1
|
Lee Y, Park SY, Lee GK, Lim HJ, Choi YR, Kim J, Han JY. Detailed characterization of combination treatment with MET inhibitor plus EGFR inhibitor in EGFR-mutant and MET-amplified non-small cell lung cancer. Transl Lung Cancer Res 2024; 13:2511-2523. [PMID: 39507027 PMCID: PMC11535821 DOI: 10.21037/tlcr-24-273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/20/2024] [Indexed: 11/08/2024]
Abstract
Background Detailed clinical data about combination treatment with MET inhibitor (METi) and EGFR inhibitor (EGFRi) is lacking in patients with EGFR-mutant, MET-amplified, and EGFRi-resistant non-small cell lung cancer (NSCLC). This study aimed to report longitudinal data on the efficacy and safety of this combination treatment. Methods We retrospectively analyzed 44 patients with advanced EGFR-mutant and MET-amplified NSCLC who were treated with any types of METi plus EGFRi after progression with EGFRi at the National Cancer Center Hospital. Longitudinal clinicogenomic data and plasma circulating tumor DNA (ctDNA) data were collected. Results The overall response rate was 74.4% and median progression-free survival (PFS) was 5.3 months [95% confidence interval (CI): 3.3-7.3]. Twenty-three patients (52.3%) required either or both treatment discontinuation due to adverse effects. The main cause of discontinuation was pneumonitis (69.2%). There was no significant difference in the PFS of patients with or without METi discontinuation [hazard ratio (HR), 0.93; 95% CI: 0.49-1.78; P=0.83]. Median clearance time of MET amplification in plasma ctDNA was measured as 63 days. Patients who stopped METi within 63 days of initiation showed poorer PFS compared to those who discontinued after (HR, 2.78; 95% CI: 1.00-7.75; P=0.050). Diverse resistance mechanisms including on-target mutations in MET (D1246H) and EGFR (C797S or T790M) were detected in 14 patients. One MET D1246H-mutant case and one EGFR C797S-mutant case responded to sitravatinib and amivantamab, respectively. Conclusions A combination of METi and EGFRi showed a promising anti-tumor effect in advanced EGFR-mutant and MET-amplified NSCLC. Pneumonitis was the main adverse effects leading to treatment discontinuation. Early discontinuation of METi negatively affected the survival outcomes.
Collapse
Affiliation(s)
- Youngjoo Lee
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center Hospital, Goyang, Republic of Korea
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center Hospital, Goyang, Republic of Korea
| | - Geon Kook Lee
- Department of Pathology, National Cancer Center Hospital, Goyang, Republic of Korea
| | - Hyun-Ju Lim
- Department of Radiology, National Cancer Center Hospital, Goyang, Republic of Korea
| | - Yu-Ra Choi
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Jaemin Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Ji-Youn Han
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center Hospital, Goyang, Republic of Korea
| |
Collapse
|
2
|
Yang M, Mandal E, Liu FX, O’Hara RM, Lesher B, Sanborn RE. Non-small cell lung cancer with MET amplification: review of epidemiology, associated disease characteristics, testing procedures, burden, and treatments. Front Oncol 2024; 13:1241402. [PMID: 38273845 PMCID: PMC10808753 DOI: 10.3389/fonc.2023.1241402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/27/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Mesenchymal-epidermal transition factor gene amplification (METamp) is being investigated as a therapeutic target in advanced non-small cell lung cancer (NSCLC). We reviewed the epidemiology and disease characteristics associated with primary and secondary METamp, as well as the testing procedures used to identify METamp, in advanced NSCLC. Economic and humanistic burdens, and the practice patterns and treatments under investigation for METamp were also examined. Methods Embase and Medline (via ProQuest), ClinicalTrials.gov, and Cochrane Controlled Register of Trials (2015-2022) were systematically searched. Conference abstracts were searched via Embase and conference proceedings websites (2020-2022). The review focused on evidence from the United States; global evidence was included for identified evidence gaps. Results The median rate of primary METamp in NSCLC across the references was 4.8% (n=4 studies) and of secondary METamp (epidermal growth factor receptor [EGFR]-mutant NSCLC) was 15% (n=10). Next-generation sequencing (NGS; n=12) and/or fluorescence in situ hybridization (FISH; n=11) were most frequently used in real-world studies and FISH testing most frequently used in clinical trials (n=9/10). METamp definitions varied among clinical trials using ISH/FISH testing (MET to chromosome 7 centromere ratio of ≥1.8 to ≥3.0; or gene copy number [GCN] ≥5 to ≥10) and among trials using NGS (tissue testing: GCN ≥6; liquid biopsy: MET copy number ≥2.1 to >5). Limited to no data were identified on the economic and humanistic burdens, and real-world treatment of METamp NSCLC. Promising preliminary results from trials enrolling patients with EGFR-mutated, METamp advanced NSCLC progressing on an EGFR-tyrosine kinase inhibitor (TKI) were observed with MET-TKIs (i.e., tepotinib, savolitinib, and capmatinib) in combination with EGFR-TKIs (i.e., gefitinib and osimertinib). For metastatic NSCLC and high-level METamp, monotherapy with capmatinib, crizotinib, and tepotinib are recommended in the 2022 published NSCLC NCCN Guidelines. Conclusion Primary METamp occurs in approximately 5% of NSCLC cases, and secondary METamp in approximately 15% of cases previously treated with an EGFR inhibitor. Variability in testing methods (including ISH/FISH and NGS) and definitions were observed. Several treatments are promising in treating METamp NSCLC. Additional studies evaluating the clinical, economic, and humanistic burdens are needed.
