1
|
Ge X, Zhang Y, Huang F, Wu Y, Pang J, Li X, Fan F, Liu H, Li S. EGFR tyrosine kinase inhibitor Almonertinib induces apoptosis and autophagy mediated by reactive oxygen species in non-small cell lung cancer cells. Hum Exp Toxicol 2021; 40:S49-S62. [PMID: 34219533 DOI: 10.1177/09603271211030554] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
Almonertinib, a new third-generation epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor, is highly selective to EGFR T790M-mutant non-small cell lung cancer (NSCLC). However, there is no available information on the form and molecular mechanism of Almonertinib-induced death in NSCLC cells. Herein, CCK-8 and colony formation assays, flow cytometry, electron microscopy, and western blots assay showed that Almonertinib inhibited NSCLC cells growth and proliferation by inducing apoptosis and autophagy which can be inhibited by a broad spectrum of caspase inhibitor Z-VAD-fmk or autophagy inhibitor chloroquine. Importantly, Almonertinib-induced autophagy was cytoprotective in NSCLC cells, and the blockade of autophagy improved cell apoptosis. In addition, Almonertinib increased reactive oxygen species (ROS) generation and clearance of ROS through pretreatment with N-acetyl-L-cysteine (NAC) inhibited the decrease of cell viability, apoptosis and increase of LC3-II induced by Almonertinib. The results of Western blot showed that both EGFR activity and downstream signaling pathways were inhibited by Almonertinib. Taken together, these findings indicated that Almonertinib induced apoptosis and autophagy by promoting ROS production in NSCLC cells.
Collapse
Affiliation(s)
- X Ge
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Y Zhang
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - F Huang
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - Y Wu
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - J Pang
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - X Li
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - F Fan
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - H Liu
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| | - S Li
- Faculty of Pharmacy, Bengbu Medical College, Bengbu, Anhui, People's Republic of China
| |
Collapse
|
2
|
Pfohl U, Pflaume A, Regenbrecht M, Finkler S, Graf Adelmann Q, Reinhard C, Regenbrecht CRA, Wedeken L. Precision Oncology Beyond Genomics: The Future Is Here-It Is Just Not Evenly Distributed. Cells 2021; 10:928. [PMID: 33920536 PMCID: PMC8072767 DOI: 10.3390/cells10040928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/14/2021] [Indexed: 12/14/2022] Open
Abstract
Cancer is a multifactorial disease with increasing incidence. There are more than 100 different cancer types, defined by location, cell of origin, and genomic alterations that influence oncogenesis and therapeutic response. This heterogeneity between tumors of different patients and also the heterogeneity within the same patient's tumor pose an enormous challenge to cancer treatment. In this review, we explore tumor heterogeneity on the longitudinal and the latitudinal axis, reviewing current and future approaches to study this heterogeneity and their potential to support oncologists in tailoring a patient's treatment regimen. We highlight how the ideal of precision oncology is reaching far beyond the knowledge of genetic variants to inform clinical practice and discuss the technologies and strategies already available to improve our understanding and management of heterogeneity in cancer treatment. We will focus on integrating multi-omics technologies with suitable in vitro models and their proficiency in mimicking endogenous tumor heterogeneity.
Collapse
Affiliation(s)
- Ulrike Pfohl
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Molekulare Biowissenschaften, Goethe Universität Frankfurt am Main, Theodor-W.-Adorno-Platz 1, 60323 Frankfurt am Main, Germany
| | - Alina Pflaume
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Manuela Regenbrecht
- Helios Klinikum Berlin-Buch, Schwanebecker Chaussee 50, 13125 Berlin, Germany;
| | - Sabine Finkler
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Quirin Graf Adelmann
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christoph Reinhard
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| | - Christian R. A. Regenbrecht
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
- Institut für Pathologie, Universitätsklinikum Göttingen, Robert-Koch-Straße 40, 37075 Göttingen, Germany
| | - Lena Wedeken
- CELLphenomics GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany; (U.P.); (A.P.); (C.R.); (Q.G.A.); (C.R.A.R.)
- ASC Oncology GmbH, Robert-Rössle-Str. 10, 13125 Berlin, Germany;
| |
Collapse
|
3
|
Yokoyama D, Mukai M, Uda S, Kishima K, Koike T, Hasegawa S, Izumi H, Yamamoto S, Tajima T, Nomura E, Makuuchi H. Efficacy of modified bevacizumab-XELOX therapy in Japanese patients with stage IV recurrent or non-resectable colorectal cancer. J Gastrointest Oncol 2021; 12:527-534. [PMID: 34012646 DOI: 10.21037/jgo-20-350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Background Neoadjuvant chemotherapy (NAC) has been conducted for patients with non-resectable colorectal cancer; however, few reports of a systematic approach to NAC exist. At our hospital, bevacizumab with capecitabine and oxaliplatin (B-mab XELOX) has been used as chemotherapy for Stage IV colorectal cancer since 2014. We aimed to evaluate the efficacy and safety of NAC with a molecular-targeting agent for Stage IV colorectal cancer. Methods A retrospective, single-institute analysis was performed including 27 patients with advanced recurrent cancer following primary tumor resection and 43 patients with non-resectable tumors and remote metastasis. At the time of resection, 17 were receiving chemotherapy. All 70 patients received at least 3 cycles of B-mab XELOX (total: 920 cycles). We determined the 1-year progression-free survival (1Y-PFS), 1-year overall survival (1Y-OS), 3Y-PFS, 3Y-OS, and number of treatment cycles. The objective response rate, clinical benefit rate, and adverse events were assessed. The number of chemotherapy cycles, survival time, and R0 surgery rate were determined for patients who underwent RO conversion surgery. Results The 1Y-PFS was 28.5% [median survival time (MST): 7.4 months], 1Y-OS was 76.6% (MST not reached), 3Y-PFS was 5.5% (MST: 7.4 months), and 3Y-OS was 26.4% (MST: 25.2 months). The mean and median number of cycles of B-mab XELOX was 13.1 and 10.5, respectively. The objective response rate was 28.6%, and the clinical benefit rate was 58.6%. Grade 1 or Grade 2 adverse events occurred in 60 patients (85.7%); however, they all resolved without intervention. A single Grade 4 event (perforation of the primary tumor) occurred in 1 patient (1.4%). RO conversion surgery was performed in 7 patients (10.0%; primary + liver in 2 patients, primary + lung in 1 patient, liver in 3 patients, and primary in 1 patient). These patients received 3 to 10 cycles preoperatively (mean: 7.3; median: 6.5). R0 surgery was achieved in 5 of the 7 patients (71.4%). Postoperative survival ranged from 1 to 26 months (MST: 8 months). Conclusions This modified regimen was safe and effective in Japanese patients, and a high quality of life/quality-adjusted life-year was achieved. To further evaluate PFS and OS, more patients are being investigated.
