1
|
Terlecka P, Krawczyk P, Grenda A, Milanowski J. MET Gene Dysregulation as a Promising Therapeutic Target in Lung Cancer-A Review. J Pers Med 2021; 11:1370. [PMID: 34945842 PMCID: PMC8705301 DOI: 10.3390/jpm11121370] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/20/2021] [Accepted: 12/13/2021] [Indexed: 11/23/2022] Open
Abstract
Several molecular abnormalities in the MET gene have been identified, including overexpression, amplification, point mutations, and "skipping mutation" in exon 14. Even though deregulated MET signaling occurs rarely in non-small cell lung cancer (NSCLC), it possesses tumorigenic activity. Since the discovery of the significant role played by MET dysregulations in resistance to epidermal growth factor receptor tyrosine kinase inhibitors (EGFR TKI), many clinical trials have been focused on mechanisms underlying this acquired resistance. Therefore, new therapeutic strategies are being considered in the personalized therapy of NSCLC patients carrying MET abnormalities. First, MET kinase inhibitors (tepotinib and capmatinib) have been shown to be effective in the first and subsequent lines of treatment in NSCLC patients with "skipping mutations" in exon 14 of MET gene. In this article, the authors show the role of MET signaling pathway alterations and describe the results of clinical trials with MET inhibitors in NSCLC patients.
Collapse
Affiliation(s)
- Paulina Terlecka
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (P.K.); (A.G.); (J.M.)
| | | | | | | |
Collapse
|
2
|
Phase I Study of Ficlatuzumab and Cetuximab in Cetuximab-Resistant, Recurrent/Metastatic Head and Neck Cancer. Cancers (Basel) 2020; 12:cancers12061537. [PMID: 32545260 PMCID: PMC7352434 DOI: 10.3390/cancers12061537] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 06/03/2020] [Accepted: 06/08/2020] [Indexed: 12/18/2022] Open
Abstract
Cetuximab, an anti-EGFR monoclonal antibody (mAb), is approved for advanced head and neck squamous cell carcinoma (HNSCC) but benefits a minority. An established tumor-intrinsic resistance mechanism is cross-talk between the EGFR and hepatocyte growth factor (HGF)/cMet pathways. Dual pathway inhibition may overcome cetuximab resistance. This Phase I study evaluated the combination of cetuximab and ficlatuzumab, an anti-HGF mAb, in patients with recurrent/metastatic HNSCC. The primary objective was to establish the recommended Phase II dose (RP2D). Secondary objectives included overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Mechanistic tumor-intrinsic and immune biomarkers were explored. Thirteen patients enrolled with no dose-limiting toxicities observed at any dose tier. Three evaluable patients were treated at Tier 1 and nine at Tier 2, which was determined to be the RP2D (cetuximab 500 mg/m2 and ficlatuzumab 20 mg/kg every 2 weeks). Median PFS and OS were 5.4 (90% CI = 1.9–11.4) and 8.9 (90% CI = 2.7–15.2) months, respectively, with a confirmed ORR of 2 of 12 (17%; 90% CI = 6–40%). High circulating soluble cMet levels correlated with poor survival. An increase in peripheral T cells, particularly the CD8+ subset, was associated with treatment response whereas progression was associated with expansion of a distinct myeloid population. This well-tolerated combination demonstrated promising activity in cetuximab-resistant, advanced HNSCC.
Collapse
|
3
|
Feng W, Zhu X. Efficacy prediction of targeted therapy for gastric cancer: The current status (Review). Mol Med Rep 2018; 18:1238-1246. [PMID: 29901092 DOI: 10.3892/mmr.2018.9145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 05/03/2018] [Indexed: 11/05/2022] Open
Abstract
Despite significant progress in the treatment of gastric cancer (GC), the prognosis remains poor and the mortality is high. Targeted drugs have been incorporated into routine treatment to improve treatment efficacy. However, the therapy response is still below 50%. Therefore, there is a need to identify predictive factors for patient response to a specific drug in order to improve the efficacy of drug therapy. The present article reviewed the predictive factors for target therapy in GC, including epidermal growth factor receptor, human epidermal receptor 2, vascular endothelial growth factor family, molecules in the mesenchymal‑epithelial transition pathway and the mammalian target of rapamycin. Additionally, the present review described the interactions between these molecules and signaling pathways.
