT3, and COX-2. Simultaneous inhibition of MET and one of these upregulated proteins led to increased cell death and inhibition of cell proliferation in resistant cells compared with either agent alone. In addition, in vivo treatment of mice bearing MET-resistant orthotopic xenografts with COX-2 or FGFR pharmacological inhibitors in combination with MET inhibitor restored sensitivity to MET inhibition and significantly inhibited tumor growth. CONCLUSIONS These data uncover the molecular basis of adaptive resistance to MET inhibitors and identify new FDA-approved multidrug therapeutic combinations that can overcome resistance.
Collapse
Affiliation(s)
- Nichola Cruickshanks
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Ying Zhang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Sarah Hine
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Myron Gibert
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Fang Yuan
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Madison Oxford
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Cassandra Grello
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Mary Pahuski
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Collin Dube
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Fadila Guessous
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,University Mohammed 6 for Health Sciences, Casablanca, Morocco
| | - Baomin Wang
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Ciana Deveau
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Karim Saoud
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Isela Gallagher
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - Julia Wulfkuhle
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - David Schiff
- Department of Neurology, University of Virginia, Charlottesville, Virginia
| | - See Phan
- Genentech Inc. South San Francisco, California
| | - Emanuel Petricoin
- George Mason University Center for Applied Proteomics and Molecular Medicine, Manassas, Virginia
| | - Roger Abounader
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia. .,Department of Neurology, University of Virginia, Charlottesville, Virginia.,The Cancer Center, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
13
|
Doi T, Yamaguchi K, Komatsu Y, Muro K, Nishina T, Nakajima TE, Tang R, Yang H, Zhang Y, Jung AS, Ang A, Yasui H. A Phase 1/1b tolerability study of rilotumumab alone or in combination with cisplatin and capecitabine in Japanese patients with gastric cancer. Jpn J Clin Oncol 2018; 47:1002-1009. [PMID: 28973403 DOI: 10.1093/jjco/hyx114] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022] Open
Abstract
Objective To evaluate the safety (including adverse events and dose-limiting toxicities [DLTs]), tolerability, pharmacokinetics and antitumor activity of the investigational MET inhibitor rilotumumab alone in patients with advanced solid tumors (Part 1) or in combination with cisplatin plus capecitabine (CX) in patients with MET-positive advanced gastric or gastroesophageal junction cancer (Part 2). Methods Adult patients received 10 or 20 mg/kg intravenous (IV) rilotumumab every 2 weeks (Part 1) or 15 mg/kg IV rilotumumab every 3 weeks plus 80 mg/m2 cisplatin on Day 1 and 1000 mg/m2 capecitabine twice daily on Days 1-14 of every 21-day cycle (Part 2). Results Nine patients enrolled in Part 1; 12 patients enrolled in Part 2. One DLT occurred (Grade 3 decreased appetite and stomatitis [Part 2]). Adverse events related to any treatment occurred in 17 patients (81%) and were Grade ≥3 in nine patients (43%). Rilotumumab pharmacokinetics appeared linear, and exposure was unaffected by CX. No patient who received rilotumumab monotherapy in Part 1 had a response. In Part 2, five of eight patients (63%) with measureable disease at baseline had a partial response and two patients (25%) had stable disease; median (95% CI) progression-free survival was 7.0 (2.4-15.4) months; overall survival was 18.2 (5.6-20.4) months. Conclusions In combination with CX, rilotumumab appeared tolerable and showed antitumor activity in Japanese patients with MET-positive gastric/gastroesophageal junction cancer. However, owing to the results of recent Phase 3 trials of MET inhibitors (including rilotumumab), further development of rilotumumab in this setting is not being pursued. ClinicalTrials.gov Identifier: NCT01791374.
