1
|
Cecchi F, Rex K, Schmidt J, Vocke CD, Lee YH, Burkett S, Baker D, Damore MA, Coxon A, Burgess TL, Bottaro DP. Rilotumumab Resistance Acquired by Intracrine Hepatocyte Growth Factor Signaling. Cancers (Basel) 2023; 15:460. [PMID: 36672409 PMCID: PMC9857108 DOI: 10.3390/cancers15020460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
Drug resistance is a long-standing impediment to effective systemic cancer therapy and acquired drug resistance is a growing problem for molecularly-targeted therapeutics that otherwise have shown unprecedented successes in disease control. The hepatocyte growth factor (HGF)/Met receptor pathway signaling is frequently involved in cancer and has been a subject of targeted drug development for nearly 30 years. To anticipate and study specific resistance mechanisms associated with targeting this pathway, we engineered resistance to the HGF-neutralizing antibody rilotumumab in glioblastoma cells harboring autocrine HGF/Met signaling, a frequent abnormality of this brain cancer in humans. We found that rilotumumab resistance was acquired through an unusual mechanism comprising dramatic HGF overproduction and misfolding, endoplasmic reticulum (ER) stress-response signaling and redirected vesicular trafficking that effectively sequestered rilotumumab and misfolded HGF from native HGF and activated Met. Amplification of MET and HGF genes, with evidence of rapidly acquired intron-less, reverse-transcribed copies in DNA, was also observed. These changes enabled persistent Met pathway activation and improved cell survival under stress conditions. Point mutations in the HGF pathway or other complementary or downstream growth regulatory cascades that are frequently associated with targeted drug resistance in other prevalent cancer types were not observed. Although resistant cells were significantly more malignant, they retained sensitivity to Met kinase inhibition and acquired sensitivity to inhibition of ER stress signaling and cholesterol biosynthesis. Defining this mechanism reveals details of a rapidly acquired yet highly-orchestrated multisystem route of resistance to a selective molecularly-targeted agent and suggests strategies for early detection and effective intervention.
Collapse
Affiliation(s)
- Fabiola Cecchi
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Karen Rex
- Amgen, Inc., Thousand Oaks, CA 91320, USA
| | | | - Cathy D. Vocke
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Young H. Lee
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sandra Burkett
- Molecular Cytogenetics Core Facility, Mouse Cancer Genetics Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21702, USA
| | | | | | | | | | - Donald P. Bottaro
- Urologic Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Tanriverdi O, Yildiz A. Current molecular and therapeutic advances in liposarcoma, rhabdomyosarcoma, leiomyosarcoma, synovial sarcoma, and angiosarcoma. J Oncol Pharm Pract 2022; 28:635-645. [PMID: 35043739 DOI: 10.1177/10781552211073139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Sarcomas probably develop after malignant transformation of embryonic mesenchymal cells and have broad spectrum histopathologically since they can develop from striated skeletal muscle and smooth muscle, fat and fibrous tissue, bone, cartilage and other mesenchymal tissues. The most common histological subtypes of soft tissue sarcoma in adults are: liposarcoma, leiomyosarcoma, poorly differentiated pleomorphic sarcoma, and gastrointestinal stromal tumor. Molecular and genetic studies of soft tissue sarcomas, which are considered as heterogeneous groups in terms of their molecular and clinical characteristics, are still an important area of interest The heterogeneity of the molecular and genetic alterations of these malignancies, which are mostly treated with surgery and chemotherapy, also offers hope to the researchers in terms of treatment targets. In this article, molecular biologic features of the soft tissue sarcomas including liposarcoma, rhabdomyosarcoma, leiomyosarcoma, synovial sarcoma, and angiosarcoma are discussed in the light of recent developments in molecular biology, targeted therapies and immunotherapy.
Collapse
Affiliation(s)
- Ozgur Tanriverdi
- Department of Medical Oncology, Faculty of Medicine, 175646Mugla Sitki Kocman University, Mugla, Turkey.,Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, 52986Mugla Sitki Kocman University, Mugla, Turkey
| | - Aysegul Yildiz
- Department of Molecular Biology and Genetics, Graduate School of Natural and Applied Sciences, 52986Mugla Sitki Kocman University, Mugla, Turkey.,Department of Molecular Biology and Genetics, Faculty of Science, Mugla Sitki Kocman University, Mugla, Turkey
| |
Collapse
|
3
|
O'Sullivan Coyne G, Kummar S, Hu J, Ganjoo K, Chow WA, Do KT, Zlott J, Bruns A, Rubinstein L, Foster JC, Juwara L, Meehan R, Piekarz R, Streicher H, Sharon E, Takebe N, Voth AR, Bottaro D, Costello R, Wright JJ, Doroshow JH, Chen AP. Clinical Activity of Single-Agent Cabozantinib (XL184), a Multi-receptor Tyrosine Kinase Inhibitor, in Patients with Refractory Soft-Tissue Sarcomas. Clin Cancer Res 2022; 28:279-288. [PMID: 34716194 PMCID: PMC8776602 DOI: 10.1158/1078-0432.ccr-21-2480] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 09/17/2021] [Accepted: 10/26/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Soft-tissue sarcomas (STS) are a rare, heterogeneous group of mesenchymal tumors. For decades the mainstay of treatment for advanced, unresectable STS has been palliative chemotherapy. High levels of activated MET receptor have been reported in various sarcoma cell lines, together with elevated vascular endothelial growth factor (VEGF) levels in patients with STS, suggesting that dual targeting of the VEGF and MET pathways with the multi-receptor tyrosine kinase inhibitor cabozantinib would result in clinical benefit in this population. PATIENTS AND METHODS We performed an open-label, multi-institution, single-arm phase II trial of single-agent cabozantinib in adult patients with advanced STS and progressive disease after at least 1 standard line of systemic therapy. Patients received 60 mg oral cabozantinib once daily in 28-day cycles, and dual primary endpoints of overall response rate and 6-month progression-free survival (PFS) were assessed. Changes in several circulating biomarkers were assessed as secondary endpoints. RESULTS Six (11.1%; 95% CI, 4.2%-22.6%) of the 54 evaluable patients enrolled experienced objective responses (all partial responses). Six-month PFS was 49.3% (95% CI, 36.2%-67.3%), with a median time on study of 4 cycles (range, 1-99). The most common grade 3/4 adverse events were hypertension (7.4%) and neutropenia (16.7%). Patients' levels of circulating hepatocyte growth factor (HGF), soluble MET, and VEGF-A generally increased after a cycle of therapy, while soluble VEGFR2 levels decreased, regardless of clinical outcome. CONCLUSIONS Cabozantinib single-agent antitumor activity was observed in patients with selected STS histologic subtypes (alveolar soft-part sarcoma, undifferentiated pleomorphic sarcoma, extraskeletal myxoid chondrosarcoma, and leiomyosarcoma) highlighting the biomolecular diversity of STS.
Collapse
Affiliation(s)
- Geraldine O'Sullivan Coyne
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Shivaani Kummar
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - James Hu
- University of Southern California, Los Angeles, California
| | - Kristen Ganjoo
- Stanford Cancer Center, Stanford University, Palo Alto, California
| | | | - Khanh T Do
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Jennifer Zlott
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Ashley Bruns
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Lawrence Rubinstein
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Jared C Foster
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Lamin Juwara
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Robert Meehan
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Richard Piekarz
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Howard Streicher
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Elad Sharon
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Naoko Takebe
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Andrea Regier Voth
- Applied/Developmental Research Directorate, Frederick National Laboratory for Cancer Research, Frederick, Maryland
| | - Donald Bottaro
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - Rene Costello
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
| | - John J Wright
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - James H Doroshow
- Center for Cancer Research, NCI, NIH, Bethesda, Maryland
- Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland
| | - Alice P Chen
- Early Clinical Trials Development Program, Division of Cancer Treatment and Diagnosis, NCI, NIH, Bethesda, Maryland.
