1
|
Melosky B, Juergens RA, Banerji S, Sacher A, Wheatley-Price P, Snow S, Tsao MS, Leighl NB, Martins I, Cheema P, Liu G, Chu QSC. The continually evolving landscape of novel therapies in oncogene-driven advanced non-small-cell lung cancer. Ther Adv Med Oncol 2025; 17:17588359241308784. [PMID: 39776537 PMCID: PMC11705342 DOI: 10.1177/17588359241308784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-small-cell lung cancer (NSCLC) is a highly heterogeneous disease that is frequently associated with a host of known oncogenic alterations. Advances in molecular diagnostics and drug development have facilitated the targeting of novel alterations such that the majority of NSCLC patients have driver mutations that are now clinically actionable. The goal of this review is to gain insights into clinical research and development principles by summary, analysis, and discussion of data on agents targeting known alterations in oncogene-driven, advanced NSCLC beyond those in the epidermal growth factor receptor (EGFR) and the anaplastic lymphoma kinase (ALK). A search of published and presented literature was conducted to identify prospective trials and integrated analyses reporting outcomes for agents targeting driver gene alterations (except those in EGFR and ALK) in molecularly selected, advanced NSCLC. Clinical efficacy data were extracted from eligible reports and summarized in text and tables. Findings show that research into alteration-directed therapies in oncogene-driven, advanced NSCLC is an extremely active research field. Ongoing research focuses on the expansion of new agents targeting both previously identified targets (particularly hepatocyte growth factor receptor (MET), human epidermal growth factor receptor 2 (HER2), and Kirsten rat sarcoma viral oncogene homolog (KRAS)) as well as novel, potentially actionable targets (such as neuregulin-1 (NRG1) and phosphatidylinositol 3-kinase (PI3K)). The refinement of biomarker selection criteria and the development of more selective and potent agents are allowing for increasingly specific and effective therapies and the expansion of clinically actionable alterations. Clinical advances in this field have resulted in a large number of regulatory approvals over the last 3 years. Future developments should focus on the continued application of alteration therapy matching principles and the exploration of novel ways to target oncogene-driven NSCLC.
Collapse
Affiliation(s)
- Barbara Melosky
- Medical Oncology, BC Cancer Agency—Vancouver, University of British Columbia, 600 West 10th Avenue, Vancouver, BC V5Z 4E6, Canada
| | | | - Shantanu Banerji
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Adrian Sacher
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Paul Wheatley-Price
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Stephanie Snow
- QEII Health Sciences Centre, Dalhousie University, Halifax, NS, Canada
| | - Ming-Sound Tsao
- University Health Network and Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Natasha B. Leighl
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | | | - Parneet Cheema
- William Osler Health System, University of Toronto, Brampton, ON, Canada
| | - Geoffrey Liu
- Princess Margaret Cancer Centre, University of Toronto, Toronto, ON, Canada
| | - Quincy S. C. Chu
- Cross Cancer Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
2
|
Chen X, Wu L, Lan G, Li X, Wang X, Zhang P, Huang W. Construction and validation of a risk prediction model for postoperative lung infection in elderly patients with lung cancer. Medicine (Baltimore) 2024; 103:e40337. [PMID: 39495987 PMCID: PMC11537623 DOI: 10.1097/md.0000000000040337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
This study aimed to analyze the risk factors for postoperative lung infection in elderly patients with lung cancer (LC) and construct a predictive model. A retrospective analysis was conducted on 192 elderly patients with LC who underwent surgical treatment in our hospital between February 2020 and May 2023. According to whether there is lung infection after surgery, they were divided into an infected group (n = 55) and a noninfected group (n = 137). Binary logistic regression was used to analyze factors influencing postoperative lung infection in elderly patients with LC. Based on the logistic regression results, a predictive model for postoperative lung infection in LC patients was constructed. The receiver operating characteristic curve was used to analyze C-reactive protein (CRP), interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), and their combination in predicting postoperative lung infection in patients with LC. There were significant differences between the infected group and the noninfected group in age, smoking history, diabetes, and perioperative antibiotic use were significantly different between the infected and noninfected groups (P < .05). The postoperative CRP, IL-6, and IGF-1 levels in the infected group were higher than those in the noninfected group on the 1st day (P < .05). Logistic regression analysis showed that age > 70 years, history of smoking, history of diabetes, prolonged use of perioperative antibiotics, and elevated CRP, IL-6, and IGF-1 levels on the 1st day after surgery were risk factors for postoperative lung infection in elderly patients with LC (P < .05). Receiver operating characteristic curve analysis showed that the area under curve values of CRP, IL-6, IGF-1, and their combination in predicting postoperative lung infection in elderly patients with LC were 0.701, 0.806, 0.737, and 0.871, P < .05), with sensitivity values of 0.443, 0.987, 0.456, and 0.835, respectively; the specificity was 0.978, 0.525, 0.991, and 0.821, respectively. Age > 70 years, smoking history, diabetes history, prolonged use of perioperative antibiotics, and elevated CRP, IL-6, and IGF-1 levels on the 1st day after surgery have an impact on postoperative lung infection in elderly patients with LC. Early postoperative monitoring of changes in CRP, IL-6, and IGF-1 levels can provide an important reference for predicting the occurrence of postoperative lung infections.