Collapse
Affiliation(s)
- Toshihiko Doi
- National Cancer Center Hospital East, Kashiwa, Chiba
| | - Kensei Yamaguchi
- Saitama Cancer Center, Kita Adachi-gun, Saitama.,Cancer Institute Hospital of Japanese Foundation for Cancer Research, Ariake, Tokyo
| | | | - Kei Muro
- Aichi Cancer Center Hospital, Nagoya
| | | | | | - Rui Tang
- Amgen Inc., Thousand Oaks, CA, USA
| | - Hui Yang
- Amgen Inc., Thousand Oaks, CA, USA
| | | | | | | | | |
Collapse
|
14
|
A non-randomized, open-label, single-arm, Phase 2 study of emibetuzumab in Asian patients with MET diagnostic positive, advanced gastric cancer. Cancer Chemother Pharmacol 2017; 80:1197-1207. [PMID: 29071414 PMCID: PMC5686250 DOI: 10.1007/s00280-017-3445-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 09/22/2017] [Indexed: 12/14/2022]
Abstract
Purpose Mesenchymal–epithelial transition factor (MET) is expressed in gastric cancer and associated with poor clinical outcomes. We assessed activity, safety, and pharmacokinetics of emibetuzumab, a bivalent monoclonal anti-MET antibody that blocks ligand-dependent and ligand-independent MET signaling. Methods This non-randomized, single-arm, Phase 2 study enrolled Asian patients with MET diagnostic positive advanced gastric adenocarcinoma. Emibetuzumab (2000 mg, intravenous) was given on days 1 and 15 (28-day cycle). The primary endpoint was 8-week progression-free survival rate. Secondary objectives included safety, pharmacokinetics, overall survival, and change in tumor size. Results Tumors from 65 patients were immunohistochemically screened to enroll 15 MET diagnostic positive patients (23% positivity; 8 Japanese, 7 Korean; 10 male). Eight-week progression-free survival rate was 0.47 (70% CI, 0.33–0.59). Disease control rate was 40% (target lesion decreases, three patients; no complete/partial responses according to RECIST). Median overall survival was 17.1 weeks (95% CI, 6.3–not achievable). No serious emibetuzumab-related adverse events or new safety signals emerged. Grade ≥ 3 possibly drug-related adverse events were hyperkalemia, hyponatremia, and hyperuricemia (one each). Emibetuzumab’s pharmacokinetics profile was similar to that observed previously. MET expression and clinical outcomes were not obviously associated. Conclusion Emibetuzumab was well tolerated with limited single-agent activity in advanced gastric adenocarcinoma.
Collapse
|
15
|
Catenacci DVT, Tebbutt NC, Davidenko I, Murad AM, Al-Batran SE, Ilson DH, Tjulandin S, Gotovkin E, Karaszewska B, Bondarenko I, Tejani MA, Udrea AA, Tehfe M, De Vita F, Turkington C, Tang R, Ang A, Zhang Y, Hoang T, Sidhu R, Cunningham D. Rilotumumab plus epirubicin, cisplatin, and capecitabine as first-line therapy in advanced MET-positive gastric or gastro-oesophageal junction cancer (RILOMET-1): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Oncol 2017; 18:1467-1482. [PMID: 28958504 DOI: 10.1016/s1470-2045(17)30566-1] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/13/2017] [Accepted: 07/17/2017] [Indexed: 12/27/2022]
Abstract
BACKGROUND Rilotumumab is a fully human monoclonal antibody that selectively targets the ligand of the MET receptor, hepatocyte growth factor (HGF). We aimed to assess the efficacy, safety, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine, and to assess potential biomarkers, in patients with advanced MET-positive gastric or gastro-oesophageal junction adenocarcinoma. METHODS This multicentre, randomised, double-blind, placebo-controlled, phase 3 study was done at 152 centres in 27 countries. We recruited adults (aged ≥18 years) with unresectable locally advanced or metastatic gastric or gastro-oesophageal junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, MET-positive tumours (≥25% of tumour cells with membrane staining of ≥1+ staining intensity), and evaluable disease, who had not received previous systemic therapy. Eligible patients were randomly assigned (1:1) via a computerised voice response system to receive rilotumumab 15 mg/kg intravenously or placebo in combination with open-label chemotherapy (epirubicin 50 mg/m2 intravenously; cisplatin 60 mg/m2 intravenously; capecitabine 625 mg/m2 orally twice daily) in 21-day cycles for up to ten cycles. After completion of chemotherapy, patients continued to receive rilotumumab or placebo monotherapy until disease progression, intolerability, withdrawal of consent, or study termination. Randomisation was stratified by disease extent and ECOG performance status. Both patients and physicians were masked to study treatment assignment. The primary endpoint was overall survival, analysed by intention to treat. We report the final analysis. This study is registered with ClinicalTrials.gov, number NCT01697072. FINDINGS Between Nov 7, 2012, and Nov 21, 2014, 609 patients were randomly assigned to rilotumumab plus epirubicin, cisplatin, and capecitabine (rilotumumab group; n=304) or placebo plus epirubicin, cisplatin, and capecitabine (placebo group; n=305). Study treatment was stopped early after an independent data monitoring committee found a higher number of deaths in the rilotumumab group than in the placebo group; all patients in the rilotumumab group subsequently discontinued all study treatment. Median follow-up was 7·7 months (IQR 3·6-12·0) for patients in the rilotumumab