| |
Collapse
|
4
|
Systems level profiling of arginine starvation reveals MYC and ERK adaptive metabolic reprogramming. Cell Death Dis 2020; 11:662. [PMID: 32814773 PMCID: PMC7438517 DOI: 10.1038/s41419-020-02899-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Abstract
Arginine auxotrophy due to the silencing of argininosuccinate synthetase 1 (ASS1) occurs in many carcinomas and in the majority of sarcomas. Arginine deiminase (ADI-PEG20) therapy exploits this metabolic vulnerability by depleting extracellular arginine, causing arginine starvation. ASS1-negative cells develop resistance to ADI-PEG20 through a metabolic adaptation that includes re-expressing ASS1. As arginine-based multiagent therapies are being developed, further characterization of the changes induced by arginine starvation is needed. In order to develop a systems-level understanding of these changes, activity-based proteomic profiling (ABPP) and phosphoproteomic profiling were performed before and after ADI-PEG20 treatment in ADI-PEG20-sensitive and resistant sarcoma cells. When integrated with metabolomic profiling, this multi-omic analysis reveals that cellular response to arginine starvation is mediated by adaptive ERK signaling and activation of the Myc–Max transcriptional network. Concomitantly, these data elucidate proteomic changes that facilitate oxaloacetate production by enhancing glutamine and pyruvate anaplerosis and altering lipid metabolism to recycle citrate for oxidative glutaminolysis. Based on the complexity of metabolic and cellular signaling interactions, these multi-omic approaches could provide valuable tools for evaluating response to metabolically targeted therapies.
Collapse
|
5
|
Kim S, Lee H, Noh J, Lee Y, Han H, Yoo DK, Kim H, Kwon S, Chung J. Efficient Selection of Antibodies Reactive to Homologous Epitopes on Human and Mouse Hepatocyte Growth Factors by Next-Generation Sequencing-Based Analysis of the B Cell Repertoire. Int J Mol Sci 2019; 20:E417. [PMID: 30669409 PMCID: PMC6359367 DOI: 10.3390/ijms20020417] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 01/16/2019] [Accepted: 01/17/2019] [Indexed: 01/05/2023] Open
Abstract
: YYB-101 is a humanized rabbit anti-human hepatocyte growth factor (HGF)-neutralizing antibody currently in clinical trial. To test the effect of HGF neutralization with antibody on anti-cancer T cell immunity, we generated surrogate antibodies that are reactive to the mouse homologue of the epitope targeted by YYB-101. First, we immunized a chicken with human HGF and monitored changes in the B cell repertoire by next-generation sequencing (NGS). We then extracted the VH gene repertoire from the NGS data, clustered it into components by sequence homology, and classified the components by the change in the number of unique VH sequences and the frequencies of the VH sequences within each component following immunization. Those changes should accompany the preferential proliferation and somatic hypermutation or gene conversion of B cells encoding HGF-reactive antibodies. One component showed significant increases in the number and frequencies of unique VH sequences and harbored genes encoding antibodies that were reactive to human HGF and competitive with YYB-101 for HGF binding. Some of the antibodies also reacted to mouse HGF. The selected VH sequences shared 98.3% identity and 98.9% amino acid similarity. It is therefore likely that the antibodies encoded by them all react to the epitope targeted by YYB-101.
Collapse
Affiliation(s)
- Soohyun Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 00380, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 00380, Korea.
| | - Hyunho Lee
- Department of Electrical Engineering and Computer Science, Seoul National University, Seoul 08826, Korea.
| | - Jinsung Noh
- Department of Electrical Engineering and Computer Science, Seoul National University, Seoul 08826, Korea.
| | - Yonghee Lee
- Department of Electrical Engineering and Computer Science, Seoul National University, Seoul 08826, Korea.
| | - Haejun Han
- Celemics, Inc., 131 Gasandigital 1-ro, Geumcheon-gu, Seoul 08506, Korea.
| | - Duck Kyun Yoo
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 00380, Korea.
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 00380, Korea.
- Genomic Medicine Institute (GMI), Medical Research Center, Seoul National University, Seoul 00380, Korea.
| | - Hyori Kim
- Convergence medicine research center, Asan Institute for Life Sciences, Asan Medical Center, Seoul 05505, Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul 05505, Korea.
| | - Sunghoon Kwon
- Department of Electrical Engineering and Computer Science, Seoul National University, Seoul 08826, Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Korea.
- Institutes of Entrepreneurial BioConvergence, Seoul National University, Seoul 08826, Korea.
- Seoul National University Hospital Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea.
- Inter-University Semiconductor Research Center, Seoul National University, Seoul 08826, Korea.
| | - Junho Chung
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 00380, Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 00380, Korea.
- Department of Biomedical Science, Seoul National University College of Medicine, Seoul 00380, Korea.
| |
Collapse
|
6
|
Kou J, Musich PR, Staal B, Kang L, Qin Y, Yao ZQ, Zhang B, Wu W, Tam A, Huang A, Hao HX, Vande Woude GF, Xie Q. Differential responses of MET activations to MET kinase inhibitor and neutralizing antibody. J Transl Med 2018; 16:253. [PMID: 30208970 PMCID: PMC6134500 DOI: 10.1186/s12967-018-1628-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Accepted: 09/05/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Aberrant MET tyrosine kinase signaling is known to cause cancer initiation and progression. While MET inhibitors are in clinical trials against several cancer types, the clinical efficacies are controversial and the molecular mechanisms toward sensitivity remain elusive. METHODS With the goal to investigate the molecular basis of MET amplification (METamp) and hepatocyte growth factor (HGF) autocrine-driven tumors in response to MET tyrosine kinase inhibitors (TKI) and neutralizing antibodies, we compared cancer cells harboring METamp (MKN45 and MHCCH97H) or HGF-autocrine (JHH5 and U87) for their sensitivity and downstream biological responses to a MET-TKI (INC280) and an anti-MET monoclonal antibody (MetMab) in vitro, and for tumor inhibition in vivo. RESULTS We find that cancer cells driven by METamp are more sensitive to INC280 than are those driven by HGF-autocrine activation. In METamp cells, INC280 induced a DNA damage response with activation of repair through the p53BP1/ATM signaling pathway. Although MetMab failed to inhibit METamp cell proliferation and tumor growth, both INC280 and MetMab reduced HGF-autocrine tumor growth. In addition, we also show that HGF stimulation promoted human HUVEC cell tube formation via the Src pathway, which was inhibited by either INC280 or MetMab. These observations suggest that in HGF-autocrine tumors, the endothelial cells are the secondary targets MET inhibitors. CONCLUSIONS Our results demonstrate that METamp and HGF-autocrine activation favor different molecular mechanisms. While combining MET TKIs and ATM inhibitors may enhance the efficacy for treating tumors harboring METamp, a combined inhibition of MET and angiogenesis pathways may improve the therapeutic efficacy against HGF-autocrine tumors.
Collapse
Affiliation(s)
- Jianqun Kou
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Phillip R Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Ben Staal
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Liang Kang
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Yuan Qin
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Zhi Q Yao
- Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.,Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA
| | - Boheng Zhang
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Weizhong Wu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Angela Tam
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Alan Huang
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - Huai-Xiang Hao
- Novartis Institutes for BioMedical Research, Cambridge, MA, 02139, USA
| | - George F Vande Woude
- Center of Cell and Cancer Biology, Van Andel Research Institute, Grand Rapids, MI, 49503, USA
| | - Qian Xie
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA. .,Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37614, USA.
| |
Collapse
|
7
|
Hoang NT, Acevedo LA, Mann MJ, Tolani B. A review of soft-tissue sarcomas: translation of biological advances into treatment measures. Cancer Manag Res 2018; 10:1089-1114. [PMID: 29785138 PMCID: PMC5955018 DOI: 10.2147/cmar.s159641] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Soft-tissue sarcomas are rare malignant tumors arising from connective tissues and have an overall incidence of about five per 100,000 per year. While this diverse family of malignancies comprises over 100 histological subtypes and many molecular aberrations are prevalent within specific sarcomas, very few are therapeutically targeted. Instead of utilizing molecular signatures, first-line sarcoma treatment options are still limited to traditional surgery and chemotherapy, and many of the latter remain largely ineffective and are plagued by disease resistance. Currently, the mechanism of sarcoma oncogenesis remains largely unknown, thus necessitating a better understanding of pathogenesis. Although substantial progress has not occurred with molecularly targeted therapies over the past 30 years, increased knowledge about sarcoma biology could lead to new and more effective treatment strategies to move the field forward. Here, we discuss biological advances in the core molecular determinants in some of the most common soft-tissue sarcomas - liposarcoma, angiosarcoma, leiomyosarcoma, rhabdomyosarcoma, Ewing's sarcoma, and synovial sarcoma - with an emphasis on emerging genomic and molecular pathway targets and immunotherapeutic treatment strategies to combat this confounding disease.