Collapse
Affiliation(s)
- Daiki Yokoyama
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Masaya Mukai
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Shuji Uda
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Kyouko Kishima
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Takuya Koike
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Sayuri Hasegawa
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Hideki Izumi
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Souichirou Yamamoto
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Takayuki Tajima
- Department of Surgery, Tokai University Tokyo Hospital, Shibuya, Tokyo, Japan
| | - Eiji Nomura
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| | - Hiroyasu Makuuchi
- Department of Surgery, Tokai University Hachioji Hospital, Hachioji, Tokyo, Japan
| |
Collapse
|
4
|
Chuang SC, Huang CW, Chen YT, Ma CJ, Tsai HL, Chang TK, Su WC, Hsu WH, Kuo CH, Wang JY. Effect of KRAS and NRAS mutations on the prognosis of patients with synchronous metastatic colorectal cancer presenting with liver-only and lung-only metastases. Oncol Lett 2020; 20:2119-2130. [PMID: 32782529 PMCID: PMC7400335 DOI: 10.3892/ol.2020.11795] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 03/26/2020] [Indexed: 12/24/2022] Open
Abstract
It has been reported that 20–25% of patients with colorectal cancer (CRC) have metastases at the time of diagnosis. Liver and lung are the most common metastatic sites. The aim of the present study was to investigate the association of KRAS and NRAS mutations with clinicopathological features and prognosis of patients with initial liver-metastasis only (LiM-only) or lung-metastasis only (LuM-only) metastatic CRC (mCRC). Overall, 166 patients with CRC with initial LiM-only (n=124) and LuM-only (n=42) were retrospectively analyzed from January 2014 to December 2017. The median follow-up time was 19.2 months (1.0–57.1 months). Patient characteristics at diagnosis were collected. Genomic DNA was isolated from frozen primary CRC tissues for targeting KRAS and NRAS. Patients with LuM-only were significantly older compared with those with LiM-only (65.5 vs. 61.5 years; P=0.05). There was no significant differences between the LiM-only and LuM-only groups in terms of sex, location of the primary tumor, serum carcinoembryonic antigen level, histological grade and RAS mutation status. KRAS mutations were detected in 43 (41.0%) patients with LiM-only and 13 (35.1%) patients with LuM-only. The overall survival time (OS) of LuM-only was more favorable compared with that of patients with LiM-only (44.5 vs. 24.7 months); however, there was no significant difference (P=0.095). The progression-free survival (PFS) and OS in the RAS wild-type group were significantly improved compared with the RAS mutant cohorts (P=0.004 and P=0.031, respectively) in the LiM-only group. In patients with stage IV CRC, those with synchronous LiM-only mCRC had a higher incidence of metastasis but a less favorable PFS and OS compared with patients with LuM-only. RAS mutation status exhibited a significant association with the survival outcome in patients with LiM-only mCRC.
Collapse
Affiliation(s)
- Shih-Chang Chuang
- Division of General and Digestive Surgery, Department of Surgery; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Ching-Wen Huang
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Yi-Ting Chen
- Department of Pathology; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C.,Department of Pathology, Faculty of Medicine, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Cheng-Jen Ma
- Division of General and Digestive Surgery, Department of Surgery; Kaohsiung Medical University Hospital, Kaohsiung 807, Taiwan, R.O.C
| | - Hsiang-Lin Tsai
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Tsung-Kun Chang
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wei-Chih Su
- Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Wen-Hung Hsu
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Chao-Hung Kuo
- Division of Gastroenterology, Department of Internal Medicine, Kaohsiung Medical University Hospital and Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| | - Jaw-Yuan Wang
- Department of Surgery, Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Division of Colorectal Surgery, Department of Surgery, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C.,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Cohort Research Center, College of Medicine, Kaohsiung 807, Taiwan, R.O.C.,Center for Cancer Research, Kaohsiung Medical University, Kaohsiung 807, Taiwan, R.O.C
| |
Collapse
|
5
|
Delineation of cell death mechanisms induced by synergistic effects of statins and erlotinib in non-small cell lung cancer cell (NSCLC) lines. Sci Rep 2020; 10:959. [PMID: 31969600 PMCID: PMC6976657 DOI: 10.1038/s41598-020-57707-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
Hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors (statins) have been shown to overcome tyrosine kinase inhibitor (TKI) resistance in epithelial growth factor receptor (EGFR) mutated non-small cell lung cancer (NSCLC) cells in vivo and in vitro. However, little is known about the putative induction of non-apoptotic cell death pathways by statins. We investigated the effects of pitavastatin and fluvastatin alone or in combination with erlotinib in three NSCLC cell lines and examined the activation of different cell death pathways. We assessed apoptosis via fluorometric caspase assay and poly (ADP-ribose) polymerase 1 (PARP) cleavage. Furthermore, annexinV/propidium iodide (PI) flow cytometry was performed. Small molecule inhibitors benzyloxycarbonyl-Val-Ala-Asp-fluoromethyl ketone (zVAD), necrostatin 1 (Nec1), ferrostatin 1 (Fer1), Ac-Lys-Lys-Norleucinal (Calp1) were used to characterise cell death pathway(s) putatively (co-)activated by pitavastatin/erlotinib co-treatment. Synergism was calculated by additivity and isobolographic analyses. Pitavastatin and fluvastatin induced cell death in EGFR TKI resistant NSCLC cells lines A549, Calu6 and H1993 as shown by caspase 3 activation and PARP cleavage. Co-treatment of cells with pitavastatin and the EGFR TKI erlotinib resulted in synergistically enhanced cytotoxicity compared to pitavastatin monotherapy. Flow cytometry indicated the induction of alternative regulated cell death pathways. However, only co-treatment with mevalonic acid (Mev) or the pan-caspase inhibitor zVAD could restore cell viability. The results show that cytotoxicity mediated by statin/erlotinib co-treatment is synergistic and can overcome erlotinib resistance in K-ras mutated NSCLC and relies only on apoptosis.