Collapse
Affiliation(s)
- Mo Yang
- North America Evidence and Value Development, North America Medical Affairs, EMD Serono, Inc., Rockland, MA, United States, an affiliate of Merck KGaA
| | - Erin Mandal
- Evidence and Access, OPEN Health, Parsippany, NJ, United States
| | - Frank X. Liu
- North America Evidence and Value Development, North America Medical Affairs, EMD Serono, Inc., Rockland, MA, United States, an affiliate of Merck KGaA
| | - Richard M. O’Hara
- North America Evidence and Value Development, North America Medical Affairs, EMD Serono, Inc., Rockland, MA, United States, an affiliate of Merck KGaA
| | - Beth Lesher
- Evidence and Access, OPEN Health, Parsippany, NJ, United States
| | - Rachel E. Sanborn
- Earle A. Chiles Research Institute, Providence Cancer Institute, Portland, OR, United States
| |
Collapse
|
3
|
Choi YR, Kang EH, Kim S, Park SY, Han JY, Lee Y. Single targeting of MET in EGFR-mutated and MET-amplified non-small cell lung cancer. Br J Cancer 2023; 128:2186-2196. [PMID: 37059804 PMCID: PMC10241937 DOI: 10.1038/s41416-023-02264-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/21/2023] [Accepted: 03/27/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND In EGFR-mutant and MET-amplified lung cancer resistant to EGFR inhibitors, double blockade of EGFR and MET is considered as a reasonable strategy despite increasing toxicity. This study evaluated the single MET inhibition in these specific tumours. METHODS We investigated the efficacy of a single MET inhibitor in EGFR-mutant, MET-amplified lung cancer cells (HCC827GR) and the matched clinical cases and patient-derived cells. Acquired resistance mechanisms to single MET inhibitor were further explored. RESULTS Single MET inhibitor sufficiently inhibited the EGFR downstream signalling and proliferation in the HCC827GR cells. The MET-inhibitor-sensitive clones had similar EGFR mutation allele frequency as the MET-inhibitor-resistant clones. The patients with EGFR-mutant, MET-amplified lung cancer resistant to EGFR inhibitors showed definite response to single MET inhibitor but the response duration was not durable. The MET gene copy number in their plasma circulating tumour DNA was significantly decreased during the treatment and was not re-increased after progression. In the cells resistant to single MET inhibitor, the EGFR pathway was reactivated, and gefitinib alone successfully suppressed their growth. CONCLUSIONS Single MET inhibition produced a short-lived response in EGFR-mutant and MET-amplified lung cancer. A further study of a novel combination therapy schedule is needed to achieve long-lasting efficacy and less toxicity.
Collapse
Affiliation(s)
- Yu-Ra Choi
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Eun Hye Kang
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Sunshin Kim
- Research Institute, National Cancer Center, Goyang, Republic of Korea
| | - Seog-Yun Park
- Department of Pathology, National Cancer Center, Goyang, Republic of Korea
| | - Ji-Youn Han
- Research Institute, National Cancer Center, Goyang, Republic of Korea
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea
| | - Youngjoo Lee
- Research Institute, National Cancer Center, Goyang, Republic of Korea.
- Division of Hematology and Oncology, Department of Internal Medicine, National Cancer Center, Goyang, Republic of Korea.
| |
Collapse
|
4
|
Osteopontin and Cancer: Insights into Its Role in Drug Resistance. Biomedicines 2023; 11:biomedicines11010197. [PMID: 36672705 PMCID: PMC9855437 DOI: 10.3390/biomedicines11010197] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/04/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023] Open
Abstract
Cancer is one of the leading causes of mortality worldwide. Currently, drug resistance is the main obstacle in cancer treatments with the underlying mechanisms of drug resistance yet to be fully understood. Osteopontin (OPN) is a member of the integrin binding glycophosphoprotein family that is overexpressed in several tumour types. It is involved in drug transport, apoptosis, stemness, energy metabolism, and autophagy, which may contribute to drug resistance. Thus, understanding the role of OPN in cancer drug resistance could be important. This review describes the OPN-based mechanisms that might contribute to cancer drug resistance, demonstrating that OPN may be a viable target for cancer therapy to reduce drug resistance in sensitive tumours.
Collapse
|
5
|
Zhu X, Lu Y, Lu S. Landscape of Savolitinib Development for the Treatment of Non-Small Cell Lung Cancer with MET Alteration-A Narrative Review. Cancers (Basel) 2022; 14:cancers14246122. [PMID: 36551608 PMCID: PMC9776447 DOI: 10.3390/cancers14246122] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 12/06/2022] [Accepted: 12/07/2022] [Indexed: 12/15/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is increasingly being treated with targeted therapies. Savolitinib (Orpathys®) is highly selective mesenchymal epithelial transition (MET)-tyrosine kinase inhibitor (TKI), which is conditionally approved in China for advanced NSCLC with MET exon 14 skipping mutations (METex14). This article summarizes the clinical development of savolitinib, as a monotherapy in NSCLC with METex14 mutation and in combination with epidermal growth factor receptor (EGFR) inhibitor in post EGFR-TKI resistance NSCLC due to MET-based acquired resistance. Preclinical models demonstrated anti-tumor activities in MET-driven cancer cell line and xenograft tumor models. The Phase Ia/Ib study established an optimized, recommended phase II dose in Chinese NSCLC patients, while TATTON study of savolitinib plus osimertinib in patients with EGFR mutant, MET-amplified and TKI-progressed NSCLC showed beneficial efficacy with acceptable safety profile. In a pivotal phase II study, Chinese patients with pulmonary sarcomatoid carcinoma, brain metastasis and other NSCLC subtype positive for METex14 mutation showed notable responses and acceptable safety profile with savolitinib. Currently, results from ongoing clinical trials are eagerly anticipated to confirm the efficacious and safety benefits of savolitinib as monotherapy and in combination with EGFR-TKI in acquired resistance setting in advanced NSCLC and its subtypes with MET alterations.