Collapse
Affiliation(s)
- Ngoc T Hoang
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Luis A Acevedo
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Michael J Mann
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| | - Bhairavi Tolani
- Thoracic Oncology Program, Department of Surgery, Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA, USA
| |
Collapse
|
8
|
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]
|
9
|
Bahrami A, Shahidsales S, Khazaei M, Ghayour-Mobarhan M, Maftouh M, Hassanian SM, Avan A. C-Met as a potential target for the treatment of gastrointestinal cancer: Current status and future perspectives. J Cell Physiol 2017; 232:2657-2673. [PMID: 28075018 DOI: 10.1002/jcp.25794] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2017] [Accepted: 01/10/2017] [Indexed: 01/05/2025]
Abstract
Aberrant activation of the HGF/c-Met signalling pathways is shown to be related with cell proliferation, progression, metastasis, and worse prognosis in several tumor types, including gastrointestinal cancers, suggesting its value as a stimulating-target for cancer-therapy. Several approaches have been developed for targeting HGF and/or c-Met, and one of them, crizotinib (dual c-Met/ALK inhibitor), is recently been approved by FDA for lung-cancers with ALK-rearrangement. The main aim of current review is to give an overview on the role of c-Met/HGF pathway in gastrointestinal cancer, in preclinical and clinical trials. Although several important matters is still remained to be elucidated on the molecular pathways underlying the antitumor effects of this therapy in gastrointestinal-cancers. Further investigations are warranted to recognize the main determinants of the activity of c-Met inhibitors, for parallel targeting signalling pathway associated/activated via MET/HGF pathway or in response to the cell resistance to anti-c-Met agents. Additionally, identification of patients that might benefit from therapy could help to increase the selectivity and efficacy of the therapy.
Collapse
Affiliation(s)
- Afsane Bahrami
- Molecular Medicine Group, Department of Modern Sciences and Technology, Mashhad University of Medical Sciences, Mashhad, Iran
- Student Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh Shahidsales
- Cancer Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Neurogenic Inflammatory Research Center and Department of Physiology, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic syndrome Research Center, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
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: 277] [Impact Index Per Article: 34.6] [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
|
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
|
12
|
Mao L, Dauchy RT, Blask DE, Dauchy EM, Slakey LM, Brimer S, Yuan L, Xiang S, Hauch A, Smith K, Frasch T, Belancio VP, Wren MA, Hill SM. Melatonin suppression of aerobic glycolysis (Warburg effect), survival signalling and metastasis in human leiomyosarcoma. J Pineal Res 2016; 60:167-77. [PMID: 26607298 DOI: 10.1111/jpi.12298] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/19/2015] [Indexed: 12/25/2022]
Abstract
Leiomyosarcoma (LMS) represents a highly malignant, rare soft tissue sarcoma with high rates of morbidity and mortality. Previously, we demonstrated that tissue-isolated human LMS xenografts perfused in situ are highly sensitive to the direct anticancer effects of physiological nocturnal blood levels of melatonin which inhibited tumour cell proliferative activity, linoleic acid (LA) uptake and metabolism to 13-hydroxyoctadecadienoic acid (13-HODE). Here, we show the effects of low pharmacological blood concentrations of melatonin following oral ingestion of a melatonin supplement by healthy adult human female subjects on tumour proliferative activity, aerobic glycolysis (Warburg effect) and LA metabolic signalling in tissue-isolated LMS xenografts perfused in situ with this blood. Melatonin markedly suppressed aerobic glycolysis and induced a complete inhibition of tumour LA uptake, 13-HODE release, as well as significant reductions in tumour cAMP levels, DNA content and [(3) H]-thymidine incorporation into DNA. Furthermore, melatonin completely suppressed the phospho-activation of ERK 1/2, AKT, GSK3β and NF-kB (p65). The addition of S20928, a nonselective melatonin antagonist, reversed these melatonin inhibitory effects. Moreover, in in vitro cell culture studies, physiological concentrations of melatonin repressed cell proliferation and cell invasion. These results demonstrate that nocturnal melatonin directly inhibited tumour growth and invasion of human LMS via suppression of the Warburg effect, LA uptake and other related signalling mechanisms. An understanding of these novel signalling pathway(s) and their association with aerobic glycolysis and LA metabolism in human LMS may lead to new circadian-based therapies for the prevention and treatment of LMS and potentially other mesenchymally derived solid tumours.
Collapse
Affiliation(s)
- Lulu Mao
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Robert T Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - David E Blask
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Erin M Dauchy
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lauren M Slakey
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Samantha Brimer
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lin Yuan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Shulin Xiang
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Adam Hauch
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Kara Smith
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Tripp Frasch
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Victoria P Belancio
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Melissa A Wren
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Steven M Hill
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, LA, USA
- Tulane Cancer Center and Louisiana Cancer Research Consortium, New Orleans, LA, USA
- Tulane Center for Circadian Biology, Tulane University School of Medicine, New Orleans, LA, USA
| |
Collapse
|
13
|
Han Z, Harris PKW, Karmakar P, Kim T, Owusu BY, Wildman SA, Klampfer L, Janetka JW. α-Ketobenzothiazole Serine Protease Inhibitors of Aberrant HGF/c-MET and MSP/RON Kinase Pathway Signaling in Cancer. ChemMedChem 2016; 11:585-99. [PMID: 26889658 DOI: 10.1002/cmdc.201500600] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Indexed: 12/20/2022]
Abstract
Upregulation of the HGF and MSP growth-factor processing serine endopeptidases HGFA, matriptase and hepsin is correlated with increased metastasis in multiple tumor types driven by c-MET or RON kinase signaling. We rationally designed P1' α-ketobenzothiazole mechanism-based inhibitors of these proteases. Structure-activity studies are presented, which resulted in the identification of potent inhibitors with differential selectivity. The tetrapeptide inhibitors span the P1-P1' substrate cleavage site via a P1' amide linker off the benzothiazole, occupying the S3' pocket. Optimized inhibitors display sub-nanomolar enzyme inhibition against one, two, or all three of HGFA, matriptase, and hepsin. Several compounds also have good selectivity against the related trypsin-like proteases, thrombin and Factor Xa. Finally, we show that inhibitors block the fibroblast (HGF)-mediated migration of invasive DU145 prostate cancer cells. In addition to prostate cancer, breast, colon, lung, pancreas, gliomas, and multiple myeloma tumors all depend on HGF and MSP for tumor survival and progression. Therefore, these unique inhibitors have potential as new therapeutics for a diverse set of tumor types.
Collapse
Affiliation(s)
- Zhenfu Han
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Peter K W Harris
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Partha Karmakar
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Tommy Kim
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA
| | - Ben Y Owusu
- Department of Oncology, Southern Research Institute, 2000 9th Ave., Birmingham, AL, 35205, USA
| | - Scott A Wildman
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA.,Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
| | - Lidija Klampfer
- Department of Oncology, Southern Research Institute, 2000 9th Ave., Birmingham, AL, 35205, USA
| | - James W Janetka
- Department of Biochemistry and Molecular Biophysics, Alvin J. Siteman Cancer Center, Washington University School of Medicine, 660 S. Euclid Ave., Saint Louis, MO, 63110, USA.
| |
Collapse
|
14
|
Lee J, Tran P, Klempner SJ. Targeting the MET Pathway in Gastric and Oesophageal Cancers: Refining the Optimal Approach. Clin Oncol (R Coll Radiol) 2016; 28:e35-44. [PMID: 26880063 DOI: 10.1016/j.clon.2016.01.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 11/09/2015] [Accepted: 12/10/2015] [Indexed: 12/21/2022]
Abstract
Gastric and oesophageal cancers are a major cause of global cancer-related morbidity and mortality. Improvements in treatment for locoregional and metastatic gastric and oesophageal cancer have been incremental and the overall prognosis remains poor. Increasingly, molecular classification has identified recurrent, therapeutically relevant, somatic alterations in gastroesophageal malignancies. However, other than ERBB2 amplification, molecularly directed therapies have not translated to improved survival. Amplification of the receptor tyrosine kinase MET is found in about 5% of gastroesophageal cancers and represents an oncogenic driver and therapeutic target. Small series have shown activity of MET-directed tyrosine kinase inhibitors, but the clinical benefit of anti-MET antibodies has been disappointing. Here we discuss the MET pathway in gastroesophageal cancers, the clinical data for MET small molecule tyrosine kinase inhibitors, anti-MET antibodies and future clinical directions for targeting MET in gastric and oesophageal cancers. To our knowledge, this is the most comprehensive review of the clinical experience with MET-directed therapies in gastric and oesophageal cancers.