Collapse
|
6
|
Angeles AKJ, Yu RTD, Cutiongco-De La Paz EM, Garcia RL. Phenotypic characterization of the novel, non-hotspot oncogenic KRAS mutants E31D and E63K. Oncol Lett 2019; 18:420-432. [PMID: 31289513 PMCID: PMC6540134 DOI: 10.3892/ol.2019.10325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Accepted: 04/02/2019] [Indexed: 12/13/2022] Open
Abstract
KRAS proto-oncogene, GTPase (KRAS) functions as a molecular switch at the apex of multiple signaling pathways controlling cell proliferation, differentiation, migration, and survival. Canonical KRAS mutants, such as those in codons 12 and 13, produce constitutively active oncoproteins that short-circuit epidermal growth factor receptor (EGFR)-initiated signaling, resulting in dysregulated downstream effectors associated with cellular transformation. Therefore, anti-EGFR therapy provides little to no clinical benefit to patients with activating KRAS mutations. Current genotyping procedures based on canonical mutation detection only account for ~40% of non-responders, highlighting the need to identify additional predictive biomarkers. In the present study, two novel non-hotspot KRAS mutations were functionally characterized in vitro: KRAS E31D was identified from a genetic screen of colorectal cancer specimens at the UP-National Institutes of Health. KRAS E63K is curated in the Catalogue of Somatic Mutations in Cancer database. Similar to the canonical mutants KRAS G12D and KRAS G13D, NIH3T3 cells overexpressing KRAS E31D and KRAS E63K showed altered morphology and were characteristically smaller, rounder, and highly refractile compared with their non-transformed counterparts. Filamentous actin staining also indicated cytoplasmic shrinkage, membrane ruffling, and formation of pseudopod protrusions. Further, they displayed higher proliferative rates and higher migratory rates in scratch wound assays compared with negative controls. These empirical findings suggest the activating impact of the novel KRAS mutations, which may contribute to resistance to anti-EGFR therapy. Complementary studies to elucidate the molecular mechanisms underlying the transforming effect of the rare mutants are required. In parallel, their oncogenic capacity in vivo should also be investigated.
Collapse
Affiliation(s)
- Arlou Kristina J Angeles
- Disease Molecular Biology and Epigenetics Laboratory, National Institute of Molecular Biology and Biotechnology, National Science Complex, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Ryan Timothy D Yu
- Disease Molecular Biology and Epigenetics Laboratory, National Institute of Molecular Biology and Biotechnology, National Science Complex, University of the Philippines Diliman, Quezon City 1101, Philippines
| | - Eva Maria Cutiongco-De La Paz
- Institute of Human Genetics, National Institutes of Health, University of the Philippines Manila, Manila 1000, Philippines.,Philippine Genome Center, University of the Philippines System, Quezon City 1101, Philippines
| | - Reynaldo L Garcia
- Disease Molecular Biology and Epigenetics Laboratory, National Institute of Molecular Biology and Biotechnology, National Science Complex, University of the Philippines Diliman, Quezon City 1101, Philippines.,Philippine Genome Center, University of the Philippines System, Quezon City 1101, Philippines
| |
Collapse
|
7
|
Liu X, Lukowski JK, Flinders C, Kim S, Georgiadis RA, Mumenthaler SM, Hummon AB. MALDI-MSI of Immunotherapy: Mapping the EGFR-Targeting Antibody Cetuximab in 3D Colon-Cancer Cell Cultures. Anal Chem 2018; 90:14156-14164. [PMID: 30479121 DOI: 10.1021/acs.analchem.8b02151] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Immunotherapies are treatments that use a patient's immune system to combat disease. One important type of immunotherapy employed in cancer treatments is the delivery of monoclonal antibodies to block growth receptors. In this manuscript, we develop a methodology that enables accurate and simple evaluation of antibody-type drug delivery using MALDI-MSI. To overcome the mass-range limitation that prevents the detection of large therapeutic antibodies, we used in situ reduction and alkylation to break disulfide bonds to generate smaller fragments. These smaller fragments are more readily ionized and detected by MALDI-MSI without loss of spatial information on the parent drug. As a proof of concept study, we evaluated the distribution of cetuximab in 3D colon cell cultures. Cetuximab is a monoclonal antibody that binds to the extracellular domain of epidermal-growth-factor receptor (EGFR), which is often overexpressed in colorectal cancer (CRC) and mediates cell differentiation, proliferation, migration, and angiogenesis. Cetuximab directly inhibits tumor growth and metastasis and induces apoptosis. By performing on-tissue reduction followed by MALDI-MSI analysis, we successfully mapped the time-dependent penetration and distribution of cetuximab in spheroids derived from two different colon-cancer cell lines (HT-29 and DLD-1). The localization patterns were further confirmed with IF staining of the drug. Changes in other biomolecules following drug treatment were also observed, including the elevation of ATP in spheroids. The developed method has also been applied to map cetuximab distribution in patient-derived colorectal-tumor organoids (CTOs). Overall, we believe this powerful label-free approach will be useful for visualizing the heterogeneous distribution of antibody drugs in tissues and tumors and will help to monitor and optimize their use in the clinic.
Collapse
Affiliation(s)
- Xin Liu
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute , University of Notre Dame , 152 McCourtney Hall , Notre Dame , Indiana 46556 , United States
| | - Jessica K Lukowski
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute , University of Notre Dame , 152 McCourtney Hall , Notre Dame , Indiana 46556 , United States.,Department of Chemistry and Biochemistry and Comprehensive Cancer Center , The Ohio State University , 414 Biomedical Research Tower , Columbus , Ohio 43210 , United States
| | - Colin Flinders
- Lawrence J. Ellison Institute for Transformative Medicine , University of Southern California , 2250 Alcazar Street, CSC 240 , Los Angeles , California 90033 , United States
| | - Seungil Kim
- Lawrence J. Ellison Institute for Transformative Medicine , University of Southern California , 2250 Alcazar Street, CSC 240 , Los Angeles , California 90033 , United States
| | - Rebecca A Georgiadis
- Department of Chemistry and Biochemistry and the Harper Cancer Research Institute , University of Notre Dame , 152 McCourtney Hall , Notre Dame , Indiana 46556 , United States
| | - Shannon M Mumenthaler
- Lawrence J. Ellison Institute for Transformative Medicine , University of Southern California , 2250 Alcazar Street, CSC 240 , Los Angeles , California 90033 , United States
| | - Amanda B Hummon
- Department of Chemistry and Biochemistry and Comprehensive Cancer Center , The Ohio State University , 414 Biomedical Research Tower , Columbus , Ohio 43210 , United States
| |
Collapse
|
8
|