group and 9·4 months (5·3-13·1) for patients in the placebo group. Median overall survival was 8·8 months (95% CI 7·7-10·2) in the rilotumumab group compared with 10·7 months (9·6-12·4) in the placebo group (stratified hazard ratio 1·34, 95% CI 1·10-1·63; p=0·003). The most common grade 3 or worse adverse events in the rilotumumab and placebo groups were neutropenia (86 [29%] of 298 patients vs 97 [32%] of 299 patients), anaemia (37 [12%] vs 43 [14%]), and fatigue (30 [10%] vs 35 [12%]). The frequency of serious adverse events was similar in the rilotumumab and placebo groups (142 [48%] vs 149 [50%]). More deaths due to adverse events occurred in the rilotumumab group than the placebo group (42 [14%] vs 31 [10%]). In the rilotumumab group, 33 (11%) of 298 patients had fatal adverse events due to disease progression, and nine (3%) had fatal events not due to disease progression. In the placebo group, 23 (8%) of 299 patients had fatal adverse events due to disease progression, and eight (3%) had fatal events not due to disease progression. INTERPRETATION Ligand-blocking inhibition of the MET pathway with rilotumumab is not effective in improving clinical outcomes in patients with MET-positive gastric or gastro-oesophageal adenocarcinoma. FUNDING Amgen.
Collapse
Affiliation(s)
| | | | - Irina Davidenko
- State Budgetary Healthcare Institution, Clinical Oncology Dispensary #1, Krasnodar Region Ministry of Healthcare, Krasnodar, Russia
| | - André M Murad
- Hospital das Clínicas da Universidade Federal de Minas Gerais, Horizonte, Brazil
| | - Salah-Eddin Al-Batran
- Institute of Clinical Cancer Research, Krankenhaus Nordwest, University Cancer Center, Frankfurt, Germany
| | - David H Ilson
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Evengy Gotovkin
- Regional Budgetary Institution of Public Health Ivanovo Regional Oncology Dispensary, Ivanovo, Russia
| | | | - Igor Bondarenko
- Dnipropetrovsk Medical Academy, City Multifield Clinical Hospital 4, Dnipropetrovsk, Ukraine
| | - Mohamedtaki A Tejani
- University of Rochester Medical Center, James P Wilmot Cancer Center, Rochester, NY, USA
| | | | - Mustapha Tehfe
- Centre Hospitalier de L'Universite de Montreal Notre-Dame, Montreal, QC, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Bendell JC, Hochster H, Hart LL, Firdaus I, Mace JR, McFarlane JJ, Kozloff M, Catenacci D, Hsu JJ, Hack SP, Shames DS, Phan SC, Koeppen H, Cohn AL. A Phase II Randomized Trial (GO27827) of First-Line FOLFOX Plus Bevacizumab with or Without the MET Inhibitor Onartuzumab in Patients with Metastatic Colorectal Cancer. Oncologist 2017; 22:264-271. [PMID: 28209746 PMCID: PMC5344636 DOI: 10.1634/theoncologist.2016-0223] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Dysregulated hepatocyte growth factor/mesenchymal-epithelial transition (MET) signaling is associated with poor prognosis and resistance to vascular endothelial growth factor inhibition in metastatic colorectal cancer (mCRC). We report outcomes from a double-blind, multicenter phase II trial of the MET inhibitor onartuzumab in combination with mFOLFOX-6 and bevacizumab for mCRC (GO27827; NCT01418222). MATERIALS AND METHODS Patients were randomized 1:1 to receive onartuzumab (10 mg/kg intravenously [IV]) or placebo plus mFOLFOX-6 and bevacizumab (5 mg/kg IV). Oxaliplatin was given for 8-12 cycles; other agents were continued until disease progression, unacceptable toxicity, or death. The primary endpoint was progression-free survival (PFS) in the intent-to-treat (ITT) and MET immunohistochemistry (IHC) expression-positive populations. RESULTS Between September 2011 and November 2012, 194 patients were enrolled. In September 2013, an interim analysis recommended stopping onartuzumab treatment due to lack of efficacy. At the time of the final analysis in February 2014, no significant improvement in PFS was seen with onartuzumab versus placebo in either the ITT or MET IHC-positive populations. An improvement in PFS was noted in the MET IHC-negative population. Neither overall survival nor response rate was improved with onartuzumab. The incidence of fatigue, peripheral edema, and deep vein thrombosis was increased with onartuzumab relative to placebo. CONCLUSION Onartuzumab combined with mFOLFOX-6 and bevacizumab did not significantly improve efficacy outcomes in either the ITT or MET IHC-positive populations. MET expression by IHC was not a predictive biomarker in this setting. The Oncologist 2017;22:264-271 IMPLICATIONS FOR PRACTICE: The addition of onartuzumab to mFOLFOX-6 plus bevacizumab did not improve outcomes in patients with previously untreated metastatic colorectal cancer in this randomized, phase II study. Although initial results with onartuzumab were promising, a number of phase II/III clinical trials have reported a lack of improvement in efficacy with onartuzumab combined with standard-of-care therapies in several tumor types. Furthermore, negative study data have been published for rilotumumab and ficlatuzumab, both of which block hepatocyte growth factor binding to the mesenchymal-epithelial transition (MET) receptor. MET immunohistochemistry was not a predictive biomarker. It remains to be seen if other biomarkers or small molecule inhibitors may be more appropriate for inhibiting this oncogenic pathway.