Collapse
Affiliation(s)
- J Lee
- Department of Medicine, University of California Irvine, Orange, CA, USA
| | - P Tran
- Division of Hematology-Oncology, University of California Irvine, Orange, CA, USA
| | - S J Klempner
- Division of Hematology-Oncology, University of California Irvine, Orange, CA, USA.
| |
Collapse
|
15
|
Marano L, Chiari R, Fabozzi A, De Vita F, Boccardi V, Roviello G, Petrioli R, Marrelli D, Roviello F, Patriti A. c-Met targeting in advanced gastric cancer: An open challenge. Cancer Lett 2015; 365:30-36. [PMID: 26049023 DOI: 10.1016/j.canlet.2015.05.028] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 12/14/2022]
Abstract
Despite significant improvements in systemic chemotherapy over the last two decades, the prognosis of patients with advanced gastric and gastroesophageal junction adenocarcinoma (GC) remains poor. Because of molecular heterogeneity, it is essential to classify tumors based on the underlying oncogenic pathways and to develop targeted therapies acting on individual tumors. High-quality research and advances in technology have contributed to the elucidation of molecular pathways underlying disease progression and have stimulated many clinical studies testing target therapies in an advanced disease setting. In particular, strong preclinical evidence for the aberrant activation of the HGF/c-Met signaling pathways in GC cancers exists. This review will cover the c-Met pathway, the mechanisms of c-Met activation and the different strategies of its inhibition. Next, we will focus on the current state of the art in the clinical evaluation of c-Met-targeted therapies and the description of ongoing randomized trials with the idea that in this disease, high quality translational research to identify and validate biomarkers is a priority task.
Collapse
Affiliation(s)
- Luigi Marano
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049 Spoleto, Italy.
| | - Rita Chiari
- Department of Medical Oncology, "Santa Maria della Misericordia" Hospital, Azienda Ospedaliera di Perugia, 06132 Perugia, Italy
| | - Alessio Fabozzi
- Division of Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi-A. Lanzara", Second University of Naples, 80131 Naples, Italy
| | - Ferdinando De Vita
- Division of Medical Oncology, Department of Clinical and Experimental Medicine "F. Magrassi-A. Lanzara", Second University of Naples, 80131 Naples, Italy
| | - Virginia Boccardi
- Section of Gerontology and Geriatrics, Department of Medicine, University of Perugia, 06132 Perugia, Italy
| | | | | | - Daniele Marrelli
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Franco Roviello
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy
| | - Alberto Patriti
- General, Minimally Invasive and Robotic Surgery, Department of Surgery, "San Matteo degli Infermi" Hospital, ASL Umbria 2, 06049 Spoleto, Italy
| |
Collapse
|
16
|
Zhang Y, Doshi S, Zhu M. Pharmacokinetics and pharmacodynamics of rilotumumab: a decade of experience in preclinical and clinical cancer research. Br J Clin Pharmacol 2015; 80:957-64. [PMID: 25912961 DOI: 10.1111/bcp.12663] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 04/06/2015] [Accepted: 04/20/2015] [Indexed: 12/11/2022] Open
Abstract
Rilotumumab is a fully human monoclonal antibody against hepatocyte growth factor, the only known ligand of the MET receptor. Over the last decade, rilotumumab has been extensively tested in preclinical studies and in clinical studies in a variety of cancer types. In this review, we examine the pharmacokinetic and pharmacodynamic data that have been collected in the rilotumumab programme to date, and discuss retrospectively how the knowledge acquired in this programme can be applied to a number of key issues in oncology drug development, including: (i) using preclinical data to inform first-in-human study design; (ii) the role of biomarkers in the identification of a target patient population; (iii) the potential for drug interactions between therapeutic proteins and other anticancer agents; and (iv) pharmacokinetic and pharmacodynamic considerations in phase 3 study design.
Collapse
Affiliation(s)
- Y Zhang
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - S Doshi
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| | - M Zhu
- Department of Clinical Pharmacology, Modeling, and Simulation, Amgen Inc, One Amgen Center Drive, Thousand Oaks, CA, 91320, USA
| |
Collapse
|
17
|
Schmitz K, Koeppen H, Binot E, Fassunke J, Künstlinger H, Ihle MA, Heydt C, Wardelmann E, Büttner R, Merkelbach-Bruse S, Rüschoff J, Schildhaus HU. MET gene copy number alterations and expression of MET and hepatocyte growth factor are potential biomarkers in angiosarcomas and undifferentiated pleomorphic sarcomas. PLoS One 2015; 10:e0120079. [PMID: 25844809 PMCID: PMC4386816 DOI: 10.1371/journal.pone.0120079] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 01/19/2015] [Indexed: 11/19/2022] Open
Abstract
Soft tissue sarcomas are a heterogeneous group of tumors with many different subtypes. In 2014 an estimated 12,020 newly diagnosed cases and 4,740 soft tissue sarcoma related deaths can be expected in the United States. Many soft tissue sarcomas are associated with poor prognosis and therapeutic options are often limited. The evolution of precision medicine has not yet fully reached the clinical treatment of sarcomas since therapeutically tractable genetic changes have not been comprehensively studied so far. We analyzed a total of 484 adult-type malignant mesenchymal tumors by MET fluorescence in situ hybridization and MET and hepatocyte growth factor immunohistochemistry. Eleven different entities were included, among them the most common and clinically relevant subtypes and tumors with specific translocations or complex genetic changes. MET protein expression was observed in 2.6% of the cases, all of which were either undifferentiated pleomorphic sarcomas or angiosarcomas, showing positivity rates of 14% and 17%, respectively. 6% of the tumors showed hepatocyte growth factor overexpression, mainly seen in undifferentiated pleomorphic sarcomas and angiosarcomas, but also in clear cell sarcomas, malignant peripheral nerve sheath tumors, leiomyosarcomas and gastrointestinal stromal tumors. MET and hepatocyte growth factor overexpression were significantly correlated and may suggest an autocrine activation in these tumors. MET FISH amplification and copy number gain were present in 4% of the tumors (15/413). Two samples, both undifferentiated pleomorphic sarcomas, fulfilled the criteria for high level amplification of MET, one undifferentiated pleomorphic sarcoma reached an intermediate level copy number gain, and 12 samples of different subtypes were categorized as low level copy number gains for MET. Our findings indicate that angiosarcomas and undifferentiated pleomorphic sarcomas rather than other frequent adult-type sarcomas should be enrolled in screening programs for clinical trials with MET inhibitors. The screening methods should include both in situ hybridization and immunohistochemistry.