Collapse
Affiliation(s)
- Xiaojie Chen
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Lixin Wu
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Gang Lan
- Department of Chest Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Xiaofeng Li
- Department of Chest Disease Diagnosis and Treatment Center, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Xuejing Wang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Ping Zhang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| | - Weihu Huang
- Department of Pulmonary and Critical Care Medicine, Zhejiang Rongjun Hospital, Jiaxing City, Zhejiang Province, China
| |
Collapse
|
3
|
Harris E, Thawani R. Current perspectives of KRAS in non-small cell lung cancer. Curr Probl Cancer 2024; 51:101106. [PMID: 38879917 DOI: 10.1016/j.currproblcancer.2024.101106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024]
Abstract
NSCLC has a diverse genomic background with mutations in key proto-oncogenic drivers including Kirsten rat sarcoma (KRAS) and epidermal growth factor receptor (EGFR). Roughly 40% of adenocarcinoma harbor Kras activating mutations regardless of smoking history. Most KRAS mutations are located at G12, which include G12C (roughly 40%), G12V (roughly 20%), and G12D (roughly 15%). KRAS mutated NSCLC have higher tumor mutational burden and some have increased PD-1 expression, which has resulted in better responses to immunotherapy than other oncogenes. While initial treatment for metastatic NSCLC still relies on chemo-immunotherapy, directly targeting KRAS has proven to be efficacious in treating patients with KRAS mutated metastatic NSCLC. To date, two G12C inhibitors have been FDA-approved, namely sotorasib and adagrasib. In this review, we summarize the different drug combinations used to target KRAS G12c, upcoming G12D inhibitors and novel therapies targeting KRAS.
Collapse
Affiliation(s)
- Ethan Harris
- Department of Medicine, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637. USA
| | - Rajat Thawani
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Ave, Chicago, IL 60637. USA.