Huxley N, Crathorne L, Varley-Campbell J, Tikhonova I, Snowsill T, Briscoe S, Peters J, Bond M, Napier M, Hoyle M. The clinical effectiveness and cost-effectiveness of cetuximab (review of technology appraisal no. 176) and panitumumab (partial review of technology appraisal no. 240) for previously untreated metastatic colorectal cancer: a systematic review and economic evaluation. Health Technol Assess 2018; 21:1-294. [PMID: 28682222 DOI: 10.3310/hta21380] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Colorectal cancer is the fourth most commonly diagnosed cancer in the UK after breast, lung and prostate cancer. People with metastatic disease who are sufficiently fit are usually treated with active chemotherapy as first- or second-line therapy. Targeted agents are available, including the antiepidermal growth factor receptor (EGFR) agents cetuximab (Erbitux®, Merck Serono UK Ltd, Feltham, UK) and panitumumab (Vecitibix®, Amgen UK Ltd, Cambridge, UK). OBJECTIVE To investigate the clinical effectiveness and cost-effectiveness of panitumumab in combination with chemotherapy and cetuximab in combination with chemotherapy for rat sarcoma (RAS) wild-type (WT) patients for the first-line treatment of metastatic colorectal cancer. DATA SOURCES The assessment included a systematic review of clinical effectiveness and cost-effectiveness studies, a review and critique of manufacturer submissions, and a de novo cohort-based economic analysis. For the assessment of effectiveness, a literature search was conducted up to 27 April 2015 in a range of electronic databases, including MEDLINE, EMBASE and The Cochrane Library. REVIEW METHODS Studies were included if they were randomised controlled trials (RCTs) or systematic reviews of RCTs of cetuximab or panitumumab in participants with previously untreated metastatic colorectal cancer with RAS WT status. All steps in the review were performed by one reviewer and checked independently by a second. Narrative synthesis and network meta-analyses (NMAs) were conducted for outcomes of interest. An economic model was developed focusing on first-line treatment and using a 30-year time horizon to capture costs and benefits. Costs and benefits were discounted at 3.5% per annum. Scenario analyses and probabilistic and univariate deterministic sensitivity analyses were performed. RESULTS The searches identified 2811 titles and abstracts, of which five clinical trials were included. Additional data from these trials were provided by the manufacturers. No data were available for panitumumab plus irinotecan-based chemotherapy (folinic acid + 5-fluorouracil + irinotecan) (FOLFIRI) in previously untreated patients. Studies reported results for RAS WT subgroups. First-line treatment with anti-EGFR therapies in combination with chemotherapy appeared to have statistically significant benefits for patients who are RAS WT. For the independent economic evaluation, the base-case incremental cost-effectiveness ratio (ICER) for RAS WT patients for cetuximab plus oxaliplatin-based chemotherapy (folinic acid + 5-fluorouracil + oxaliplatin) (FOLFOX) compared with FOLFOX was £104,205 per quality-adjusted life-year (QALY) gained; for panitumumab plus FOLFOX compared with FOLFOX was £204,103 per QALY gained; and for cetuximab plus FOLFIRI compared with FOLFIRI was £122,554 per QALY gained. The ICERs were sensitive to treatment duration, progression-free survival, overall survival (resected patients only) and resection rates. LIMITATIONS The trials included RAS WT populations only as subgroups. No evidence was available for panitumumab plus FOLFIRI. Two networks were used for the NMA and model, based on the different chemotherapies (FOLFOX and FOLFIRI), as insufficient evidence was available to the assessment group to connect these networks. CONCLUSIONS Although cetuximab and panitumumab in combination with chemotherapy appear to be clinically beneficial for RAS WT patients compared with chemotherapy alone, they are likely to represent poor value for money when judged by cost-effectiveness criteria currently used in the UK. It would be useful to conduct a RCT in patients with RAS WT. STUDY REGISTRATION This study is registered as PROSPERO CRD42015016111. FUNDING The National Institute for Health Research Health Technology Assessment programme.
Collapse
Affiliation(s)
- Nicola Huxley
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Louise Crathorne
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Jo Varley-Campbell
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Irina Tikhonova
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Tristan Snowsill
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Simon Briscoe
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Jaime Peters
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Mary Bond
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| | - Mark Napier
- Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Martin Hoyle
- Peninsula Technology Assessment Group (PenTAG), University of Exeter Medical School, Exeter, UK
| |
Collapse
|
9
|
Park S, Kim JY, Lee SH, Suh B, Keam B, Kim TM, Kim DW, Heo DS. KRAS G12C mutation as a poor prognostic marker of pemetrexed treatment in non-small cell lung cancer. Korean J Intern Med 2017; 32:514-522. [PMID: 28407465 PMCID: PMC5432792 DOI: 10.3904/kjim.2015.299] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/12/2015] [Accepted: 10/19/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND/AIMS The predictive and prognostic value of KRAS mutation and its type of mutations in non-small cell lung cancer (NSCLC) are controversial. This clinical study was designed to investigate the predictive value of KRAS mutations and its mutation types to pemetrexed and gemcitabine based treatment. METHODS Advanced NSCLC patients tested for KRAS mutation (n = 334) were retrospectively reviewed and 252 patients with wild type epidermal growth factor receptor and no anaplastic lymphoma kinase fusion were enrolled for the analysis. KRAS mutations were observed in 45 subjects with mutation type as followed: G12C (n = 13), G12D (n = 12), G12V (n = 12), other (n = 8). Response rate (RR), progression-free survival (PFS), and overall survival (OS) of pemetrexed singlet and gemcitabine based chemotherapy were analysis. RESULTS Age, sex, performance status were well balanced between subjects with or without KRAS mutations. No difference was observed in RR. Hazard ratio (HR) of PFS for pemetrexed treated subjects with G12C mutation compared to subjects with KRAS wild type was 1.96 (95% confidential interval [CI], 1.01 to 3.79; p = 0.045), but other mutations failed to show clinical significance. By analysis done by PFS, compared to the subjects with transition mutation, HR was 1.48 (95% CI, 0.64 to 3.40; p = 0.360) for subjects with transversion mutation on pemetrexed treatment and 0.41 (95% CI, 0.19 to 0.87; p = 0.020) for subjects treated with gemcitabine based chemotherapy. No difference was observed in OS. CONCLUSIONS In this study, different drug sensitivity was observed according to the type of KRAS mutation. NSCLC subpopulations with different KRAS mutation type should be considered as different subgroups and optimal chemotherapy regimens should be searched in further confirmative studies.
Collapse
Affiliation(s)
- Sehhoon Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Ji-Yeon Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Se-Hoon Lee
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
- Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul 03080, Korea Tel: +82-2-2072-2199 Fax: +82-2-762-9662 E-mail:
| | - Beomseok Suh
- Department of Family Medicine and Health Promotion Center, Seoul National University Hospital, Seoul, Korea
| | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Tae Min Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dong-Wan Kim
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Dae Seog Heo
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| |
Collapse
|
10
|
Jain S, Shankaran V. The Economics of Personalized Therapy in Metastatic Colorectal Cancer. CURRENT COLORECTAL CANCER REPORTS 2016. [DOI: 10.1007/s11888-016-0318-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
11
|
Pai SG, Fuloria J. Novel therapeutic agents in the treatment of metastatic colorectal cancer. World J Gastrointest Oncol 2016; 8:99-104. [PMID: 26798440 PMCID: PMC4714150 DOI: 10.4251/wjgo.v8.i1.99] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/20/2015] [Accepted: 12/04/2015] [Indexed: 02/05/2023] Open
Abstract
Over the past couple of decades considerable progress has been made in the management of metastatic colorectal cancers (mCRC) leading to a significant improvement in five-year survival. Although part of this success has been rightly attributed to aggressive surgical management and advances in other adjunct treatments, our understanding of the pathogenesis of cancer and emergence of newer molecular targets for colon cancer has created a powerful impact. In this review article we will discuss various targeted therapies in the management of mCRC. Newer agents on the horizon soon to be incorporated in clinical practice will be briefly reviewed as well.