Collapse
Affiliation(s)
- Katja Schmitz
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Institute of Pathology, University Hospital Göttingen, Göttingen, Germany
| | - Hartmut Koeppen
- Genentech Inc., South San Francisco, California, United States of America
| | - Elke Binot
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jana Fassunke
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Helen Künstlinger
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Michaela A. Ihle
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Carina Heydt
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Eva Wardelmann
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Gerhard Domagk Institute of Pathology, University Hospital Münster, Münster, Germany
| | - Reinhard Büttner
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | | | | | - Hans-Ulrich Schildhaus
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
- Institute of Pathology, University Hospital Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
18
|
Zhang Y, Jain RK, Zhu M. Recent Progress and Advances in HGF/MET-Targeted Therapeutic Agents for Cancer Treatment. Biomedicines 2015; 3:149-181. [PMID: 28536405 PMCID: PMC5344234 DOI: 10.3390/biomedicines3010149] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 02/25/2015] [Accepted: 03/03/2015] [Indexed: 12/31/2022] Open
Abstract
The hepatocyte growth factor (HGF): MET axis is a ligand-mediated receptor tyrosine kinase pathway that is involved in multiple cellular functions, including proliferation, survival, motility, and morphogenesis. Aberrancy in the HGF/MET pathway has been reported in multiple tumor types and is associated with tumor stage and prognosis. Thus, targeting the HGF/MET pathway has become a potential therapeutic strategy in oncology development in the last two decades. A number of novel therapeutic agents-either as therapeutic proteins or small molecules that target the HGF/MET pathway-have been tested in patients with different tumor types in clinical studies. In this review, recent progress in HGF/MET pathway-targeted therapy for cancer treatment, the therapeutic potential of HGF/MET-targeted agents, and challenges in the development of such agents will be discussed.
Collapse
Affiliation(s)
- Yilong Zhang
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| | - Rajul K Jain
- Kite Pharma, Inc., 2225 Colorado Avenue, Santa Monica, CA 90404, USA.
| | - Min Zhu
- Department of Clinical Pharmacology, Modeling and Simulation, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320, USA.
| |
Collapse
|
19
|
Exposure-response analysis of rilotumumab in gastric cancer: the role of tumour MET expression. Br J Cancer 2015; 112:429-37. [PMID: 25584489 PMCID: PMC4453660 DOI: 10.1038/bjc.2014.649] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 11/17/2014] [Accepted: 12/08/2014] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Rilotumumab, an investigational, monoclonal antibody, inhibits MET-mediated signalling. In a randomized phase 2 trial of rilotumumab±epirubicin/cisplatin/capecitabine in gastric or oesophagogastric junction cancer, patients receiving rilotumumab showed a trend towards improved survival, especially in MET-positive patients, but no clear dose-response relationship was observed. Exposure-response and biomarker analyses were used for dose selection and to differentiate patient subpopulations that may benefit most from treatment. Here, we analyse rilotumumab exposure-survival and exposure-safety and the impact of MET expression on these relationships. METHODS Individual rilotumumab exposure parameters were generated using population pharmacokinetic modelling. Relationships among rilotumumab dose (7.5 and 15 mg kg(-1)), exposure, and clinical outcomes (progression-free survival (PFS) and overall survival (OS)) were evaluated with Cox regression models and Kaplan-Meier plots. MET status and other baseline covariates were evaluated in subgroup and multivariate analyses. Treatment-emergent adverse events were summarised by exposure. RESULTS Among MET-positive patients, higher rilotumumab exposure, vs placebo and low exposure, was associated with improved median PFS (80% CI: 7.0 (5.7-9.7) vs 4.4 (2.9-4.9) and 5.5 (4.2-6.8) months) and OS (13.4 (10.6-18.6) vs 5.7 (4.7-10.2) and 8.1 (6.9-11.1) months) without increased toxicity. No rilotumumab benefit was seen among MET-negative patients. CONCLUSIONS Rilotumumab had an exposure-dependent treatment effect in patients with MET-positive gastric or oesophagogastric junction cancer.
Collapse
|
20
|
Vita FD, Martino ND, Fabozzi A, Laterza MM, Ventriglia J, Savastano B, Petrillo A, Gambardella V, Sforza V, Marano L, Auricchio A, Galizia G, Ciardiello F, Orditura M. Clinical management of advanced gastric cancer: the role of new molecular drugs. World J Gastroenterol 2014; 20:14537-14558. [PMID: 25356019 PMCID: PMC4209522 DOI: 10.3748/wjg.v20.i40.14537] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 02/19/2014] [Accepted: 06/02/2014] [Indexed: 02/06/2023] Open
Abstract
Gastric cancer is the fourth most common malignant neoplasm and the second leading cause of death for cancer in Western countries with more than 20000 new cases yearly diagnosed in the United States. Surgery represents the main approach for this disease but, notwithstanding the advances in surgical techniques, we observed a minimal improvement in terms of overall survival with a significant increasing of relapsing disease rates. Despite the development of new drugs has significantly improved the effectiveness of chemotherapy, the prognosis of patients with unresectable or metastatic gastric adenocarcinoma remains poor. Recently, several molecular target agents have been investigated; in particular, trastuzumab represents the first target molecule showing improvements in overall survival in human epithelial growth factor 2-positive gastric cancer patients. New molecules targeting vascular epithelial growth factor, mammalian target of rapamycin, and anti hepatocyte growth factor-c-Met pathway are also under investigation, with interesting results. Anyway, it seems necessary to select more accurately the population to treat with new agents by the identification of new biomarkers in order to optimize the results. In this paper we review the actual "scenario" of targeted treatments, also focusing on the new agents in development for gastric cancer and gastro-esophageal carcinoma, discussing their efficacy and potential applications in clinical practice.
Collapse
|
21
|
Keire PA, Bressler SL, Lemire JM, Edris B, Rubin BP, Rahmani M, McManus BM, van de Rijn M, Wight TN. A role for versican in the development of leiomyosarcoma. J Biol Chem 2014; 289:34089-103. [PMID: 25320080 DOI: 10.1074/jbc.m114.607168] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Leiomyosarcoma (LMS) is a mesenchymal cancer that occurs throughout the body. Although LMS is easily recognized histopathologically, the cause of the disease remains unknown. Versican, an extracellular matrix proteoglycan, increases in LMS. Microarray analyses of 80 LMSs and 24 leiomyomas showed a significant elevated expression of versican in human LMS versus benign leiomyomas. To explore the importance of versican in this smooth muscle cell tumor, we used versican-directed siRNA to knock down versican expression in a LMS human cell line, SK-LMS-1. Decreased versican expression was accompanied by slower rates of LMS cell proliferation and migration, increased adhesion, and decreased accumulation of the extracellular matrix macromolecule hyaluronan. Addition of purified versican to cells expressing versican siRNA restored cell proliferation to the level of LMS controls, increased the pericellular coat and the retention of hyaluronan, and decreased cell adhesion in a dose-dependent manner. The presence of versican was not only synergistic with hyaluronan in increasing cell proliferation, but the depletion of versican decreased hyaluronan synthase expression and decreased the retention of hyaluronan. When LMS cells stably expressing versican siRNA were injected into nude mice, the resulting tumors displayed significantly less versican and hyaluronan staining, had lower volumes, and had reduced levels of mitosis as compared with controls. Collectively, these results suggest a role for using versican as a point of control in the management and treatment of LMS.