| |
Collapse
|
4
|
Zahmatyar M, Kharaz L, Abiri Jahromi N, Jahanian A, Shokri P, Nejadghaderi SA. The safety and efficacy of binimetinib for lung cancer: a systematic review. BMC Pulm Med 2024; 24:379. [PMID: 39090580 PMCID: PMC11295668 DOI: 10.1186/s12890-024-03178-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Accepted: 07/22/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND Lung cancer, accounting for a significant proportion of global cancer cases and deaths, poses a considerable health burden. Non-small cell lung cancer (NSCLC) patients have a poor prognosis and limited treatment options due to late-stage diagnosis and drug resistance. Dysregulated of the mitogen-activated protein kinase (MAPK) pathway, which is implicated in NSCLC pathogenesis, underscores the potential of MEK inhibitors such as binimetinib. Despite promising results in other cancers, comprehensive studies evaluating the safety and efficacy of binimetinib in lung cancer are lacking. This systematic review aimed to investigate the safety and efficacy of binimetinib for lung cancer treatment. METHODS We searched PubMed, Scopus, Web of Science, and Google Scholar until September 2023. Clinical trials evaluating the efficacy or safety of binimetinib for lung cancer treatment were included. Studies were excluded if they included individuals with conditions unrelated to lung cancer, investigated other treatments, or had different types of designs. The quality assessment was conducted utilizing the National Institutes of Health tool. RESULTS Seven studies with 228 participants overall were included. Four had good quality judgments, and three had fair quality judgments. The majority of patients experienced all-cause adverse events, with diarrhea, fatigue, and nausea being the most commonly reported adverse events of any grade. The objective response rate (ORR) was up to 75%, and the median progression-free survival (PFS) was up to 9.3 months. The disease control rate after 24 weeks varied from 41% to 64%. Overall survival (OS) ranged between 3.0 and 18.8 months. Notably, treatment-related adverse events were observed in more than 50% of patients, including serious adverse events such as colitis, febrile neutropenia, and pulmonary infection. Some adverse events led to dose limitation and drug discontinuation in five studies. Additionally, five studies reported cases of death, mostly due to disease progression. The median duration of treatment ranged from 14.8 weeks to 8.4 months. The most common dosage of binimetinib was 30 mg or 45 mg twice daily, sometimes used in combination with other agents like encorafenib or hydroxychloroquine. CONCLUSIONS Only a few studies have shown binimetinib to be effective, in terms of improving OS, PFS, and ORR, while most of the studies found nonsignificant efficacy with increased toxicity for binimetinib compared with traditional chemotherapy in patients with lung cancer. Further large-scale randomized controlled trials are recommended.
Collapse
Affiliation(s)
- Mahdi Zahmatyar
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ladan Kharaz
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ali Jahanian
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pourya Shokri
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Aria Nejadghaderi
- HIV/STI Surveillance Research Center, WHO Collaborating Center for HIV Surveillance, Institute for Futures Studies in Health, Kerman University of Medical Sciences, Kerman, Iran.
- Systematic Review and Meta‑analysis Expert Group (SRMEG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
5
|
Sahu P, Mitra A, Ganguly A. Targeting KRAS and SHP2 signaling pathways for immunomodulation and improving treatment outcomes in solid tumors. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 386:167-222. [PMID: 38782499 DOI: 10.1016/bs.ircmb.2024.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
Historically, KRAS has been considered 'undruggable' inspite of being one of the most frequently altered oncogenic proteins in solid tumors, primarily due to the paucity of pharmacologically 'druggable' pockets within the mutant isoforms. However, pioneering developments in drug design capable of targeting the mutant KRAS isoforms especially KRASG12C-mutant cancers, have opened the doors for emergence of combination therapies comprising of a plethora of inhibitors targeting different signaling pathways. SHP2 signaling pathway, primarily known for activation of intracellular signaling pathways such as KRAS has come up as a potential target for such combination therapies as it emerged to be the signaling protein connecting KRAS and the immune signaling pathways and providing the link for understanding the overlapping regions of RAS/ERK/MAPK signaling cascade. Thus, SHP2 inhibitors having potent tumoricidal activity as well as role in immunomodulation have generated keen interest in researchers to explore its potential as combination therapy in KRAS mutant solid tumors. However, the excitement with these combination therapies need to overcome challenges thrown up by drug resistance and enhanced toxicity. In this review, we will discuss KRAS and SHP2 signaling pathways and their roles in immunomodulation and regulation of tumor microenvironment and also analyze the positive effects and drawbacks of the different combination therapies targeted at these signaling pathways along with their present and future potential to treat solid tumors.