Collapse
Affiliation(s)
- Johanna C Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, Tennessee, USA
| | | | - Lowell L Hart
- Sarah Cannon Research Institute/Florida Cancer Specialists, Fort Myers, Florida, USA
| | - Irfan Firdaus
- Sarah Cannon Research Institute/Oncology Hematology Care, Cincinnati, Ohio, USA
| | - Joseph R Mace
- Sarah Cannon Research Institute/Florida Cancer Specialists, St Petersburg, Florida, USA
| | - Joshua J McFarlane
- Sarah Cannon Research Institute/Virginia Cancer Institute, Richmond, Virginia, USA
| | | | | | - Jessie J Hsu
- Genentech Inc., South San Francisco, California, USA
| | | | | | - See-Chun Phan
- Genentech Inc., South San Francisco, California, USA
| | | | - Allen L Cohn
- Rocky Mountain Cancer Center, Denver, Colorado, USA
| |
Collapse
|
17
|
Spigel DR, Edelman MJ, O'Byrne K, Paz-Ares L, Mocci S, Phan S, Shames DS, Smith D, Yu W, Paton VE, Mok T. Results From the Phase III Randomized Trial of Onartuzumab Plus Erlotinib Versus Erlotinib in Previously Treated Stage IIIB or IV Non-Small-Cell Lung Cancer: METLung. J Clin Oncol 2016; 35:412-420. [PMID: 27937096 DOI: 10.1200/jco.2016.69.2160] [Citation(s) in RCA: 215] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Purpose The phase III OAM4971g study (METLung) examined the efficacy and safety of onartuzumab plus erlotinib in patients with locally advanced or metastatic non-small-cell lung cancer selected by MET immunohistochemistry whose disease had progressed after treatment with a platinum-based chemotherapy regimen. Patients and Methods Patients were randomly assigned at a one-to-one ratio to receive onartuzumab (15 mg/kg intravenously on day 1 of each 21-day cycle) plus daily oral erlotinib 150 mg or intravenous placebo plus daily oral erlotinib 150 mg. The primary end point was overall survival (OS) in the intent-to-treat population. Secondary end points included median progression-free survival, overall response rate, biomarker analysis, and safety. Results A total of 499 patients were enrolled (onartuzumab, n = 250; placebo, n = 249). Median OS was 6.8 versus 9.1 months for onartuzumab versus placebo (stratified hazard ratio [HR], 1.27; 95% CI, 0.98 to 1.65; P = .067), with a greater number of deaths in the onartuzumab arm (130 [52%] v 114 [46%]). Median progression-free survival was 2.7 versus 2.6 months (stratified HR, 0.99; 95% CI, 0.81 to 1.20; P = .92), and overall response rate was 8.4% and 9.6% for onartuzumab versus placebo, respectively. Exploratory analyses using MET fluorescence in situ hybridization status and gene expression showed no benefit for onartuzumab; patients with EGFR mutations showed a trend toward shorter OS with onartuzumab treatment (HR, 4.68; 95% CI, 0.97 to 22.63). Grade 3 to 5 adverse events were reported by 56.0% and 51.2% of patients, with serious AEs in 33.9% and 30.7%, for experimental versus control arms, respectively. Conclusion Onartuzumab plus erlotinib did not improve clinical outcomes, with shorter OS in the onartuzumab arm, compared with erlotinib in patients with MET-positive non-small-cell lung cancer.