Collapse
Affiliation(s)
- Wanjing Feng
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| | - Xiaodong Zhu
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, P.R. China
| |
Collapse
|
4
|
Chivu-Economescu M, Matei L, Necula LG, Dragu DL, Bleotu C, Diaconu CC. New therapeutic options opened by the molecular classification of gastric cancer. World J Gastroenterol 2018; 24:1942-1961. [PMID: 29760539 PMCID: PMC5949709 DOI: 10.3748/wjg.v24.i18.1942] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 04/12/2018] [Accepted: 04/23/2018] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer (GC) is one of the most lethal and aggressive cancers, being the third cause of cancer related death worldwide. Even with radical gastrectomy and the latest generation of molecular chemotherapeutics, the numbers of recurrence and mortality remains high. This is due to its biological heterogeneity based on the interaction between multiple factors, from genomic to environmental factors, diet or infections with various pathogens. Therefore, understanding the molecular characteristics at a genomic level is critical to develop new treatment strategies. Recent advances in GC molecular classification provide the unique opportunity to improve GC therapy by exploiting the biomarkers and developing novel targeted therapy specific to each subtype. This article highlights the molecular characteristics of each subtype of gastric cancer that could be considered in shaping a therapeutic decision, and also presents the completed and ongoing clinical trials addressed to those targets. The implementation of the novel molecular classification system will allow a preliminary patient selection for clinical trials, a mandatory issue if it is desired to test the efficacy of a certain inhibitor to the given target. This will represent a substantial advance as well as a powerful tool for targeted therapy. Nevertheless, translating the scientific results into new personalized treatment opportunities is needed in order to improve clinical care, the survival and quality of life of patients with GC.
Collapse
Affiliation(s)
- Mihaela Chivu-Economescu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Lilia Matei
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Laura G Necula
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
- Nicolae Cajal Institute, Titu Maiorescu University, Bucharest 040441, Romania
| | - Denisa L Dragu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Coralia Bleotu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| | - Carmen C Diaconu
- Department of Cellular and Molecular Pathology, Stefan S. Nicolau Institute of Virology, Bucharest 030304, Romania
| |
Collapse
|
5
|
Lucas AT, Price LSL, Schorzman AN, Storrie M, Piscitelli JA, Razo J, Zamboni WC. Factors Affecting the Pharmacology of Antibody-Drug Conjugates. Antibodies (Basel) 2018; 7:E10. [PMID: 31544862 PMCID: PMC6698819 DOI: 10.3390/antib7010010] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 01/30/2018] [Accepted: 02/01/2018] [Indexed: 12/12/2022] Open
Abstract
Major advances in therapeutic proteins, including antibody-drug conjugates (ADCs), have created revolutionary drug delivery systems in cancer over the past decade. While these immunoconjugate agents provide several advantages compared to their small-molecule counterparts, their clinical use is still in its infancy. The considerations in their development and clinical use are complex, and consist of multiple components and variables that can affect the pharmacologic characteristics. It is critical to understand the mechanisms employed by ADCs in navigating biological barriers and how these factors affect their biodistribution, delivery to tumors, efficacy, and toxicity. Thus, future studies are warranted to better understand the complex pharmacology and interaction between ADC carriers and biological systems, such as the mononuclear phagocyte system (MPS) and tumor microenvironment. This review provides an overview of factors that affect the pharmacologic profiles of ADC therapies that are currently in clinical use and development.