Collapse
Affiliation(s)
- Paul A Keire
- From the Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101, Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Steven L Bressler
- From the Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101
| | - Joan M Lemire
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Badreddin Edris
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, and
| | - Brian P Rubin
- Department of Pathology, University of Washington, Seattle, Washington 98195
| | - Maziar Rahmani
- James Hogg iCAPTURE Centre for Cardiovascular and Pulmonary Research, St. Paul's Hospital, Room 166, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada, and Department of Pathology and Laboratory Medicine, University of British Columbia, Room G227, 2211 Wesbrook Mall, Vancouver, British Columbia V6T 2A1, Canada
| | - Bruce M McManus
- James Hogg iCAPTURE Centre for Cardiovascular and Pulmonary Research, St. Paul's Hospital, Room 166, 1081 Burrard Street, Vancouver, British Columbia V6Z 1Y6, Canada, and Department of Pathology and Laboratory Medicine, University of British Columbia, Room G227, 2211 Wesbrook Mall, Vancouver, British Columbia V6T 2A1, Canada
| | - Matt van de Rijn
- Department of Pathology, Stanford University School of Medicine, Stanford, California 94305, and
| | - Thomas N Wight
- From the Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, Washington 98101, Department of Pathology, University of Washington, Seattle, Washington 98195,
| |
Collapse
|
22
|
Iveson T, Donehower RC, Davidenko I, Tjulandin S, Deptala A, Harrison M, Nirni S, Lakshmaiah K, Thomas A, Jiang Y, Zhu M, Tang R, Anderson A, Dubey S, Oliner KS, Loh E. Rilotumumab in combination with epirubicin, cisplatin, and capecitabine as first-line treatment for gastric or oesophagogastric junction adenocarcinoma: an open-label, dose de-escalation phase 1b study and a double-blind, randomised phase 2 study. Lancet Oncol 2014; 15:1007-18. [PMID: 24965569 DOI: 10.1016/s1470-2045(14)70023-3] [Citation(s) in RCA: 233] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Dysregulation of the hepatocyte growth factor (HGF)/MET pathway promotes tumour growth and metastasis. Rilotumumab is a fully human, monoclonal antibody that neutralises HGF. We aimed to assess the safety, efficacy, biomarkers, and pharmacokinetics of rilotumumab combined with epirubicin, cisplatin, and capecitabine (ECX) in patients with advanced gastric or oesophagogastric junction cancer. METHODS We recruited patients (≥18 years old) with unresectable locally advanced or metastatic gastric or oesophagogastric junction adenocarcinoma, an Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1, who had not received previous systemic therapy, from 43 sites worldwide. Phase 1b was an open-label, dose de-escalation study to identify a safe dose of rilotumumab (initial dose 15 mg/kg intravenously on day 1) plus ECX (epirubicin 50 mg/m(2) intravenously on day 1, cisplatin 60 mg/m(2) intravenously on day 1, capecitabine 625 mg/m(2) twice a day orally on days 1-21, respectively), administered every 3 weeks. The phase 1b primary endpoint was the incidence of dose-limiting toxicities in all phase 1b patients who received at least one dose of rilotumumab and completed the dose-limiting toxicity assessment window (first cycle of therapy). Phase 2 was a double-blind study that randomly assigned patients (1:1:1) using an interactive voice response system to receive rilotumumab 15 mg/kg, rilotumumab 7·5 mg/kg, or placebo, plus ECX (doses as above), stratified by ECOG performance status and disease extent. The phase 2 primary endpoint was progression-free survival (PFS), analysed by intention to treat. The study is registered with ClinicalTrials.gov, number NCT00719550. FINDINGS Seven of the nine patients enrolled in the phase 1b study received at least one dose of rilotumumab 15 mg/kg, only two of whom had three dose-limiting toxicities: palmar-plantar erythrodysesthesia, cerebral ischaemia, and deep-vein thrombosis. In phase 2, 121 patients were randomly assigned (40 to rilotumumab 15 mg/kg; 42 to rilotumumab 7·5 mg/kg; 39 to placebo). Median PFS was 5·1 months (95% CI 2·9-7·0) in the rilotumumab 15 mg/kg group, 6·8 months (4·5-7·5) in the rilotumumab 7·5 mg/kg group, 5·7 months (4·5-7·0) in both rilotumumab groups combined, and 4·2 months (2·9-4·9) in the placebo group. The hazard ratio for PFS events compared with placebo was 0·69 (80% CI 0·49-0·97; p=0·164) for rilotumumab 15 mg/kg, 0·53 (80% CI 0·38-0·73; p=0·009) for rilotumumab 7·5 mg/kg, and 0·60 (80% CI 0·45-0·79; p=0·016) for combined rilotumumab. Any grade adverse events more common in the combined rilotumumab group than in the placebo group included haematological adverse events (neutropenia in 44 [54%] of 81 patients vs 13 [33%] of 39 patients; anaemia in 32 [40%] vs 11 [28%]; and thrombocytopenia in nine [11%] vs none), peripheral oedema (22 [27%] vs three [8%]), and venous thromboembolism (16 [20%] vs five [13%]). Grade 3-4 adverse events more common with rilotumumab included neutropenia (36 [44%] vs 11 [28%]) and venous thromboembolism (16 [20%] vs four [10%]). Serious adverse events were balanced between groups except for anaemia, which occurred more frequently in the combined rilotumumab group (ten [12%] vs none). INTERPRETATION Rilotumumab plus ECX had no unexpected safety signals and showed greater activity than placebo plus ECX. A phase 3 study of the combination in MET-positive gastric and oesophagogastric junction cancer is in progress. FUNDING Amgen Inc.
Collapse
Affiliation(s)
| | | | - Irina Davidenko
- State Institution of Public Health "Regional Clinical Oncology Dispensary", Krasnodar, Russia
| | | | - Andrzej Deptala
- Central Clinical Hospital MSW, Warsaw Medical University, Warsaw, Poland
| | | | - Somanath Nirni
- Indo-American Cancer Institute and Research Centre, Hyderabad, India
| | | | | | | | - Min Zhu
- Amgen Inc, Thousand Oaks, CA, USA
| | - Rui Tang
- Amgen Inc, Thousand Oaks, CA, USA
| | | | | | | | - Elwyn Loh
- Amgen Inc, South San Francisco, CA, USA
| |
Collapse
|
23
|
Understanding the impact of 2D and 3D fibroblast cultures on in vitro breast cancer models. PLoS One 2013; 8:e76373. [PMID: 24124550 PMCID: PMC3790689 DOI: 10.1371/journal.pone.0076373] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2013] [Accepted: 08/26/2013] [Indexed: 12/21/2022] Open
Abstract
The utilization of 3D, physiologically relevant in vitro cancer models to investigate complex interactions between tumor and stroma has been increasing. Prior work has generally focused on the cancer cells and, the role of fibroblast culture conditions on tumor-stromal cell interactions is still largely unknown. Here, we focus on the stroma by comparing functional behaviors of human mammary fibroblasts (HMFs) cultured in 2D and 3D and their effects on the invasive progression of breast cancer cells (MCF10DCIS.com). We identified increased levels of several paracrine factors from HMFs cultured in 3D conditions that drive the invasive transition. Using a microscale co-culture model with improved compartmentalization and sensitivity, we demonstrated that HMFs cultured in 3D intensify the promotion of the invasive progression through the HGF/c-Met interaction. This study highlights the importance of the 3D stromal microenvironment in the development of multiple cell type in vitro cancer models.
Collapse
|
24
|
Rex K, Lewis XZ, Gobalakrishnan S, Glaus C, Silva MD, Radinsky R, Burgess TL, Gambhir SS, Coxon A. Evaluation of the antitumor effects of rilotumumab by PET imaging in a U-87 MG mouse xenograft model. Nucl Med Biol 2013; 40:458-63. [DOI: 10.1016/j.nucmedbio.2013.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2012] [Accepted: 01/15/2013] [Indexed: 01/03/2023]
|
25
|
Lusby K, Savannah KB, Demicco EG, Zhang Y, Ghadimi MP, Young ED, Colombo C, Lam R, Dogan TE, Hornick JL, Lazar AJ, Hunt KK, Anderson ML, Creighton CJ, Lev D, Pollock RE. Uterine leiomyosarcoma management, outcome, and associated molecular biomarkers: a single institution's experience. Ann Surg Oncol 2013; 20:2364-72. [PMID: 23334251 DOI: 10.1245/s10434-012-2834-0] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Indexed: 12/18/2022]
Abstract
BACKGROUND Uterine leiomyosarcoma (ULMS) is an aggressive, rapidly progressive tumor lacking clinical and molecular predictors of outcome. METHODS ULMS patients (n = 349) were classified by disease status at presentation to MDACC as having intra-abdominal (n = 157) or distant metastatic disease (n = 192). Patient, tumor, treatment, and outcome variables were retrospectively retrieved. Formalin-fixed, paraffin-embedded tumor and control tissues from these patients (n = 109) were assembled in a tissue microarray and evaluated for hormone receptors and markers of angiogenesis, cell-cycle progression and survival. Patient, tumor, and treatment variables were correlatively analyzed. RESULTS The 5- and 10-year disease-specific survival (DSS) for the cohort was 42 and 27 %, respectively. Patients with primary intra-abdominal tumors had better outcomes than those with recurrent intraperitoneal tumors. Whites had a more favorable prognosis. In patients with intra-abdominal tumors, only mitotic count >10M/10HPF portended poorer prognosis. Patients with pulmonary metastasis had improved outcomes with "curative" metastasectomy. ULMS samples exhibited loss of ER and PR expression, overexpressed Ki-67, and altered p53, Rb, p16, cytoplasmic β-catenin, EGFR, PDGFR-α, PDGFR-β, and AXL levels. Metastatic tumors had increased VEGF, Ki-67, and survivin expression versus localized disease. Survivin and β-catenin expression were associated with intraperitoneal recurrence; high bcl-2 expression predicted longer DSS. CONCLUSIONS Analysis of both clinicopathologic factors and immunohistochemical biomarkers in ULMS identified several prognostic clinical and molecular factors, suggesting that further study may lead to improved ULMS understanding and treatment.