Collapse
Affiliation(s)
- Xiaokuan Zhu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yao Lu
- AstraZeneca China, Shanghai 201200, China
| | - Shun Lu
- Department of Shanghai Lung Cancer Center, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence:
| |
Collapse
|
6
|
Liu L, Qu J, Heng J, Zhou C, Xiong Y, Yang H, Jiang W, Zeng L, Zhu S, Zhang Y, Tan J, Hu C, Deng P, Yang N. A Large Real-World Study on the Effectiveness of the Combined Inhibition of EGFR and MET in EGFR-Mutant Non-Small-Cell Lung Cancer After Development of EGFR-TKI Resistance. Front Oncol 2021; 11:722039. [PMID: 34660287 PMCID: PMC8517073 DOI: 10.3389/fonc.2021.722039] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 08/23/2021] [Indexed: 12/16/2022] Open
Abstract
Background MET proto-oncogene amplification (amp) is an important mechanism underlying acquired resistance to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs). However, the optimal treatment strategy after acquiring MET-amp-mediated EGFR-TKI resistance remains controversial. Our study compared three treatment strategies for patients with EGFR-mutant non-small-cell lung cancer (NSCLC) who were detected with MET-amp at EGFR-TKI progression using next-generation sequencing. Methods Of the 70 patients included in the study, 38 received EGFR-TKI + crizotinib, 10 received crizotinib monotherapy, and 22 received chemotherapy. Clinical outcomes and molecular profiles were analyzed. Results The objective response rate was 48.6% for EGFR-TKI + crizotinib group, 40.0% for crizotinib monotherapy group, and 18.2% for chemotherapy group. Patients who received EGFR-TKI + crizotinib had significantly longer progression-free survival than those who received crizotinib or chemotherapy (5.0 vs. 2.3 vs. 2.9 months, p = 0.010), but overall survival was comparable (10.0 vs. 4.1 vs. 8.5 months, p = 0.088). TP53 mutation (58.5%) and EGFR-amp (42.9%) were frequent concurrent mutations of the cohort. Progression-free survival was significantly longer for patients with either concurrent TP53 mutation (n = 17) (6.0 vs. 2.3 vs. 2.9 months, p = 0.009) or EGFR-amp (n = 13) (5.0 vs. 1.2 vs. 2.4 months, p = 0.016) in the EGFR-TKI + crizotinib group than the other two regimen. Potential acquired resistance mechanisms to EGFR-TKI + crizotinib included EGFR-T790M (n = 2), EGFR-L718Q (n = 1), EGFR-S645C (n = 1), MET-D1228H (n = 1), BRAF-V600E (n = 1), NRAS-Q61H (n = 1), KRAS-amp (n = 1), ERBB2-amp (n = 1), CDK4-amp (n = 1), and MYC-amp (n = 1). Conclusion Our study provides real-world clinical evidence from a large cohort that simultaneous inhibition of EGFR and MET could be a more effective therapeutic strategy for patients with MET-amp acquired from EGFR-TKI therapy.
Collapse
Affiliation(s)
- Li Liu
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Jingjing Qu
- Department of Respiratory Disease, Thoracic Disease Centre, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jianfu Heng
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China.,Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Chunhua Zhou
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Yi Xiong
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China.,Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Haiyan Yang
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Wenjuan Jiang
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Liang Zeng
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Songlin Zhu
- Department of Clinical Pharmaceutical Research Institution, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Yongchang Zhang
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Jiarong Tan
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Pengbo Deng
- Department of Respiratory Medicine, Xiangya Hospital of Central South University, Changsha, China
| | - Nong Yang
- Department of Lung Cancer and Gastroenterology, Hunan Cancer Hospital, Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| |
Collapse
|
7
|
Drusbosky LM, Dawar R, Rodriguez E, Ikpeazu CV. Therapeutic strategies in METex14 skipping mutated non-small cell lung cancer. J Hematol Oncol 2021; 14:129. [PMID: 34425853 PMCID: PMC8381548 DOI: 10.1186/s13045-021-01138-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
METex14 skipping mutations occur in about 3–4% of lung adenocarcinoma patients and 1–2% of patients with other lung cancer histology. The MET receptor tyrosine kinase and its ligand hepatocyte growth factor (HGF) are established oncogenic drivers of NSCLC. A mutation that results in loss of exon 14 in the MET gene leads to dysregulation and inappropriate signaling that is associated with increased responsiveness to MET TKIs. Results from GEOMETRY mono-1 and VISION Phase I/II clinical trials demonstrated significant clinical activity in patients treated with the MET Exon 14 skipping mutation inhibitors capmatinib and tepotinib with tolerable toxicity profile. In the GEOMETRY mono-1 trial, capmatinib was especially active in treatment-naïve patients supporting the upfront testing of this oncogenic driver. Tepotinib demonstrated superior activity in the pretreated patients in the VISION trial. Savolitinib is another MET TKI that has shown efficacy in the first- and second-line settings, including patients with aggressive pulmonary sarcomatoid carcinoma. These studies have demonstrated that these TKIs can cross the blood brain barrier and demonstrated some activity toward CNS metastases. MET Exon 14 skipping mutation is detected by NGS-based testing of liquid or tissue biopsies, with preference for RNA-based NGS. The activity of capmatinib and tepotinib is limited by the development of acquired resistance. Current research is focused on strategies to overcome resistance and improve the effectiveness of these agents. Our aim is to review the current status of MET Exon 14 skipping mutation as it pertains NSCLC.