Collapse
Affiliation(s)
- Andrew T Lucas
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Lauren S L Price
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Allison N Schorzman
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - Mallory Storrie
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | | | - Juan Razo
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
| | - William C Zamboni
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
- UNC Eshelman School of Pharmacy, Chapel Hill, NC 27599, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| |
Collapse
|
6
|
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
|
7
|
Glisson B, Besse B, Dols MC, Dubey S, Schupp M, Jain R, Jiang Y, Menon H, Nackaerts K, Orlov S, Paz-Ares L, Ramlau R, Tang R, Zhang Y, Zhu M. A Randomized, Placebo-Controlled, Phase 1b/2 Study of Rilotumumab or Ganitumab in Combination With Platinum-Based Chemotherapy as First-Line Treatment for Extensive-Stage Small-Cell Lung Cancer. Clin Lung Cancer 2017; 18:615-625.e8. [DOI: 10.1016/j.cllc.2017.05.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Revised: 04/26/2017] [Accepted: 05/02/2017] [Indexed: 11/28/2022]
|
8
|
Tarhini AA, Rafique I, Floros T, Tran P, Gooding WE, Villaruz LC, Burns TF, Friedland DM, Petro DP, Farooqui M, Gomez-Garcia J, Gaither-Davis A, Dacic S, Argiris A, Socinski MA, Stabile LP, Siegfried JM. Phase 1/2 study of rilotumumab (AMG 102), a hepatocyte growth factor inhibitor, and erlotinib in patients with advanced non-small cell lung cancer. Cancer 2017; 123:2936-2944. [PMID: 28472537 DOI: 10.1002/cncr.30717] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/09/2017] [Accepted: 01/11/2017] [Indexed: 11/06/2022]
Abstract
BACKGROUND Activation of the mesenchymal-epidermal transition factor (MET) tyrosine kinase and its ligand, hepatocyte growth factor (HGF), is implicated in resistance to epidermal growth factor receptor (EGFR) inhibitors. In this phase 1/2 trial, rilotumumab (an anti-HGF antibody) combined with erlotinib was evaluated in patients with metastatic, previously treated non-small cell lung cancer. METHODS In phase 1, a dose de-escalation design was adopted with rilotumumab starting at 15 mg/kg intravenously every 3 weeks and oral erlotinib 150 mg daily. In phase 2, the disease control rate (DCR) (according to Response Evaluation Criteria in Solid Tumors) of the combination was evaluated using a Simon 2-stage design. The biomarkers examined included 10 plasma-circulating molecules associated with the EGFR and MET pathways. RESULTS Without indications for de-escalation, the recommended phase 2 dose was dose level 0. Overall, 45 response-evaluable patients were enrolled (13 with squamous carcinoma, 32 with adenocarcinoma; 2 had confirmed EGFR mutations, 33 had confirmed wild-type [WT] EGFR, and 7 had KRAS mutations). The DCR for all patients was 60% (90% confidence interval [CI], 47.1%-71.3%). Median progression-free survival was 2.6 months (90% CI, 1.4-2.7 months), and median overall survival was 6.6 months (90% CI, 5.6-8.9 months). Among patients with WT EGFR, the DCR was 60.6% (90% CI, 46.3%-73.3%), median progression-free survival was 2.6 months (90% CI, 1.4-2.7 months), and median overall survival was 7.0 months (90% CI, 5.6-13.4 months). Elevated baseline levels of neuregulin 1 were associated with longer progression-free survival (hazard ratio, 0.41; 95% CI, 0.19-0.87), whereas elevated amphiregulin levels were associated with more rapid progression (hazard ratio, 2.14; 95% CI, 1.48-3.08). CONCLUSIONS Combined rilotumumab and erlotinib had an acceptable safety profile, and the DCR met the prespecified criteria for success. In the EGFR WT group, the DCR exceeded published reports for erlotinib alone. High circulating levels of neuregulin 1 may indicate sensitivity to this combination. Cancer 2017;123:2936-44. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Ahmad A Tarhini
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Imran Rafique
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Theofanis Floros
- Department of Medicine, Athens Naval & Veterans Hospital, Athens, Greece
| | - Phu Tran
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - William E Gooding
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Liza C Villaruz
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Timothy F Burns
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania.,Department of Medicine, Athens Naval & Veterans Hospital, Athens, Greece
| | - David M Friedland
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Daniel P Petro
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Mariya Farooqui
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Jose Gomez-Garcia
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| | - Autumn Gaither-Davis
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sanja Dacic
- Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania.,University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Athanassios Argiris
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Mark A Socinski
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania
| | - Laura P Stabile
- University of Pittsburgh Medical Center and Cancer Institute, Pittsburgh, Pennsylvania.,Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Jill M Siegfried
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
9
|
Kim H, Hong SH, Kim JY, Kim IC, Park YW, Lee SJ, Song SW, Kim JJ, Park G, Kim TM, Kim YH, Park JB, Chung J, Kim IH. Preclinical development of a humanized neutralizing antibody targeting HGF. Exp Mol Med 2017; 49:e309. [PMID: 28336956 PMCID: PMC5382562 DOI: 10.1038/emm.2017.21] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 12/23/2016] [Indexed: 12/17/2022] Open
Abstract
Hepatocyte growth factor (HGF) and its receptor, cMET, play critical roles in cell proliferation, angiogenesis and invasion in a wide variety of cancers. We therefore examined the anti-tumor activity of the humanized monoclonal anti-HGF antibody, YYB-101, in nude mice bearing human glioblastoma xenografts as a single agent or in combination with temozolomide. HGF neutralization, The extracellular signal-related kinases 1 and 2 (ERK1/2) phosphorylation, and HGF-induced scattering were assessed in HGF-expressing cell lines treated with YYB-101. To support clinical development, we also evaluated the preclinical pharmacokinetics and toxicokinetics in cynomolgus monkeys, and human and cynomolgus monkey tissue was stained with YYB-101 to test tissue cross-reactivity. We found that YYB-101 inhibited cMET activation in vitro and suppressed tumor growth in the orthotopic mouse model of human glioblastoma. Combination treatment with YYB-101 and temozolomide decreased tumor growth and increased overall survival compared with the effects of either agent alone. Five cancer-related genes (TMEM119, FST, RSPO3, ROS1 and NBL1) were overexpressed in YYB-101-treated mice that showed tumor regrowth. In the tissue cross-reactivity assay, critical cross-reactivity was not observed. The terminal elimination half-life was 21.7 days. Taken together, the in vitro and in vivo data demonstrated the anti-tumor efficacy of YYB-101, which appeared to be mediated by blocking the HGF/cMET interaction. The preclinical pharmacokinetics, toxicokinetics and tissue cross-reactivity data support the clinical development of YYB-101 for advanced cancer.