Collapse
|
12
|
Zheng Z, He X, Xie C, Hua S, Li J, Wang T, Yao M, Vignarajan S, Teng Y, Hejazi L, Liu B, Dong Q. Targeting cytosolic phospholipase A2 α in colorectal cancer cells inhibits constitutively activated protein kinase B (AKT) and cell proliferation. Oncotarget 2015; 5:12304-16. [PMID: 25365190 PMCID: PMC4322978 DOI: 10.18632/oncotarget.2639] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 10/28/2014] [Indexed: 01/05/2023] Open
Abstract
A constitutive activation of protein kinase B (AKT) in a hyper-phosphorylated status at Ser473 is one of the hallmarks of anti-EGFR therapy-resistant colorectal cancer (CRC). The aim of this study was to examine the role of cytosolic phospholipase A2α (cPLA2α) on AKT phosphorylation at Ser473 and cell proliferation in CRC cells with mutation in phosphoinositide 3-kinase (PI3K). AKT phosphorylation at Ser473 was resistant to EGF stimulation in CRC cell lines of DLD-1 (PIK3CAE545K mutation) and HT-29 (PIK3CAP499T mutation). Over-expression of cPLA2α by stable transfection increased basal and EGF-stimulated AKT phosphorylation and proliferation in DLD-1 cells. In contrast, silencing of cPLA2α with siRNA or inhibition with Efipladib decreased basal and EGF-stimulated AKT phosphorylation and proliferation in HT-29. Treating animals transplanted with DLD-1 with Efipladib (10 mg/kg, i.p. daily) over 14 days reduced xenograft growth by >90% with a concomitant decrease in AKT phosphorylation. In human CRC tissue, cPLA2α expression and phosphorylation were increased in 63% (77/120) compared with adjacent normal mucosa determined by immunohistochemistry. We conclude that cPLA2α is required for sustaining AKT phosphorylation at Ser473 and cell proliferation in CRC cells with PI3K mutation, and may serve as a potential therapeutic target for treatment of CRC resistant to anti-EGFR therapy.
Collapse
Affiliation(s)
- Zhong Zheng
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiangyi He
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chanlu Xie
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia. School of Science and Health, The University of Western Sydney, Australia
| | - Sheng Hua
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Jianfang Li
- Shanghai Key Laboratory of Gastric Neoplasms, Department of Surgery, Shanghai Institute of Digestive Surgery, and Gastroenterology, Ruijin Hospital, Jiaotong University School of Medicine, Shanghai, China
| | - Tingfeng Wang
- Department of General Surgery, Nanhui Central Hospital. Shanghai, China
| | - Mu Yao
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Soma Vignarajan
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Ying Teng
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia
| | - Leila Hejazi
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia. School of Science and Health, The University of Western Sydney, Australia
| | - Bingya Liu
- Shanghai Key Laboratory of Gastric Neoplasms, Department of Surgery, Shanghai Institute of Digestive Surgery, and Gastroenterology, Ruijin Hospital, Jiaotong University School of Medicine, Shanghai, China
| | - Qihan Dong
- Central Clinical School and Bosch Institute, The University of Sydney and Department of Endocrinology and Sydney Cancer Centre, Royal Prince Alfred Hospital, Sydney, Australia. School of Science and Health, The University of Western Sydney, Australia
| |
Collapse
|
13
|
Steinbach C, Steinbrücker C, Pollok S, Walther K, Clement JH, Chen Y, Petersen I, Cialla-May D, Weber K, Popp J. KRAS mutation screening by chip-based DNA hybridization--a further step towards personalized oncology. Analyst 2015; 140:2747-54. [PMID: 25706807 DOI: 10.1039/c4an02086c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of predictive biomarkers can help to improve therapeutic options for the individual cancer patient. For the treatment of colon cancer patients with anti-EGFR-based drugs, the KRAS mutation status has to be determined to pre-select responders that will benefit from this medication. Amongst others, array-based tests have been established for profiling of the KRAS mutation status. Within this article we describe an on-chip hybridization technique to screen therapeutic relevant KRAS codon 12 mutations. The DNA chip-based platform enables the reliable discrimination of selected mutations by allele-specific hybridization. Here, silver deposits represent robust endpoint signals that allow for a simple naked eye rating. With the here presented assay concept a precise identification of heterozygous and homozygous KRAS mutations, even against a background of up to 95% wild-type DNA, was realizable. The applicability of the test was successfully proven for various cancer cell lines as well as clinical tumour samples. Thus, the chip-based DNA hybridization technique seems to be a promising tool for KRAS mutation analysis to further improve personalized cancer treatment.
Collapse
Affiliation(s)
- Christine Steinbach
- Leibniz Institute of Photonic Technology Jena, Albert-Einstein-Str. 9, 07745 Jena, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Natalicchio MI, Improta G, Zupa A, Cursio OE, Stampone E, Possidente L, Teresa Gerardi AM, Vita G, Martini M, Cassano A, Piccoli C, Romito S, Aieta M, Antonetti R, Barone C, Landriscina M. Pyrosequencing evaluation of low-frequency KRAS mutant alleles for EGF receptor therapy selection in metastatic colorectal carcinoma. Future Oncol 2014; 10:713-23. [PMID: 24799053 DOI: 10.2217/fon.13.233] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
AIM To evaluate whether pyrosequencing (PS) improves the KRAS mutational status predictive value. PATIENTS & METHODS A retrospective analysis of KRAS mutations by PS and direct sequencing (DS) in 192 metastatic colorectal carcinomas (mCRCs), subgrouped in 51 KRAS mutated at PS and 141 KRAS wild-type at DS. RESULTS DS failed to detect low-frequency KRAS mutations in four out of 51 mCRCs, whereas PS detected 12 additional low-frequency KRAS mutations in 141 mCRCs KRAS wild-type at DS. After reanalyzing by PS 97 KRAS wild-type tumors treated with anti-EGF receptor (EGFR) antibodies, nine additional mutations were revealed in nonresponders, whereas none of responders exhibited a KRAS-mutated genotype. Of note, KRAS-mutated tumors upon PS showed a worst progression-free survival after EGFR therapy. Finally, PS allowed the detection of additional NRAS, BRAF and exon 20 PIK3CA mutations mostly in KRAS wild-type mCRCs resistant to EGFR therapy. CONCLUSION PS detection of low-frequency mutations may improve the KRAS predictive value for EGFR therapy selection.