Collapse
Affiliation(s)
- Kristelle Lusby
- Department of Surgical Oncology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
van der Graaf WTA, Gelderblom H. New systemic therapy options for advanced sarcomas. Curr Treat Options Oncol 2012; 13:306-17. [PMID: 22661203 PMCID: PMC3419831 DOI: 10.1007/s11864-012-0196-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The outcome of patients with advanced soft tissue sarcomas (STS) has not improved much during the last decade. Apart from non-pleomorphic rhabdomyosarcoma, adjuvant chemotherapy has no standard role in high risk STS. In metastatic disease little progress has been made, but during recent years much effort has been put into the development of better clinical study protocols, with stratification of patients to at least the most common histological subtypes, preventing the dilution of potential treatment efficacy when measuring results over the total heterogeneous group of STS. The outcome of patients with advanced STS is however not only dependent on the introduction of new drugs, but also on the availability of dedicated sarcoma centers in which multidisciplinary teams with the input of all experts from different disciplines, such as pathology, radiology, nuclear medicine, surgery, orthopedics radiotherapy and medical oncology is present. Long delay, wrong histological diagnoses, under- and overtreatment are not in the favor of these patients, neither with regard to outcome, nor with respect to short- and long-term toxicity. Disappointedly, centralization is not a routine part of daily care of STS patients and their care givers. Patient advocacy groups are more and more aware of the relevance of treatment in centers of expertise and are active in guiding the patients to these hospitals. At the same time the sarcoma centers should be pro-active in putting patients into clinical trials, also for rare indications within the STS group, as only in this way a better outcome for this group of patients can be reached.
Collapse
Affiliation(s)
- Winette T A van der Graaf
- Department of Medical Oncology, Radboud University Nijmegen Medical Centre, PO Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | | |
Collapse
|
27
|
Feng Y, Ma PC. Anti-MET targeted therapy has come of age: the first durable complete response with MetMAb in metastatic gastric cancer. Cancer Discov 2012; 1:550-4. [PMID: 22586678 DOI: 10.1158/2159-8290.cd-11-0289] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The MET/hepatocyte growth factor (HGF) signaling pathway plays important roles in oncogenesis and tumor progression in a variety of human cancers. MET/HGF drives an invasive signaling program that can be dysregulated in human cancers through a number of activating mechanisms, including mutations, overexpression, amplification, alternative splicing, and HGF ligand-induced autocrine/paracrine loop signaling. As a testimony of MET-targeting therapeutics is beginning to come to clinical fruition, Catenacci and colleagues report the first case of durable complete response under an anti-MET receptor monoclonal antibody, MetMAb, in a patient with chemotherapy-refractory, advanced gastric cancer metastatic to the liver, found to have high MET gene polysomy and remarkably high serum HGF level. Serum and tissue studies also revealed predictive biomarkers for therapeutic response to MET inhibition.
Collapse
Affiliation(s)
- Yan Feng
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
28
|
Ryan CJ, Rosenthal M, Ng S, Alumkal J, Picus J, Gravis G, Fizazi K, Forget F, Machiels JP, Srinivas S, Zhu M, Tang R, Oliner KS, Jiang Y, Loh E, Dubey S, Gerritsen WR. Targeted MET inhibition in castration-resistant prostate cancer: a randomized phase II study and biomarker analysis with rilotumumab plus mitoxantrone and prednisone. Clin Cancer Res 2012; 19:215-24. [PMID: 23136195 DOI: 10.1158/1078-0432.ccr-12-2605] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate the efficacy, safety, biomarkers, and pharmacokinetics of rilotumumab, a fully human, monoclonal antibody against hepatocyte growth factor (HGF)/scatter factor, combined with mitoxantrone and prednisone (MP) in patients with castration-resistant prostate cancer (CRPC). EXPERIMENTAL DESIGN This double-blinded phase II study randomized (1:1:1) patients with progressive, taxane-refractory CRPC to receive MP (12 mg/m(2) i.v. day 1, 5 mg twice a day orally days 1-21, respectively) plus 15 mg/kg rilotumumab, 7.5 mg/kg rilotumumab, or placebo (i.v. day 1) every 3 weeks. The primary endpoint was overall survival (OS). RESULTS One hundred and forty-four patients were randomized. Median OS was 12.2 versus 11.1 months [HR, 1.10; 80% confidence interval (CI), 0.82-1.48] in the combined rilotumumab versus control arms. Median progression-free survival was 3.0 versus 2.9 months (HR, 1.02; 80% CI, 0.79-1.31). Treatment appeared well tolerated with peripheral edema (24% vs. 8%) being more common with rilotumumab. A trend toward unfavorable OS was observed in patients with high tumor MET expression regardless of treatment. Soluble MET levels increased in all treatment arms. Total HGF levels increased in the rilotumumab arms. Rilotumumab showed linear pharmacokinetics when co-administered with MP. CONCLUSIONS Rilotumumab plus MP had manageable toxicities and showed no efficacy improvements in this estimation study. High tumor MET expression may identify patients with CRPC with poorer prognosis.
Collapse
Affiliation(s)
- Charles J Ryan
- University of California San Francisco Helen Diller Family Comprehensive Cancer Center, San Francisco, CA 94115, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Thompson PA, Chintagumpala M. Targeted therapy in bone and soft tissue sarcoma in children and adolescents. Curr Oncol Rep 2012; 14:197-205. [PMID: 22302601 DOI: 10.1007/s11912-012-0223-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Pediatric soft-tissue and bone sarcomas are a heterogeneous group of tumors of mesenchymal origin which affect approximately 1,500 children in the United States each year. Using multimodal therapy (surgery, radiation, and chemotherapy),the overall 5-year survival rate for children with soft-tissue and bone sarcomas is approximately 60%–70%. However, the prognosis for children with metastatic or recurrent disease is poor; and, furthermore, the improvements in the overall cure rate have slowed. It is highly unlikely that further advances in the treatment of pediatric soft-tissue and bone sarcomas will come from traditional cytotoxic chemotherapy. Based on research advances in understanding the biology of pediatric soft-tissue and bone sarcomas, improved cure rates will likely be driven by new types of treatment which target the specific abnormalities within these tumors. These new targeted therapies may include small molecules, antibodies, or other immunotherapies. This review briefly describes the biology of the major types of pediatric sarcomas, discusses potential targets for new therapy, and highlights some recent and current clinical trials using targeted therapy.
Collapse
Affiliation(s)
- Patrick A Thompson
- Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX 77030-2399, USA.
| | | |
Collapse
|
30
|
Horn LC, Hommel N, Roschlau U, Bilek K, Hentschel B, Einenkel J. Peritumoral stromal remodeling, pattern of invasion and expression of c-met/HGF in advanced squamous cell carcinoma of the cervix uteri, FIGO stages III and IV. Eur J Obstet Gynecol Reprod Biol 2012; 163:76-80. [PMID: 22480414 DOI: 10.1016/j.ejogrb.2012.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2011] [Revised: 02/26/2012] [Accepted: 03/09/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE Different patterns of invasion (PIs) have prognostic impact in several types of cancer and are associated with different grades of peritumoral stromal remodeling, characterized by the desmoplastic stromal response (DSR). One key regulator influencing cellular motility and peritumoral stromal response is c-met/HGF. This study evaluates the association between different PI, peritumoral DSR and its correlation to the expression of c-met/HGF in squamous cell carcinomas of the uterine cervix (CX). STUDY DESIGN 131 advanced stage CX (FIGO III/IV) were re-evaluated histologically regarding PI, using a two-level scoring system. The tumor grows in solid cords/trabeculae in finger-like PI and in very small groups or single cells in spray-like PI. DSR was categorized as none/weak and moderate/strong. The tumors were stained with antibodies against c-met and HGF. The staining of >30% of tumor cells was defined as overexpression. The PI was correlated to the prognostic outcome, different categories of DSR and expression status of c-met and HGF. RESULTS 66.4% of the tumors showed a finger-like, and 33.6% a spray-like PI. The spray-like PI showed a reduced two-year overall survival when compared to the finger-like PI (14.0% vs. 29.1%, respectively; p=0.012), and was associated with moderate/strong DSR. The majority of the tumors showed overexpression of c-met (85.4%) and HGF (74.8%). There was no correlation between the expression status of c-met/HGF and the FIGO stage, peritumoral DSR or the prognostic outcome. CONCLUSIONS Spray-like PI is of prognostic impact in cervical carcinoma FIGO III/IV and is associated with strong peritumoral stromal remodeling. There is no prognostic impact of the immunohistochemical expression of c-met/HGF in advanced stage cervical carcinomas.