Collapse
Affiliation(s)
- Hyori Kim
- Asan Institute for Life Sciences, Asan Medical Center, Songpa-gu, Seoul, Republic of Korea
| | - Sung Hee Hong
- National OncoVenture, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jung Yong Kim
- National OncoVenture, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - In-Chull Kim
- National OncoVenture, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Young-Whan Park
- National OncoVenture, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Song-Jae Lee
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co. Ltd., Guro-gu, Seoul, Republic of Korea
| | - Seong-Won Song
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co. Ltd., Guro-gu, Seoul, Republic of Korea
| | - Jung Ju Kim
- Yooyoung Central Research Institute, Yooyoung Pharmaceutical Co. Ltd., Guro-gu, Seoul, Republic of Korea
| | - Gunwoo Park
- Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Jongno-gu, Seoul, Republic of Korea.,Cancer Research Institute, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea
| | - Yun-Hee Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea.,Research Institute, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Jong Bae Park
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| | - Junho Chung
- Cancer Research Institute, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea.,Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Jongno-gu, Seoul, Republic of Korea
| | - In-Hoo Kim
- Graduate School of Cancer Science and Policy, National Cancer Center, Goyang-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
10
|
Zhang Y, Kuchimanchi M, Zhu M, Doshi S, Hoang T, Kasichayanula S. Assessment of pharmacokinetic interaction between rilotumumab and epirubicin, cisplatin and capecitabine (ECX) in a Phase 3 study in gastric cancer. Br J Clin Pharmacol 2016; 83:1048-1055. [PMID: 27966237 DOI: 10.1111/bcp.13179] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 10/20/2016] [Accepted: 10/27/2016] [Indexed: 02/06/2023] Open
Abstract
AIMS Rilotumumab is a fully human monoclonal antibody investigated for the treatment of MET-positive gastric cancer. The aim of this study was to evaluate the potential pharmacokinetic (PK)-based drug-drug interaction (DDI) between rilotumumab and epirubicin (E), cisplatin(C) and capecitabine (X). METHODS This was a Phase 3 double-blind, placebo-controlled study, in which rilotumumab, epirubicin and cisplatin were administered intravenously at 15 mg kg-1 , 50 mg m-2 , and 60 mg m-2 Q3W, respectively, while capecitabine was given orally at 625 mg m-2 twice daily. Rilotumumab PK samples were taken at pre-dose and at the end-of-infusion from all patients in cycles 1, 3, 5 and 7. ECX PK samples were taken in cycle 3 from patients who participated in the intensive PK assessment. ECX PK was assessed by non-compartmental (NCA) analyses and PK parameters were compared between two arms. Rilotumumab PK was assessed by comparing the observed rilotumumab serum concentrations with model-predicted concentrations using a population PK model developed from previous Phase 1 and Phase 2 studies. RESULTS The study enrolled 609 patients. ECX plasma concentrations in the presence and absence of rilotumumab were similar, as demonstrated by the geometric mean ratios for Cmax and AUC, which were close to 1.0, suggesting ECX PK was not affected by co-administration of rilotumumab. The observed rilotumumab serum concentrations were similar to the values predicted by population PK modelling on the basis of a prediction-corrected visual predictive check, indicating rilotumumab exposure was not affected by co-administration of ECX. CONCLUSIONS The results suggest lack of PK-based DDI between rilotumumab and ECX.