Collapse
|
15
|
Myers MB, McKinzie PB, Wang Y, Meng F, Parsons BL. ACB-PCR quantification of somatic oncomutation. Methods Mol Biol 2014; 1105:345-63. [PMID: 24623241 DOI: 10.1007/978-1-62703-739-6_27] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Allele-specific competitive blocker-polymerase chain reaction (ACB-PCR) is a sensitive approach for the selective amplification of an allele. Using the ACB-PCR technique, hotspot point mutations in oncogenes and tumor-suppressor genes (oncomutations) are being developed as quantitative biomarkers of cancer risk. ACB-PCR employs a mutant specific primer (with a 3'-penultimate mismatch relative to the mutant DNA sequence, but a double 3'-terminal mismatch relative to the wild-type DNA sequence) to selectively amplify rare mutant DNA molecules. A blocker primer (having a non-extendable 3'-end and with a 3'-penultimate mismatch relative to the wild-type DNA sequence, but a double 3'-terminal mismatch relative to the mutant DNA sequence) is included in ACB-PCR to selectively repress amplification from the abundant wild-type molecules. Consequently, ACB-PCR is capable of quantifying the level of a single basepair substitution mutation in a DNA population when present at a mutant:wild type ratio of 10(-5) or greater. Quantification of rare mutant alleles is achieved by parallel analysis of unknown samples and mutant fraction (MF) standards (defined mixtures of mutant and wild-type DNA sequences). The ability to quantify specific mutations with known association to cancer has several important applications, including evaluating the carcinogenic potential of chemical exposures in rodent models and in the diagnosis and treatment of cancer. This chapter provides a step-by-step description of the ACB-PCR methodology as it has been used to measure human KRAS codon 12 GGT to GAT mutation.
Collapse
Affiliation(s)
- Meagan B Myers
- Division of Genetic and Molecular Toxicology, National Center for Toxicological Research, U.S. Food and Drug Administration, 3900 NCTR Road, HFT-120, Jefferson, AR, 72079, USA,
| | | | | | | | | |
Collapse
|
16
|
Qin Y, Liu XJ, Li L, Liu XJ, Li Y, Gao RJ, Shao RG, Zhen YS. MMP-2/9-oriented combinations enhance antitumor efficacy of EGFR/HER2-targeting fusion proteins and gemcitabine. Oncol Rep 2014; 32:121-30. [PMID: 24807584 DOI: 10.3892/or.2014.3169] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Accepted: 04/02/2014] [Indexed: 11/05/2022] Open
Abstract
To increase the antitumor efficacy, in the present study, we proposed several settings of matrix metalloproteinase (MMP)-2/9-oriented combinations that comprise the MMP-2/9-targeting fusion protein dFv-LDP and the MMP inhibitor doxycycline (DOX) in association with EGFR/HER2-bispecific fusion protein Ec-LDP-Hr, its enediyne-energized analogue Ec-LDP-Hr-AE, and gemcitabine (GEM). The expressions of various fusion proteins were detected by western blot analysis. Proliferation and migration inhibition of cells were determined by MTT and Transwell assay, respectively. The binding capability of dFv-LDP and Ec-LDP-Hr to cancer cells was examined by ELISA, cell immunofluorescence coimmunoprecipitation and confocal assays. Animal experiments were set to investigate the antitumor efficacy of various combinations against colorectal carcinoma HCT-15 xenograft in athymic mice. These two targeting proteins dFv-LDP and Ec-LDP-Hr had strong binding capabilities and antiproliferation effects on various cancer cell lines. Enhanced therapeutic efficacy in vivo was observed in the MMP-2/9-targeting fusion protein dFv-LDP integrated combinations including: i) dFv-LDP and Ec-LDP-Hr, ii) dFv-LDP and enediyne-energized fusion protein Ec-LDP-Hr-AE, iii) dFv-LDP and Ec-LDP-Hr-AE plus DOX, and iv) dFv-LDP and GEM plus DOX against colorectal cancer HCT-15 xenograft in athymic mice. In setting iii, DOX (20 mg/kg), dFv-LDP (20 mg/kg) and Ec-LDP-Hr-AE (0.3 mg/kg) alone suppressed tumor growth by 35, 49.7 and 67.5%, respectively. The combination of dFv-LDP and Ec-LDP-Hr-AE was 75.1%. Furthermore, this combination plus DOX showed stronger efficacy with an inhibitory rate of 82.7%. In setting iv, the combination of dFv-LDP and GEM suppressed tumor growth by 66.3%. Notably, the tumor inhibitory rate of the dFv-LDP/GEM/DOX combination reached 85.5%, producing initial shrinkage after the first administration. The MMP-2/9-oriented combination strategy that employs the MMP-2/9-targeting antibody-based fusion protein and the small molecular inhibitor DOX as the basic composed agents may enhance antitumor efficacy in association with the EGFR/HER2-targeting fusion protein and GEM. This multiple targeting approach may be useful for enhancing antitumor efficacy against colorectal cancer.
Collapse
Affiliation(s)
- Ye Qin
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Xiu-Jun Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Liang Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Xu-Jie Liu
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Yi Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Rui-Juan Gao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Rong-Guang Shao
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| | - Yong-Su Zhen
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, P.R. China
| |
Collapse
|
17
|
Werner HMJ, Trovik J, Halle MK, Wik E, Akslen LA, Birkeland E, Bredholt T, Tangen IL, Krakstad C, Salvesen HB. Stathmin protein level, a potential predictive marker for taxane treatment response in endometrial cancer. PLoS One 2014; 9:e90141. [PMID: 24587245 PMCID: PMC3934991 DOI: 10.1371/journal.pone.0090141] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 01/21/2014] [Indexed: 12/30/2022] Open
Abstract
Stathmin is a prognostic marker in many cancers, including endometrial cancer. Preclinical studies, predominantly in breast cancer, have suggested that stathmin may additionally be a predictive marker for response to paclitaxel. We first evaluated the response to paclitaxel in endometrial cancer cell lines before and after stathmin knock-down. Subsequently we investigated the clinical response to paclitaxel containing chemotherapy in metastatic endometrial cancer in relation to stathmin protein level in tumors. Stathmin level was also determined in metastatic lesions, analyzing changes in biomarker status on disease progression. Knock-down of stathmin improved sensitivity to paclitaxel in endometrial carcinoma cell lines with both naturally higher and lower sensitivity to paclitaxel. In clinical samples, high stathmin level was demonstrated to be associated with poor response to paclitaxel containing chemotherapy and to reduced disease specific survival only in patients treated with such combination. Stathmin level increased significantly from primary to metastatic lesions. This study suggests, supported by both preclinical and clinical data, that stathmin could be a predictive biomarker for response to paclitaxel treatment in endometrial cancer. Re-assessment of stathmin level in metastatic lesions prior to treatment start may be relevant. Also, validation in a randomized clinical trial will be important.