Collapse
Affiliation(s)
| | - Richa Dawar
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA
| | - Estelamari Rodriguez
- Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA
| | - Chukwuemeka V Ikpeazu
- University of Miami Sylvester Comprehensive Cancer Center, 8100 SW 10th Street, Ste 3310F, Plantation, FL, 33324, USA. .,Division of Medical Oncology, Department of Internal Medicine, University of Miami Miller School of Medicine, 1475 NW 12th Avenue, Miami, FL, 33136, USA.
| |
Collapse
|
8
|
Generation and Characterization of a New Preclinical Mouse Model of EGFR-Driven Lung Cancer with MET-Induced Osimertinib Resistance. Cancers (Basel) 2021; 13:cancers13143441. [PMID: 34298655 PMCID: PMC8307933 DOI: 10.3390/cancers13143441] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/01/2021] [Accepted: 07/07/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary The use of targeted therapy has changed the clinical management of lung cancer patients, increasing both their life quality and expectancy. Conversely, the appearance of resistance occurs in almost all patients receiving this therapy. In this regard, new strategies combining different therapies could delay or even eliminate the appearance of resistance. However, in order to develop new therapeutic treatments, we need preclinical mouse models that recapitulate human disease. In the present study, we developed a new state-of-the-art mouse model that summarizes all features occurring in EGFR-mutated patients that relapse after osimertinib after acquisition of MET amplification. Abstract Despite the introduction of epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) to treat advanced lung cancer harboring EGFR-activating mutations, the prognosis remains unfavorable because of intrinsic and/or acquired resistance. We generated a new state-of-the-art mouse strain harboring the human EGFRT790M/L858R oncogene and MET overexpression (EGFR/MET strain) that mimics the MET amplification occurring in one out of five patients with EGFR-mutated lung cancer that relapsed after treatment with osimertinib, a third-generation anti-EGFR TKI. We found that survival was reduced in EGFR/MET mice compared with mice harboring only EGFRT790M/L858R (EGFR strain). Moreover, EGFR/MET-driven lung tumors were resistant to osimertinib, recapitulating the phenotype observed in patients. Conversely, as also observed in patients, the crizotinib (anti-MET TKI) and osimertinib combination improved survival and reduced tumor burden in EGFR/MET mice, further validating the model’s value for preclinical studies. We also found that in EGFR/MET mice, MET overexpression negatively regulated EGFR activity through MIG6 induction, a compensatory mechanism that allows the coexistence of the two onco-genic events. Our data suggest that single EGFR or MET inhibition might not be a good therapeutic option for EGFR-mutated lung cancer with MET amplification, and that inhibition of both pathways should be the best clinical choice in these patients.
Collapse
|
9
|
When the MET receptor kicks in to resist targeted therapies. Oncogene 2021; 40:4061-4078. [PMID: 34031544 DOI: 10.1038/s41388-021-01835-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/07/2021] [Indexed: 02/04/2023]
Abstract
Although targeted therapies have increased the life expectancy of patients with druggable molecular alterations directly involved in tumor development, the efficacy of these therapies is limited by acquired resistances leading to treatment failure. Most targeted therapies, including ones exploiting therapeutic antibodies and kinase inhibitors, are directed against receptor tyrosine kinases (RTKs) or major signaling hubs. Resistances to these therapies arise when inhibition of these targets is bypassed through activation of alternative signaling pathways. In recent years, activation of the receptor tyrosine kinase MET has been shown to promote resistance to various targeted therapies. This casts MET as important actor in resistance. In this review, we describe how the MET receptor triggers resistance to targeted therapies against RTKs such as EGFR, VEGFR, and HER2 and against signaling hubs such as BRAF. We also describe how MET can be its own resistance factor, as illustrated by on-target resistance of lung tumors harboring activating mutations causing MET exon 14 skipping. Interestingly, investigation of all these situations reveals functional physiological relationships between MET and the target of the therapy to which the cancer becomes resistant, suggesting that resistance stems from preexisting mechanisms. Identification of MET as a resistance factor opens the way to co-treatment strategies that are being tested in current clinical trials.
Collapse
|
10
|
Aubanel M, Swalduz A, Avrillon V, Doublet L, Mastroianni B, Neidhardt-Bérard EM, Pérol M. Combining EGFR and MET Inhibition With Crizotinib in EGFR-mutated Lung Adenocarcinoma Harboring MET Amplification: A Brief Report. Clin Lung Cancer 2020; 21:e601-e606. [DOI: 10.1016/j.cllc.2020.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 04/26/2020] [Accepted: 05/14/2020] [Indexed: 02/01/2023]
|
11
|
Wessels S, Muley T, Christopoulos P, Meister M, Heinzmann-Groth I, Warth A, Herpel E, Hummler S, Klingmüller U, Kuon J, Heussel CP, Eberhardt R, Herth FJF, Winter H, Bischoff H, Stenzinger A, Reck M, Huber RM, Thomas M, Schneider MA. Comprehensive serial biobanking in advanced NSCLC: feasibility, challenges and perspectives. Transl Lung Cancer Res 2020; 9:1000-1014. [PMID: 32953480 PMCID: PMC7481602 DOI: 10.21037/tlcr-20-137] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND Availability of tumor material at baseline and disease progression is increasingly important for patient management in non-small-cell lung cancer (NSCLC), especially for the application of targeted therapies like tyrosine kinase inhibitors and for immune checkpoint inhibitor treatment. Here we report the experience of prospective biomaterial acquisition in advanced NSCLC from a pilot project. METHODS Main objective was the longitudinal collection of high-quality, cryoconserved biopsies in addition to formalin-fixed paraffin-embedded (FFPE) biopsies required for routine diagnostics, along with blood samples and detailed clinical annotation using standardized questionnaires. RESULTS Over five years, 205 patients were enrolled for the project, yielding 387 cryoconserved biopsies and 1,098 serum, plasma and buffy-coat samples. The feasibility of obtaining the cryoconserved biopsies in addition to the FFPE biopsies was 89% for newly diagnosed cases, but dropped down to 56% and 47% at first and second disease progression, respectively. While forceps biopsy was the preferred procedure for tissue acquisition, the highest tissue amounts were received using the cryobiopsy method. Biopsies had a median tumor cellularity of 34% and yielded in median 13.6 µg DNA and 12 µg RNA (median RIN =8). During the five-year project, a maximum of 38 follow-up blood samples per patient were assembled in up to four therapy lines. CONCLUSIONS Despite the poor condition and limited prognosis of most NSCLC patients, this serial biomaterial acquisition including routine collection of cryoconserved biopsies is feasible to support individualized management. The standardized collection of high-quality material has enabled and enriched several translational research studies that can advance therapeutic options.