Collapse
Affiliation(s)
- L-C Horn
- Institute of Pathology, Division of Breast, Gynecologic and Perinatal Pathology, University of Leipzig, Leipzig, Germany.
| | | | | | | | | | | |
Collapse
|
31
|
HGF/c-Met pathway is one of the mediators of sunitinib-induced tumor cell type-dependent metastasis. Cancer Lett 2012; 320:48-55. [PMID: 22269210 DOI: 10.1016/j.canlet.2012.01.026] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 01/11/2012] [Accepted: 01/14/2012] [Indexed: 11/24/2022]
Abstract
Recent studies in several tumor models indicated that treatment with angiogenic inhibitors may trigger induction of metastasis to other organs. Here we investigated modes of resistance and invasion in several tumor cell lines including 4T1 (breast), H460 (lung) and Colo205 (colorectal) using sunitinib at doses comparable to clinically utilized regimen. In comparison with vehicle-treated tumors, sunitinib increased metastasis to lung in 4T1 tumors and to peritoneal lymph node in Colo205 tumors. However, the same treatment did not induce invasiveness in H460 tumors, further suggesting that accelerating metastasis during treatment with angiogenic inhibitors is tumor cell-type dependent. Interestingly, Crizotinib (a dual inhibitor of c-Met and ALK pathways) as single agent or in combination with sunitinib reduced metastasis in all models tested suggesting a role for c-Met/HGF pathway in intrinsic- or sunitinib-induced-metastasis. Moreover, ELISA data showed that while c-Met is highly enriched in tumor cells, HGF is secreted mainly by the stroma (mouse HGF) suggesting a paracrine fashion for c-Met pathway activation in the tumors. In conclusion, our findings indicate that sunitinib-induced metastasis is tumor cell-type dependent and further supports a rationale for combination of anti-angiogenics and c-Met inhibition in the clinic.
Collapse
|
32
|
Abstract
Soft tissue sarcomas (STS) are malignancies of mesenchymal origin that represent approximately 1% of cancers in adults. Systematic research into the treatment of STS is challenging given its rarity and disease heterogeneity. Despite the ability to histologically subtype STS, only recently has our approach to therapy begun to differentiate along these lines. The purpose of this review is to highlight emerging therapeutic targets and therapies that hold the potential to add to the current state of systemic treatment for STS.
Collapse
Affiliation(s)
- Jason L Smith
- Divisions of Hematology, Medical Oncology, Cellular Therapy, Duke University Medical Center, Durham, NC 27710, USA.
| | | |
Collapse
|
33
|
Abstract
The use of monoclonal antibodies (mAbs) has become a general approach for specifically targeting and treating human disease. In oncology, the therapeutic utility of mAbs is usually evaluated in the context of treatment with standard of care, as well as other small molecule targeted therapies. Many anti-cancer antibody modalities have achieved validation, including the targeting of growth factor and angiogenesis pathways, the induction of tumor cell killing or apoptosis, and the blocking of immune inhibitory mechanisms to stimulate anti-tumor responses. But, as with other targeted therapies, few antibodies are curative because of biological complexities that underlie tumor formation and redundancies in molecular pathways that enable tumors to adapt and show resistance to treatment. This review discusses the combinations of antibody therapeutics that are emerging to improve efficacy and durability within a specific biological mechanism (e.g., immunomodulation or the inhibition of angiogenesis) and across multiple biological pathways (e.g., inhibition of tumor growth and induction of tumor cell apoptosis).
Collapse
|
34
|
Zhang YW, Staal B, Essenburg C, Su Y, Kang L, West R, Kaufman D, Dekoning T, Eagleson B, Buchanan SG, Vande Woude GF. MET kinase inhibitor SGX523 synergizes with epidermal growth factor receptor inhibitor erlotinib in a hepatocyte growth factor-dependent fashion to suppress carcinoma growth. Cancer Res 2010; 70:6880-90. [PMID: 20643778 DOI: 10.1158/0008-5472.can-10-0898] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The hepatocyte growth factor (HGF)-MET pathway supports several hallmark cancer traits, and it is frequently activated in a broad spectrum of human cancers (http://www.vai.org/met/). With the development of many cancer drugs targeting this pathway, there is a need for relevant in vivo model systems for preclinical evaluation of drug efficacy. Here, we show that production of the human HGF ligand in transgenic severe combined immunodeficient mice (hHGF(tg)-SCID mice) enhances the growth of many MET-expressing human carcinoma xenografts, including those derived from lung, breast, kidney, colon, stomach, and pancreas. In this model, the MET-specific small-molecule kinase inhibitor SGX523 partially inhibits the HGF-dependent growth of lung, breast, and pancreatic tumors. However, much greater growth suppression is achieved by combinatorial inhibition with the epidermal growth factor receptor (EGFR) kinase inhibitor erlotinib. Together, these results validate the hHGF(tg)-SCID mouse model for in vivo determination of MET sensitivity to drug inhibition. Our findings also indicate that simultaneously targeting the MET and EGFR pathways can provide synergistic inhibitory effects for the treatment of cancers in which both pathways are activated.
Collapse
Affiliation(s)
- Yu-Wen Zhang
- Van Andel Research Institute, Grand Rapids, MI, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Buchanan IM, Scott T, Tandle AT, Burgan WE, Burgess TL, Tofilon PJ, Camphausen K. Radiosensitization of glioma cells by modulation of Met signalling with the hepatocyte growth factor neutralizing antibody, AMG102. J Cell Mol Med 2010; 15:1999-2006. [PMID: 20629992 PMCID: PMC2976812 DOI: 10.1111/j.1582-4934.2010.01122.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The hepatocyte growth factor (HGF)/Met signalling pathway is up-regulated in many cancers, with downstream mediators playing a role in DNA double strand break repair. Previous studies have shown increased radiosensitization of tumours through modulation of Met signalling by genetic methods. We investigated the effects of the anti-HGF monoclonal antibody, AMG102, on the response to ionizing radiation in a model of glioblastoma multiforme in vitro and in vivo. Radiosensitivity was evaluated in vitro in the U-87 MG human glioma cell line. Met activation was measured by Western blot, and the effect on survival following radiation was evaluated by clonogenic assay. Mechanism of cell death was evaluated by apoptosis and mitotic catastrophe assays. DNA damage was quantitated by γH2AX foci and neutral comet assay. Growth kinetics of subcutaneous tumours was used to assess the effects of AMG102 on in vivo tumour radiosensitivity. AMG102 inhibited Met activation after irradiation. An enhancement of radiation cell killing was shown with no toxicity using drug alone. Retention of γH2AX foci at 6 and 24 hrs following the drug/radiation combination indicated an inhibition of DNA repair following radiation, and comet assay confirmed DNA damage persisting over the same duration. At 48 and 72 hrs following radiation, a significant increase of cells undergoing mitotic catastrophe was seen in the drug/radiation treated cells. Growth of subcutaneous tumours was slowed in combination treated mice, with an effect that was greater than additive for each modality individually. Modulation of Met signalling with AMG102 may prove a novel radiation sensitizing strategy. Our data indicate that DNA repair processes downstream of Met are impaired leading to increased cell death through mitotic catastrophe.
Collapse
Affiliation(s)
- Ian M Buchanan
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|