Collapse
Affiliation(s)
- Henrica M. J. Werner
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
- * E-mail:
| | - Jone Trovik
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Mari K. Halle
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Elisabeth Wik
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Lars A. Akslen
- Centre for Cancer Biomarkers, Department of Clinical Medicine, The University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Even Birkeland
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Therese Bredholt
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Ingvild L. Tangen
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
| | - Helga B. Salvesen
- Centre for Cancer Biomarkers, Department of Clinical Science, The University of Bergen, Bergen, Norway
- Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
18
|
Macerelli M, Caramella C, Faivre L, Besse B, Planchard D, Polo V, Ngo Camus M, Celebic A, Koubi-Pick V, Lacroix L, Pignon JP, Soria JC. Does KRAS mutational status predict chemoresistance in advanced non-small cell lung cancer (NSCLC)? Lung Cancer 2014; 83:383-8. [PMID: 24439569 DOI: 10.1016/j.lungcan.2013.12.013] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Revised: 12/15/2013] [Accepted: 12/20/2013] [Indexed: 01/06/2023]
Abstract
BACKGROUND Clinical implications of KRAS mutational status in advanced non-small cell lung cancer (NSCLC) remain unclear. To clarify this point, we retrospectively explored whether KRAS mutations could impact tumor response, and disease control rate (DCR) to first-line platinum-based chemotherapy (CT) as well as progression-free survival (PFS) or overall survival (OS). METHODS Between June 2009 and June 2012, 340 patients with advanced (stage IIIB/IV) NSCLC were reviewed in a single institution (Institut Gustave Roussy). Two hundred and one patients had a biomolecular profile and received a platinum-based first-line CT. Patients with an unknown mutational status or with actionable alterations were excluded. We retained two groups: patients with KRAS mutated tumor (MUT) and patients with wild-type KRAS/EGFR (WT). Multivariate analyses with Cox model were used. Survival curves were calculated with Kaplan-Meier method. RESULTS One hundred and eight patients were included in the analysis: 39 in the MUT group and 69 in the WT group. Baseline radiological assessment demonstrated more brain (P=0.01) and liver (P=0.04) metastases in MUT patients. DCR was 76% for MUT vs. 91% for WT group (P=0.03), regardless of the type of platinum-based CT (use of pemetrexed or not). Although no statistically significant differences were found, shorter PFS (4.9 vs. 6.0 months; P=0.79) and OS (10.3 vs. 13.2 months; P=0.40) were observed for patients with KRAS mutated tumors in univariate analysis. CONCLUSIONS KRAS mutant tumors had a lower DCR after the first-line platinum-based CT, but this difference did not translate in PFS or OS. The presence of KRAS mutations may confer a more aggressive disease, with greater baseline incidence of hepatic and cerebral metastases.
Collapse
Affiliation(s)
- M Macerelli
- Department of Medical Oncology, Gustave Roussy, Villejuif, France; Department of Medical Oncology, University Hospital, Udine, Italy.
| | - C Caramella
- Department of Radiology, Gustave Roussy, Villejuif, France
| | - L Faivre
- Department of Biostatistics and Epidemiology, Gustave Roussy, Villejuif, France
| | - B Besse
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - D Planchard
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - V Polo
- Department of Medical Oncology, Gustave Roussy, Villejuif, France; Department of Second Medical Oncology, Oncologic Venetian Institute, Padua, Italy
| | - M Ngo Camus
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - A Celebic
- Department of Biostatistics and Epidemiology, Gustave Roussy, Villejuif, France
| | - V Koubi-Pick
- Translational Research Laboratory, Gustave Roussy, Villejuif, France
| | - L Lacroix
- Translational Research Laboratory, Gustave Roussy, Villejuif, France
| | - J P Pignon
- Department of Biostatistics and Epidemiology, Gustave Roussy, Villejuif, France
| | - J C Soria
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| |
Collapse
|
19
|
Liu ET, Johnston PG. Personalized medicine: does the molecular suit fit? Oncologist 2013; 18:653-4. [PMID: 23814161 PMCID: PMC4063388 DOI: 10.1634/theoncologist.2013-0191] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 05/16/2013] [Indexed: 01/03/2023] Open
Affiliation(s)
- Edison T. Liu
- The Jackson Laboratory Cancer Center, Bar Harbor, Maine, USA
| | - Patrick G. Johnston
- Dentistry and Biomedical Sciences, Queen's University Belfast, United Kingdom
| |
Collapse
|
20
|
Pradhan MP, Nagulapalli K, Palakal MJ. Cliques for the identification of gene signatures for colorectal cancer across population. BMC SYSTEMS BIOLOGY 2012; 6 Suppl 3:S17. [PMID: 23282040 PMCID: PMC3524317 DOI: 10.1186/1752-0509-6-s3-s17] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Colorectal cancer (CRC) is one of the most commonly diagnosed cancers worldwide. Studies have correlated risk of CRC development with dietary habits and environmental conditions. Gene signatures for any disease can identify the key biological processes, which is especially useful in studying cancer development. Such processes can be used to evaluate potential drug targets. Though recognition of CRC gene-signatures across populations is crucial to better understanding potential novel treatment options for CRC, it remains a challenging task. Results We developed a topological and biological feature-based network approach for identifying the gene signatures across populations. In this work, we propose a novel approach of using cliques to understand the variability within population. Cliques are more conserved and co-expressed, therefore allowing identification and comparison of cliques across a population which can help researchers study gene variations. Our study was based on four publicly available expression datasets belonging to four different populations across the world. We identified cliques of various sizes (0 to 7) across the four population networks. Cliques of size seven were further analyzed across populations for their commonality and uniqueness. Forty-nine common cliques of size seven were identified. These cliques were further analyzed based on their connectivity profiles. We found associations between the cliques and their connectivity profiles across networks. With these clique connectivity profiles (CCPs), we were able to identify the divergence among the populations, important biological processes (cell cycle, signal transduction, and cell differentiation), and related gene pathways. Therefore the genes identified in these cliques and their connectivity profiles can be defined as the gene-signatures across populations. In this work we demonstrate the power and effectiveness of cliques to study CRC across populations. Conclusions We developed a new approach where cliques and their connectivity profiles helped elucidate the variation and similarity in CRC gene profiles across four populations with unique dietary habits.