Collapse
Affiliation(s)
- David R Spigel
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Martin J Edelman
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Kenneth O'Byrne
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Luis Paz-Ares
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Simonetta Mocci
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - See Phan
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - David S Shames
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Dustin Smith
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Wei Yu
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Virginia E Paton
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | - Tony Mok
- David R. Spigel, Sarah Cannon Research Institute, Nashville, TN; Martin J. Edelman, University of Maryland Greenebaum Cancer Center, Baltimore, MD; Kenneth O'Byrne, Queensland University of Technology, Brisbane, Queensland, Australia; Luis Paz-Ares, Hospital Universitario Doce de Octubre and Centro Nacional de Investigaciones Oncológicas, Madrid, Spain; Simonetta Mocci, See Phan, David S. Shames, Dustin Smith, Wei Yu, and Virginia E. Paton, Genentech, South San Francisco, CA; and Tony Mok, Chinese University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| |
Collapse
|
18
|
Ye S, Li J, Hao K, Yan J, Zhou H. The Efficacy and Risk Profile of c-Met inhibitors in Non-small Cell Lung Cancer: a Meta-analysis. Sci Rep 2016; 6:35770. [PMID: 27786238 PMCID: PMC5081544 DOI: 10.1038/srep35770] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Accepted: 10/03/2016] [Indexed: 12/17/2022] Open
Abstract
c-MET inhibitors are considered as a kind of novel drugs in non-small cell lung cancer (NSCLC) treatment. However, the results of different clinical studies involving c-MET inhibitors were not consistent. In this report, we performed Meta-analysis to investigate the beneficial and harmful effects of these drugs from 9 studies including 1611 patients in target drug groups and 1605 patients in control groups. As a result, patients in target drugs group had longer progression free survival (PFS) (HR 0.80, 95% CI 0.66-0.99, p = 0.04) but not overall survival (OS) than those in control group, especially in Asian (HR 0.57, 95% CI 0.42-0.76, p < 0.001), Non-squamous (HR 0.79, 95% CI 0.64-0.97, p = 0.03), Phase III (HR 0.66, 95% CI 0.50-0.86, p = 0.002), previous treated (HR 0.77, 95% CI 0.63-0.95, p = 0.01) and small molecular compounds subgroups (HR 0.62, 95% CI 0.50-0.78, p < 0.001). In addition, target drugs did not affect the objective response rate (ORR) but improved disease control rate (DCR) (RR 1.22, 95% CI 1.02-1.46, p = 0.03) of NSCLC patients. Our study first indicated that targeting c-MET therapies improved PFS and DCR in advanced or metastatic NSCLC patients, especially in previous treated Asian patients with adenocarcinoma.
Collapse
Affiliation(s)
- Sa Ye
- Department of Respiratory Medicine, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
| | - Jiuke Li
- Department of Ophthalmology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ke Hao
- Department of Blood Transfusion, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
| | - Jianping Yan
- Department of Respiratory Medicine, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
| | - Hongbin Zhou
- Department of Respiratory Medicine, Zhejiang Provincial People’s Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
19
|
Castoldi R, Schanzer J, Panke C, Jucknischke U, Neubert NJ, Croasdale R, Scheuer W, Auer J, Klein C, Niederfellner G, Kobold S, Sustmann C. TetraMabs: simultaneous targeting of four oncogenic receptor tyrosine kinases for tumor growth inhibition in heterogeneous tumor cell populations. Protein Eng Des Sel 2016; 29:467-475. [PMID: 27578890 PMCID: PMC5036864 DOI: 10.1093/protein/gzw037] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 07/11/2016] [Indexed: 11/14/2022] Open
Abstract
Monoclonal antibody-based targeted tumor therapy has greatly improved treatment options for patients. Antibodies against oncogenic receptor tyrosine kinases (RTKs), especially the ErbB receptor family, are prominent examples. However, long-term efficacy of such antibodies is limited by resistance mechanisms. Tumor evasion by a priori or acquired activation of other kinases is often causative for this phenomenon. These findings led to an increasing number of combination approaches either within a protein family, e.g. the ErbB family or by targeting RTKs of different phylogenetic origin like HER1 and cMet or HER1 and IGF1R. Progress in antibody engineering technology enabled generation of clinical grade bispecific antibodies (BsAbs) to design drugs inherently addressing such resistance mechanisms. Limited data are available on multi-specific antibodies targeting three or more RTKs. In the present study, we have evaluated the cloning, eukaryotic expression and purification of tetraspecific, tetravalent Fc-containing antibodies targeting HER3, cMet, HER1 and IGF1R. The antibodies are based on the combination of single-chain Fab and Fv fragments in an IgG1 antibody format enhanced by the knob-into-hole technology. They are non-agonistic and inhibit tumor cell growth comparable to the combination of four parental antibodies. Importantly, TetraMabs show improved apoptosis induction and tumor growth inhibition over individual monospecific or BsAbs in cellular assays. In addition, a mimicry assay to reflect heterogeneous expression of antigens in a tumor mass was established. With this novel in vitro assay, we can demonstrate the superiority of a tetraspecific antibody to bispecific tumor antigen-binding antibodies in early pre-clinical development.