Collapse
Affiliation(s)
- Sabine Wessels
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Thomas Muley
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Petros Christopoulos
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michael Meister
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Ingrid Heinzmann-Groth
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Arne Warth
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
| | - Esther Herpel
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
- NCT Tissue Bank, National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Simone Hummler
- Department of Pneumology and Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Ursula Klingmüller
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Division Systems Biology of Signal Transduction, German Cancer Research Centre (DKFZ), INF 280, Heidelberg, Germany
| | - Jonas Kuon
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Claus-Peter Heussel
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology, University Hospital, D-69120 Heidelberg, Germany
| | - Ralf Eberhardt
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Diagnostic and Interventional Radiology with Nuclear Medicine, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Felix J F Herth
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Pneumology and Critical Care Medicine, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Department of Surgery, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Helge Bischoff
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| | - Albrecht Stenzinger
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Institute of Pathology, University Hospital Heidelberg, D-69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), Heidelberg Site, Germany
| | - Martin Reck
- Department of Thoracic Oncology, Lung Clinic Grosshansdorf, D-22927 Grosshansdorf, Germany
- Airway Research Centre North (ARCN), German Centre for Lung Research (DZL), Grosshansdorf, Germany
| | - Rudolf Maria Huber
- University Hospital Munich and Thoracic Oncology Centre Munich, D-80337 Munich, Germany
- Comprehensive Pneumology Centre Munich (CPC-M), German Centre for Lung Research (DZL), Munich, Germany
| | - Michael Thomas
- Department of Thoracic Oncology, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Marc A Schneider
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
- Translational Research Unit, Thoraxklinik at University Hospital Heidelberg, D-69126 Heidelberg, Germany
| |
Collapse
|
12
|
He Q, Zhu X, Shi X, Lin C, Jin Y, Yan J, He J, Yu X. cfDNA changes for monitoring of targeted therapy in a primary EGFR mutation lung adenocarcinoma. Transl Lung Cancer Res 2020; 9:807-810. [PMID: 32676342 PMCID: PMC7354132 DOI: 10.21037/tlcr-20-442] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Qiong He
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Xian Zhu
- Department of Cardiovascular Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310022, China
| | - Xun Shi
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Chen Lin
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Yin Jin
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| | - Junrong Yan
- Nanjing Geneseeq Technology Inc., Nanjing 210032, China
| | - Jiachen He
- Nanjing Geneseeq Technology Inc., Nanjing 210032, China
| | - Xinmin Yu
- Institute of Cancer and Basic Medicine (ICBM), Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Cancer Hospital of the University of Chinese Academy of Sciences, Hangzhou 310022, China.,Department of Medical Oncology, Zhejiang Cancer Hospital, Hangzhou 310022, China
| |
Collapse
|
13
|
Yu T, Xia Q, Gong T, Wang J, Zhong D. Molecular mechanism of acquired drug resistance in the EGFR-TKI resistant cell line HCC827-TR. Thorac Cancer 2020; 11:1129-1138. [PMID: 32163227 PMCID: PMC7180561 DOI: 10.1111/1759-7714.13342] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/12/2020] [Accepted: 01/14/2020] [Indexed: 01/10/2023] Open
Abstract
Background The first‐line standard treatment of non‐small cell lung cancer (NSCLC) with EGFR mutation is EGFR‐tyrosine kinase inhibitors (TKIs). However, most patients will develop acquired resistance after 9–13 months. This study investigated novel molecular mechanisms of acquired resistance to EGFR‐TKIs to identify a potential new treatment for EGFR‐TKI resistant NSCLC patients. Methods We established an EGFR‐TKI resistant cell line (HCC827‐TR) by culturing the HCC827‐P cell line through continuous erlotinib culture. We used Sanger sequencing, RT‐PCR, and western blot to rule out known resistance mechanisms in HCC827‐TR cells, including EGFR‐T790M and MET, PTEN, or EGFR expression changes. Next‐generation sequencing was performed and identified differentially expressed genes between two cell lines and examined the genes with GO and KEGG pathway database analyses. We also examined the molecular alterations in COSMIC and GDSC databases and performed hazard predictions using SIFT, PolyPhen‐2, Mutation Taster, and CADD. Results Our results identified FGF2 as a differentially expressed gene with a G101T point mutation in HCC827‐TR cells that showed high mutation frequency and hazard score. HCC827‐TR cells showed elevated FGF2 compared to parental cells. It is noteworthy that treatment with the FGFR inhibitor AZD4547 could restore the sensitivity of HCC872‐TR cells to erlotinib. Conclusions An erlotinib‐resistant cell line HCC827‐TR was successfully constructed and we identified the EGFR‐TKI resistance mechanism involving the FGF2 gene mutation. Targeted inhibition of the FGF2/FGFR signaling pathway may effectively restore the sensitivity of the resistant cells to erlotinib. These results suggest a novel treatment strategy for EGFR‐TKI resistant NSCLC patients. Key points Significant findings of the study: Identifies a novel molecular mechanism for EGFR‐TKI acquired resistance. What this study adds: A potential novel strategy for the treatment of EGFR‐TKI resistant NSCLC patients.