Collapse
Affiliation(s)
- Meeta P Pradhan
- School of Informatics, Indiana University Purdue University Indianapolis, IN, USA
| | | | | |
Collapse
|
21
|
Gasch C, Bauernhofer T, Pichler M, Langer-Freitag S, Reeh M, Seifert AM, Mauermann O, Izbicki JR, Pantel K, Riethdorf S. Heterogeneity of epidermal growth factor receptor status and mutations of KRAS/PIK3CA in circulating tumor cells of patients with colorectal cancer. Clin Chem 2012; 59:252-60. [PMID: 23136247 DOI: 10.1373/clinchem.2012.188557] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Molecular characterization of circulating tumor cells (CTCs) is pivotal to increasing the diagnostic specificity of CTC assays and investigating therapeutic targets and their downstream pathways on CTCs. We focused on epidermal growth factor receptor (EGFR) and genes relevant for its inhibition in patients with colorectal cancer (CRC). METHODS We used the CellSearch® system for CTC detection in peripheral blood samples from 49 patients with metastatic CRC (mCRC) and 32 patients with nonmetastatic CRC (nmCRC). We assessed EGFR expression in 741 CTCs from 27 patients with mCRC and 6 patients with nmCRC using a fluorescein-conjugated antibody with the CellSearch Epithelial Cell Kit. DNA of a single CTC isolated by micromanipulation was propagated by whole-genome amplification and analyzed by quantitative PCR for EGFR gene amplification and sequencing for KRAS (v-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog), BRAF (v-raf murine sarcoma viral oncogene homolog B1), and PIK3CA (phosphatidylinositol-4,5-bisphosphate 3-kinase, catalytic subunit α) mutations. RESULTS At least 2 CTCs were detected in 24 of 49 patients with mCRC and 7 of 32 patients with nmCRC. In 7 of 33 patients, CTCs with increased EGFR expression were identified. Heterogeneity in EGFR expression was observed between CTCs from the same patient. EGFR gene amplification was found in 7 of 26 CTCs from 3 patients. The investigated BRAF gene locus was not mutated in 44 analyzed CTCs, whereas KRAS mutations were detected in 5 of 15 CTCs from 1 patient and PIK3CA mutations in 14 of 36 CTCs from 4 patients. CONCLUSIONS Molecular characterization of single CTCs demonstrated considerable intra- and interpatient heterogeneity of EGFR expression and genetic alterations in EGFR, KRAS, and PIK3CA, possibly explaining the variable response rates to EGFR inhibition in patients with CRC.
Collapse
Affiliation(s)
- Christin Gasch
- Department of Tumor Biology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Duffy MJ, McGowan PM, Crown J. Targeted therapy for triple-negative breast cancer: where are we? Int J Cancer 2012; 131:2471-7. [PMID: 22581656 DOI: 10.1002/ijc.27632] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2012] [Accepted: 04/05/2012] [Indexed: 12/29/2022]
Abstract
Breast cancers that are negative for estrogen receptor (ER), progesterone receptors (PR) and HER2, using standard clinical assays, have been dubbed triple-negative (TN). Unlike other molecular subtypes of invasive breast cancer, validated targeted therapies are currently unavailable for patients with TN breast cancer. Preclinical studies however, have identified several potential targets such as epidermal growth factor receptor (EGFR), SRC, MET and poly ADP ribose polymerase 1/2 (PARP1/2). Because of tumor heterogeneity, it is unlikely that any single targeted therapy will be efficacious in all patients with TN breast cancer. The rational way forward for treating these patients is likely to be biomarker-driven, combination targeted therapies or combination of targeted therapy with cytotoxic chemotherapy.
Collapse
Affiliation(s)
- Michael J Duffy
- UCD Clinical Research Centre, St. Vincent's University Hospital, Dublin, Ireland.
| | | | | |
Collapse
|
23
|
Abstract
Advances in our understanding of the intricate molecular mechanisms for transformation of a normal cell to a cancer cell, and the aberrant control of complementary pathways, have presented a much more complex set of challenges for the diagnostic and therapeutic disciplines than originally appreciated. The oncology field has entered an era of personalized medicine where treatment selection for each cancer patient is becoming individualized or customized. This advance reflects the molecular and genetic composition of the tumors and progress in biomarker technology, which allow us to align the most appropriate treatment according to the patient's disease. There is a worldwide acceptance that advances in our ability to identify predictive biomarkers and provide them as companion diagnostics for stratifying and subgrouping patients represents the next leap forward in improving the quality of clinical care in oncology. As such, we are progressing from a population-based empirical 'one drug fits all' treatment model, to a focused personalized approach where rational companion diagnostic tests support the drug's clinical utility by identifying the most responsive patient subgroup.
Collapse
|
24
|
Voon PJ, Kong HL. Tumour Genetics and Genomics to Personalise Cancer Treatment. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2011. [DOI: 10.47102/annals-acadmedsg.v40n8p362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Personalising cancer treatment to optimise therapeutic efficacy while minimising exposure to the toxicities of ineffective drugs is the holy grail of medical oncology. Clinical parameters and conventional histopathological characterisations of cancers are no longer adequate to guide the practising oncologists in treatment planning. The explosion of knowledge in cancer molecular biology has led to the availability of tumour-specific molecules that serve as predictive and prognostic markers. In breast cancer, HER-2 positivity is a good predictor for success of anti-HER-2 trastuzumab monoclonal antibody therapy. K-ras mutational status predicts the likelihood of response to anti-EGFR monoclonal antibodies in advanced colorectal cancers. Similarly, EGFR mutational status in pulmonary adenocarcinoma is highly predictive for responses or otherwise to tyrosine kinase inhibitors. Notwithstanding our deeper understanding of tumour biology and the availability of predictive and prognostic laboratory tools, we are still far from achieving our dream of the perfect personalised cancer treatment, as each tumour in a particular patient is unique to itself. A much coveted, real-time, anti-tumour drug sensitivity testing in the future may one day pave the way for truly treating the right tumour with the right drug in the right patient.
Key words: Personalised cancer treatment, Predictive markers, Prognostic markers
Collapse
Affiliation(s)
| | - Hwai Loong Kong
- Mount Elizabeth Medical Centre & Novena Medical Center, Singapore
| |
Collapse
|
25
|
Harrison S, Benziger H. The molecular biology of colorectal carcinoma and its implications: A review. Surgeon 2011; 9:200-10. [DOI: 10.1016/j.surge.2011.01.011] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Revised: 01/17/2011] [Accepted: 01/23/2011] [Indexed: 02/07/2023]
|
26
|
Use of molecular markers for predicting therapy response in cancer patients. Cancer Treat Rev 2010; 37:151-9. [PMID: 20685042 DOI: 10.1016/j.ctrv.2010.07.004] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2010] [Revised: 07/07/2010] [Accepted: 07/09/2010] [Indexed: 12/27/2022]
Abstract
Predictive markers are factors that are associated with upfront response or resistance to a particular therapy. Predictive markers are important in oncology as tumors of the same tissue of origin vary widely in their response to most available systemic therapies. Currently recommended oncological predictive markers include both estrogen and progesterone receptors for identifying patients with breast cancers likely to benefit from hormone therapy, HER-2 for the identification of breast cancer patients likely to benefit from trastuzumab, specific K-RAS mutations for the identification of patients with advanced colorectal cancer unlikely to benefit from either cetuximab or panitumumab and specific EGFR mutations for selecting patients with advanced non-small-cell lung cancer for treatment with tyrosine kinase inhibitors such as gefitinib and erlotinib. The availability of predictive markers should increase drug efficacy and decrease toxicity, thus leading to a more personalized approach to cancer treatment.
Collapse
|