Collapse
Affiliation(s)
- Raffaella Castoldi
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Jürgen Schanzer
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Christian Panke
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Ute Jucknischke
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Natalie J Neubert
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Rebecca Croasdale
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Werner Scheuer
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Johannes Auer
- pRED, Roche Pharma Research & Early Development, Roche Large Molecule Research, Roche Innovation Center, Munich, Nonnenwald 2, 82377 Penzberg, Germany
| | - Christian Klein
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Zuerich, Switzerland, Wagistrasse 18, 8952 Schlieren
| | - Gerhard Niederfellner
- pRED, Roche Pharma Research & Early Development, Roche Innovation Center, Munich, Germany
| | - Sebastian Kobold
- Center of Integrated Protein Science Munich (CIPS-M) and Division of Clinical Pharmacology, Department of Medicine IV, Klinikum der Universität München, Lindwurmstraße 2a, 80337 Munich, Germany, Member of the German Center for Lung Research (DZL)
| | - Claudio Sustmann
- pRED, Roche Pharma Research & Early Development, Roche Large Molecule Research, Roche Innovation Center, Munich, Nonnenwald 2, 82377 Penzberg, Germany
| |
Collapse
|
20
|
Van Der Steen N, Giovannetti E, Pauwels P, Peters GJ, Hong DS, Cappuzzo F, Hirsch FR, Rolfo C. cMET Exon 14 Skipping: From the Structure to the Clinic. J Thorac Oncol 2016; 11:1423-32. [PMID: 27223456 DOI: 10.1016/j.jtho.2016.05.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 05/02/2016] [Accepted: 05/03/2016] [Indexed: 12/28/2022]
Abstract
The abnormal stimulation of the multiple signal transduction pathways downstream of the receptor tyrosine kinase mesenchymal-epithelial transition factor (cMET) promotes cellular transformation, tumor motility, and invasion. Therefore, cMET has been the focus of prognostic and therapeutic studies in different tumor types, including non-small cell lung cancer. In particular, several cMET inhibitors have been developed as innovative therapeutic candidates and are currently under investigation in clinical trials. However, one of the challenges in establishing effective targeted treatments against cMET remains the accurate identification of biomarkers for the selection of responsive subsets of patients. Recently, splice site mutations have been discovered in cMET that lead to the skipping of exon 14, impairing the breakdown of the receptor. Patients with NSCLC who are carrying this splice variant typically overexpress the cMET receptor and show a response to small molecule inhibitors of cMET. Here, we review the main differences at the structural level between the wild-type and the splice variants of cMET and their influence on cMET signaling. We clarify the reason why this variant responds to small molecule inhibitors and their prognostic/predictive role.
Collapse
Affiliation(s)
- Nele Van Der Steen
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Department of Pathology, Antwerp University Hospital, Edegem, Antwerp, Belgium; Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands; Cancer Pharmacology Lab, Italian Association for Cancer Research Start-Up Unit, University of Pisa, Hospital of Cisanello, Pisa, Italy
| | - Patrick Pauwels
- Department of Pathology, Antwerp University Hospital, Edegem, Antwerp, Belgium; Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - David S Hong
- Investigational Cancer Therapeutics, Division of Cancer Medicine, The University of Texas M. D. Anderson Cancer Center, Houston, Texas
| | | | - Fred R Hirsch
- Division of Medical Oncology, University of Colorado, Aurora, Colorado
| | - Christian Rolfo
- Center for Oncological Research, University of Antwerp, Wilrijk, Antwerp, Belgium; Phase I Early Clinical Trials Unit, Oncology Department, Antwerp University Hospital, Antwerp, Belgium.
| |
Collapse
|