Collapse
Affiliation(s)
- Tao Yu
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Qian Xia
- Tianjin Medical University, Tianjin, China
| | - Ting Gong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Wang
- Department of Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - DianSheng Zhong
- Department of Medical Oncology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Tafe LJ. Non-Small Cell Lung Cancer as a Precision Oncology Paradigm: Emerging Targets and Tumor Mutational Burden (TMB). Adv Anat Pathol 2020; 27:3-10. [PMID: 31567128 DOI: 10.1097/pap.0000000000000244] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Non-small cell lung cancer (NSCLC), since the recognition of epidermal growth factor receptor (EGFR) mutations that sensitized tumors to EGFR tyrosine kinase inhibitors, has been a poster child for precision oncology in solid tumors. The emergence of resistance to the EGFR tyrosine kinase inhibitors led to the unveiling of multiple resistance mechanisms that are now recognized to be frequent mechanisms across multiple tumor types. Coevolution of technological advancements in testing methods available to clinical laboratories now has identified a growing number of molecularly defined subsets of NSCLC that have new therapeutic implications. In addition, identifying patients eligible for immunotherapy is another goal for precision oncology. Recently, studies suggest that TMB may be a promising biomarker for selecting patients with NSCLC for immunotherapy. This review focuses on emerging potentially targetable alterations specifically in RET, ERBB2 (HER2), MET, and KRAS and current evidence and controversies surrounding TMB testing.
Collapse
|
15
|
Combination of Crizotinib and Osimertinib or Erlotinib Might Overcome MET-Mediated Resistance to EGFR Tyrosine Kinase Inhibitor in EGFR-Mutated Adenocarcinoma. J Thorac Oncol 2019; 13:e232-e234. [PMID: 30368417 DOI: 10.1016/j.jtho.2018.07.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 07/10/2018] [Accepted: 07/10/2018] [Indexed: 01/08/2023]
|
16
|
Moosavi F, Giovannetti E, Saso L, Firuzi O. HGF/MET pathway aberrations as diagnostic, prognostic, and predictive biomarkers in human cancers. Crit Rev Clin Lab Sci 2019; 56:533-566. [PMID: 31512514 DOI: 10.1080/10408363.2019.1653821] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Cancer is a major cause of death worldwide. MET tyrosine kinase receptor [MET, c-MET, hepatocyte growth factor (HGF) receptor] pathway activation is associated with the appearance of several hallmarks of cancer. The HGF/MET pathway has emerged as an important actionable target across many solid tumors; therefore, biomarker discovery becomes essential in order to guide clinical intervention and patient stratification with the aim of moving towards personalized medicine. The focus of this review is on how the aberrant activation of the HGF/MET pathway in tumor tissue or the circulation can provide diagnostic and prognostic biomarkers and predictive biomarkers of drug response. Many meta-analyses have shown that aberrant activation of the MET pathway in tumor tissue, including MET gene overexpression, gene amplification, exon 14 skipping and other activating mutations, is almost invariably associated with shorter survival and poor prognosis. Most meta-analyses have been performed in non-small cell lung cancer (NSCLC), breast, head and neck cancers as well as colorectal, gastric, pancreatic and other gastrointestinal cancers. Furthermore, several studies have shown the predictive value of MET biomarkers in the identification of patients who gain the most benefit from HGF/MET targeted therapies administered as single or combination therapies. The highest predictive values have been observed for response to foretinib and savolitinib in renal cancer, as well as tivantinib in NSCLC and colorectal cancer. However, some studies, especially those based on MET expression, have failed to show much value in these stratifications. This may be rooted in lack of standardization of methodologies, in particular in scoring systems applied in immunohistochemistry determinations or absence of oncogenic addiction of cancer cells to the MET pathway, despite detection of overexpression. Measurements of amplification and mutation aberrations are less likely to suffer from these pitfalls. Increased levels of MET soluble ectodomain (sMET) in circulation have also been associated with poor prognosis; however, the evidence is not as strong as it is with tissue-based biomarkers. As a diagnostic biomarker, sMET has shown its value in distinguishing cancer patients from healthy individuals in prostate and bladder cancers and in melanoma. On the other hand, increased circulating HGF has also been presented as a valuable prognostic and diagnostic biomarker in many cancers; however, there is controversy on the predictive value of HGF as a biomarker. Other biomarkers such as circulating tumor DNA (ctDNA) and tumor HGF levels have also been briefly covered. In conclusion, HGF/MET aberrations can provide valuable diagnostic, prognostic and predictive biomarkers and represent vital assets for personalized cancer therapy.
Collapse
Affiliation(s)
- Fatemeh Moosavi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, VU University Medical Center (VUmc) , Amsterdam , The Netherlands.,Cancer Pharmacology Lab, AIRC Start Up Unit, Fondazione Pisana per la Scienza Onlus , Pisa , Italy
| | - Luciano Saso
- Department of Physiology and Pharmacology, "Vittorio Erspamer," Sapienza University , Rome , Italy
| | - Omidreza Firuzi
- Medicinal and Natural Products Chemistry Research Center, Shiraz University of Medical Sciences , Shiraz , Iran
| |
Collapse
|
17
|
Fernandes M, Duplaquet L, Tulasne D. Proteolytic cleavages of MET: the divide-and-conquer strategy of a receptor tyrosine kinase. BMB Rep 2019. [PMID: 30670153 PMCID: PMC6507848 DOI: 10.5483/bmbrep.2019.52.4.024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Membrane-anchored full-length MET stimulated by its ligand HGF/SF induces various biological responses, including survival, growth, and invasion. This panel of responses, referred to invasive growth, is required for embryogenesis and tissue regeneration in adults. On the contrary, MET deregulation is associated with tumorigenesis in many kinds of cancer. In addition to its well-documented ligand-stimulated downstream signaling, the receptor can be cleaved by proteases such as secretases, caspases, and calpains. These cleavages are involved either in MET receptor inactivation or, more interestingly, in generating active fragments that can modify cell fate. For instance, MET fragments can promote cell death or invasion. Given a large number of proteases capable of cleaving MET, this receptor appears as a prototype of proteolytic-cleavage-regulated receptor tyrosine kinase. In this review, we describe and discuss the mechanisms and consequences, both physiological and pathological, of MET proteolytic cleavages.