Collapse
Affiliation(s)
- Yilong Zhang
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Mita Kuchimanchi
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Min Zhu
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Sameer Doshi
- Clinical Pharmacology, Modeling and Simulation, Amgen Inc., Thousand Oaks, California, 91320
| | - Tien Hoang
- Clinical Development, Amgen Inc., Thousand Oaks, California, 91320
| | | |
Collapse
|
11
|
Huang F, Ma Z, Pollan S, Yuan X, Swartwood S, Gertych A, Rodriguez M, Mallick J, Bhele S, Guindi M, Dhall D, Walts AE, Bose S, de Peralta Venturina M, Marchevsky AM, Luthringer DJ, Feller SM, Berman B, Freeman MR, Alvord WG, Vande Woude G, Amin MB, Knudsen BS. Quantitative imaging for development of companion diagnostics to drugs targeting HGF/MET. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2016; 2:210-222. [PMID: 27785366 PMCID: PMC5068192 DOI: 10.1002/cjp2.49] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/07/2016] [Indexed: 02/06/2023]
Abstract
The limited clinical success of anti-HGF/MET drugs can be attributed to the lack of predictive biomarkers that adequately select patients for treatment. We demonstrate here that quantitative digital imaging of formalin fixed paraffin embedded tissues stained by immunohistochemistry can be used to measure signals from weakly staining antibodies and provides new opportunities to develop assays for detection of MET receptor activity. To establish a biomarker panel of MET activation, we employed seven antibodies measuring protein expression in the HGF/MET pathway in 20 cases and up to 80 cores from 18 human cancer types. The antibodies bind to epitopes in the extra (EC)- and intracellular (IC) domains of MET (MET4EC, SP44_METIC, D1C2_METIC), to MET-pY1234/pY1235, a marker of MET kinase activation, as well as to HGF, pSFK or pMAPK. Expression of HGF was determined in tumour cells (T_HGF) as well as in stroma surrounding cancer (St_HGF). Remarkably, MET4EC correlated more strongly with pMET (r = 0.47) than SP44_METIC (r = 0.21) or D1C2_METIC (r = 0.08) across 18 cancer types. In addition, correlation coefficients of pMET and T_HGF (r = 0.38) and pMET and pSFK (r = 0.56) were high. Prediction models of MET activation reveal cancer-type specific differences in performance of MET4EC, SP44_METIC and anti-HGF antibodies. Thus, we conclude that assays to predict the response to HGF/MET inhibitors require a cancer-type specific antibody selection and should be developed in those cancer types in which they are employed clinically.
Collapse
Affiliation(s)
- Fangjin Huang
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Zhaoxuan Ma
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Sara Pollan
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Xiaopu Yuan
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Steven Swartwood
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Arkadiusz Gertych
- Departments of Surgery Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Maria Rodriguez
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Jayati Mallick
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Sanica Bhele
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Maha Guindi
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Deepti Dhall
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Ann E Walts
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Shikha Bose
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Mariza de Peralta Venturina
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Alberto M Marchevsky
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Daniel J Luthringer
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Stephan M Feller
- Institute of Molecular Medicine, Martin-Luther-University 06120 Halle Germany
| | - Benjamin Berman
- Department of Biomedical Sciences Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Michael R Freeman
- Department of Biomedical SciencesCedars-Sinai Medical CenterLos AngelesCalifornia90048USA; Departments of SurgeryCedars-Sinai Medical CenterLos AngelesCalifornia90048USA; Cancer Biology Program, Departments of Medicine, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical CenterLos AngelesCalifornia90048USA
| | - W Gregory Alvord
- Data Management Services, Inc., National Cancer Institute at Frederick Frederick Maryland 21702 USA
| | - George Vande Woude
- Laboratory of Molecular Oncology Center for Cancer and Cell Biology, Van Andel Research Institute Grand Rapids Michigan 49503 USA
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine Cedars-Sinai Medical Center Los Angeles California 90048 USA
| | - Beatrice S Knudsen
- Department of Biomedical SciencesCedars-Sinai Medical CenterLos AngelesCalifornia90048USA; Department of Pathology and Laboratory MedicineCedars-Sinai Medical CenterLos AngelesCalifornia90048USA
| |
Collapse
|