Collapse
Affiliation(s)
- Marie Fernandes
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| | - Leslie Duplaquet
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| | - David Tulasne
- University of Lille, CNRS, Institut Pasteur de Lille, UMR 8161 - M3T - Mechanisms of Tumorigenesis and Target Therapies, F-59000 Lille, France
| |
Collapse
|
18
|
Panda M, Biswal BK. Cell signaling and cancer: a mechanistic insight into drug resistance. Mol Biol Rep 2019; 46:5645-5659. [PMID: 31280421 DOI: 10.1007/s11033-019-04958-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/27/2019] [Indexed: 12/22/2022]
Abstract
Drug resistance is a major setback for advanced therapeutics in multiple cancers. The increasing prevalence of this resistance is a growing concern and bitter headache for the researchers since a decade. Hence, it is essential to revalidate the existing strategies available for cancer treatment and to look after a novel therapeutic approach for target based killing of cancer cells at the genetic level. This review outlines the different mechanisms enabling resistance including drug efflux, drug target alternation, alternative splicing, the release of the extracellular vesicle, tumor heterogeneity, epithelial-mesenchymal transition, tumor microenvironment, the secondary mutation in the receptor, epigenetic alternation, heterodimerization of receptors, amplification of target and amplification of components rather than the target. Furthermore, existing evidence and the role of various signaling pathways like EGFR, Ras, PI3K/Akt, Wnt, Notch, TGF-β, Integrin-ECM signaling in drug resistance are explained. Lastly, the prevention of this resistance by a contemporary therapeutic strategy, i.e., a combination of specific signaling pathway inhibitors and the cocktail of a cancer drug is summarized showing the new treatment strategies.
Collapse
Affiliation(s)
- Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India
| | - Bijesh K Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology, Sundargarh, Rourkela, Odisha, 769008, India.
| |
Collapse
|
19
|
Hindy JR, Souaid T, Kourie HR, Kattan J. Targeted therapies in urothelial bladder cancer: a disappointing past preceding a bright future? Future Oncol 2019; 15:1505-1524. [PMID: 30977669 DOI: 10.2217/fon-2018-0459] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Bladder cancer (BC) is the most frequent cancer affecting the urinary tract. With the growing era of targeted therapies around the 2000s, many trials evaluated the efficacy of targeted therapy in advanced BC. However, no approval was given yet to any form of targeted therapy when it comes to BC. The aim of this paper was to report the most pivotal trials that evaluated different families of targeted therapy in the treatment of BC, according to their biomarkers (FGFR3, EGFR, HER2, VEGF and PI3K/AKT/mTOR). The ongoing trials testing targeted therapies in advanced BC were then summarized. Finally, the different immunotherapies approved for this disease and their potential combination with targeted therapy were addressed.
Collapse
Affiliation(s)
- Joya-Rita Hindy
- Medical Genetics Unit, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Tarek Souaid
- Medical Genetics Unit, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Hampig Raphael Kourie
- Medical Genetics Unit, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon.,Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| | - Joseph Kattan
- Department of Oncology, Faculty of Medicine, Saint Joseph University, Beirut, Lebanon
| |
Collapse
|
20
|
Lai GGY, Lim TH, Lim J, Liew PJR, Kwang XL, Nahar R, Aung ZW, Takano A, Lee YY, Lau DPX, Tan GS, Tan SH, Tan WL, Ang MK, Toh CK, Tan BS, Devanand A, Too CW, Gogna A, Ong BH, Koh TPT, Kanesvaran R, Ng QS, Jain A, Rajasekaran T, Yuan J, Lim TKH, Lim AST, Hillmer AM, Lim WT, Iyer NG, Tam WL, Zhai W, Tan EH, Tan DSW. Clonal MET Amplification as a Determinant of Tyrosine Kinase Inhibitor Resistance in Epidermal Growth Factor Receptor-Mutant Non-Small-Cell Lung Cancer. J Clin Oncol 2019; 37:876-884. [PMID: 30676858 DOI: 10.1200/jco.18.00177] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
PURPOSE Mesenchymal epithelial transition factor ( MET) activation has been implicated as an oncogenic driver in epidermal growth factor receptor ( EGFR)-mutant non-small-cell lung cancer (NSCLC) and can mediate primary and secondary resistance to EGFR tyrosine kinase inhibitors (TKI). High copy number thresholds have been suggested to enrich for response to MET inhibitors. We examined the clinical relevance of MET copy number gain (CNG) in the setting of treatment-naive metastatic EGFR-mutant-positive NSCLC. PATIENTS AND METHODS MET fluorescence in situ hybridization was performed in 200 consecutive patients identified as metastatic treatment-naïve EGFR-mutant-positive. We defined MET-high as CNG greater than or equal to 5, with an additional criterion of MET/centromeric portion of chromosome 7 ratiο greater than or equal to 2 for amplification. Time-to-treatment failure (TTF) to EGFR TKI in patients identified as MET-high and -low was estimated by Kaplan-Meier method and compared using log-rank test. Multiregion single-nucleotide polymorphism array analysis was performed on 13 early-stage resected EGFR-mutant-positive NSCLC across 59 sectors to investigate intratumoral heterogeneity of MET CNG. RESULTS Fifty-two (26%) of 200 patients in the metastatic cohort were MET-high at diagnosis; 46 (23%) had polysomy and six (3%) had amplification. Median TTF was 12.2 months (95% CI, 5.7 to 22.6 months) versus 13.1 months (95% CI, 10.6 to 15.0 months) for MET-high and -low, respectively ( P = .566), with no significant difference in response rate regardless of copy number thresholds. Loss of MET was observed in three of six patients identified as MET-high who underwent postprogression biopsies, which is consistent with marked intratumoral heterogeneity in MET CNG observed in early-stage tumors. Suboptimal response (TTF, 1.0 to 6.4 months) to EGFR TKI was observed in patients with coexisting MET amplification (five [3.2%] of 154). CONCLUSION Although up to 26% of TKI-naïve EGFR-mutant-positive NSCLC harbor high MET CNG by fluorescence in situ hybridization, this did not significantly affect response to TKI, except in patients identified as MET-amplified. Our data underscore the limitations of adopting arbitrary copy number thresholds and the need for cross-assay validation to define therapeutically tractable MET pathway dysregulation in EGFR-mutant-positive NSCLC.
Collapse
Affiliation(s)
| | | | - John Lim
- 1 National Cancer Centre Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | - Mei-Kim Ang
- 1 National Cancer Centre Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | - Amit Jain
- 1 National Cancer Centre Singapore, Singapore
| | | | - Ju Yuan
- 3 Genome Institute of Singapore, Singapore
| | | | | | | | | | | | | | | | | | - Daniel S W Tan
- 1 National Cancer Centre Singapore, Singapore.,3 Genome Institute of Singapore, Singapore
| |
Collapse
|
21
|
Crizotinib treatment for patients with EGFR mutation positive NSCLC that acquire cMET amplification after EGFR TKI therapy results in short-lived and heterogeneous responses. Lung Cancer 2018; 124:130-134. [DOI: 10.1016/j.lungcan.2018.07.030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 07/05/2018] [Accepted: 07/20/2018] [Indexed: 12/15/2022]
|
22
|
Runcie K, Budman DR, John V, Seetharamu N. Bi-specific and tri-specific antibodies- the next big thing in solid tumor therapeutics. Mol Med 2018; 24:50. [PMID: 30249178 PMCID: PMC6154901 DOI: 10.1186/s10020-018-0051-4] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 09/10/2018] [Indexed: 12/31/2022] Open
Abstract
Antibody-based therapy has revitalized the world of cancer therapeutics since rituximab was first approved for the treatment of Non-Hodgkin's Lymphoma. Monoclonal antibodies against cancer antigens have been successful strategies for only a handful of cancer types due to many reasons including lack of antibody specificity and complex nature of tumor milieu which interfere with antibody efficacy. Polyspecific antibodies are promising class of anti-cancer agents which can be directed at multiple tumor antigens to eradicate tumor cells more precisely and effectively. They may overcome some of these limitations and have already changed treatment landscape for some malignancies such as B cell acute lymphoblastic leukemia. Pre-clinical studies and early phase clinical trials have demonstrated that this approach may be an effective strategy even for solid tumors. This review focuses on the development of bispecific and trispecific antibody therapy for the treatment of solid tumor malignancies and highlights the potential they hold for future therapies to come.
Collapse
Affiliation(s)
- Karie Runcie
- Department of Medicine, Hofstra-Northwell School of Medicine, Hempstead, USA
| | - Daniel R. Budman
- Department of Medicine, Hofstra-Northwell School of Medicine, Hempstead, USA
- Division of Hematology and Medical Oncology, Hofstra-Northwell School of Medicine, Hempstead, USA
| | - Veena John
- Department of Medicine, Hofstra-Northwell School of Medicine, Hempstead, USA
- Division of Hematology and Medical Oncology, Hofstra-Northwell School of Medicine, Hempstead, USA
| | - Nagashree Seetharamu
- Department of Medicine, Hofstra-Northwell School of Medicine, Hempstead, USA
- Division of Hematology and Medical Oncology, Hofstra-Northwell School of Medicine, Hempstead, USA
| |
Collapse
|
23
|
Vanden Bempt I, Wauters E, Vansteenkiste J. Genetic profiling of cell-free DNA from cerebrospinal fluid: opening the barrier to leptomeningeal metastasis in EGFR-mutant NSCLC. Ann Oncol 2018; 29:789-791. [DOI: 10.1093/annonc/mdy053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
24
|
Fumagalli C, Vacirca D, Rappa A, Passaro A, Guarize J, Rafaniello Raviele P, de Marinis F, Spaggiari L, Casadio C, Viale G, Barberis M, Guerini-Rocco E. The long tail of molecular alterations in non-small cell lung cancer: a single-institution experience of next-generation sequencing in clinical molecular diagnostics. J Clin Pathol 2018. [PMID: 29535211 DOI: 10.1136/jclinpath-2018-205032] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND Molecular profiling of advanced non-small cell lung cancers (NSCLC) is essential to identify patients who may benefit from targeted treatments. In the last years, the number of potentially actionable molecular alterations has rapidly increased. Next-generation sequencing allows for the analysis of multiple genes simultaneously. AIMS To evaluate the feasibility and the throughput of next-generation sequencing in clinical molecular diagnostics of advanced NSCLC. METHODS A single-institution cohort of 535 non-squamous NSCLC was profiled using a next-generation sequencing panel targeting 22 actionable and cancer-related genes. RESULTS 441 non-squamous NSCLC (82.4%) harboured at least one gene alteration, including 340 cases (63.6%) with clinically relevant molecular aberrations. Mutations have been detected in all but one gene (FGFR1) of the panel. Recurrent alterations were observed in KRAS, TP53, EGFR, STK11 and MET genes, whereas the remaining genes were mutated in <5% of the cases. Concurrent mutations were detected in 183 tumours (34.2%), mostly impairing KRAS or EGFR in association with TP53 alterations. CONCLUSIONS The study highlights the feasibility of targeted next-generation sequencing in clinical setting. The majority of NSCLC harboured mutations in clinically relevant genes, thus identifying patients who might benefit from different targeted therapies.
Collapse
Affiliation(s)
- Caterina Fumagalli
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Davide Vacirca
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Alessandra Rappa
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Antonio Passaro
- Division of Thoracic Oncology, European Institute of Oncology, Milan, Italy
| | - Juliana Guarize
- Division of Thoracic Surgery, European Institute of Oncology, Milan, Italy
| | | | - Filippo de Marinis
- Division of Thoracic Oncology, European Institute of Oncology, Milan, Italy
| | - Lorenzo Spaggiari
- Division of Thoracic Surgery, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chiara Casadio
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Giuseppe Viale
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Massimo Barberis
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology and Laboratory Medicine, European Institute of Oncology, Milan, Italy.,Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| |
Collapse
|