Collapse
Affiliation(s)
- Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Ankita Mitra
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY, United States
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar, Jharkhand, India.
| |
Collapse
|
6
|
Hu C, Mi W, Li F, Zhu L, Ou Q, Li M, Li T, Ma Y, Zhang Y, Xu Y. Optimizing drug combination and mechanism analysis based on risk pathway crosstalk in pan cancer. Sci Data 2024; 11:74. [PMID: 38228620 PMCID: PMC10791624 DOI: 10.1038/s41597-024-02915-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 01/03/2024] [Indexed: 01/18/2024] Open
Abstract
Combination therapy can greatly improve the efficacy of cancer treatment, so identifying the most effective drug combination and interaction can accelerate the development of combination therapy. Here we developed a computational network biological approach to identify the effective drug which inhibition risk pathway crosstalk of cancer, and then filtrated and optimized the drug combination for cancer treatment. We integrated high-throughput data concerning pan-cancer and drugs to construct miRNA-mediated crosstalk networks among cancer pathways and further construct networks for therapeutic drug. Screening by drug combination method, we obtained 687 optimized drug combinations of 83 first-line anticancer drugs in pan-cancer. Next, we analyzed drug combination mechanism, and confirmed that the targets of cancer-specific crosstalk network in drug combination were closely related to cancer prognosis by survival analysis. Finally, we save all the results to a webpage for query ( http://bio-bigdata.hrbmu.edu.cn/oDrugCP/ ). In conclusion, our study provided an effective method for screening precise drug combinations for various cancer treatments, which may have important scientific significance and clinical application value for tumor treatment.
Collapse
Affiliation(s)
- Congxue Hu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Wanqi Mi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Feng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Lun Zhu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Qi Ou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Maohao Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Tengyue Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yuheng Ma
- Department of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot, 010100, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China
| | - Yingqi Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081, China.
- Department of Pharmacy, Inner Mongolia Medical University, Jinshan Development Zone, Hohhot, 010100, China.
| |
Collapse
|
7
|
Saltos AN, Creelan BC, Tanvetyanon T, Chiappori AA, Antonia SJ, Shafique MR, Ugrenovic-Petrovic M, Sansil S, Neuger A, Ozakinci H, Boyle TA, Kim J, Haura EB, Gray JE. A phase I/IB trial of binimetinib in combination with erlotinib in NSCLC harboring activating KRAS or EGFR mutations. Lung Cancer 2023; 183:107313. [PMID: 37499521 DOI: 10.1016/j.lungcan.2023.107313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/29/2023]
Abstract
BACKGROUND Activating mutations in EGFR or KRAS are highly prevalent in NSCLC, share activation of the MAPK pathway and may be amenable to combination therapy to prevent negative feedback activation. METHODS In this phase 1/1B trial, we tested the combination of binimetinib and erlotinib in patients with advanced NSCLC with at least 1 prior line of treatment (unless with activating EGFR mutation which could be treatment-naïve). A subsequent phase 1B expansion accrued patients with either EGFR- or KRAS-mutation using the recommended phase 2 dose (RP2D) from Phase 1. The primary objective was to evaluate the safety of binimetinib plus erlotinib and establish the RP2D. RESULTS 43 patients enrolled (dose-escalation = 23; expansion = 20). 17 harbored EGFR mutation and 22 had KRAS mutation. The RP2D was erlotinib 100 mg daily and binimetinib 15 mg BID × 5 days/week. Common AEs across all doses included diarrhea (69.8%), rash (44.2%), fatigue (32.6%), and nausea (32.6%), and were primarily grade 1/2. Among KRAS mutant patients, 1 (5%) had confirmed partial response and 8 (36%) achieved stable disease as best overall response. Among EGFR mutant patients, 9 were TKI-naïve with 8 (89%) having partial response, and 8 were TKI-pretreated with no partial responses and 1 (13%) stable disease as best overall response. CONCLUSIONS Binimetinib plus erlotinib demonstrated a manageable safety profile and modest efficacy including one confirmed objective response in a KRAS mutant patient. While clinical utility of this specific combination was limited, these results support development of combinations using novel small molecule inhibitors of RAS, selective EGFR- and other MAPK pathway inhibitors, many of which have improved therapeutic indices. CLINICAL TRIAL REGISTRATION NCT01859026.
Collapse
Affiliation(s)
- Andreas N Saltos
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA.
| | - Ben C Creelan
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Tawee Tanvetyanon
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Alberto A Chiappori
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Scott J Antonia
- Center for Cancer Immunotherapy, Duke Cancer Institute, 20 Duke Medicine Cir., Durham, NC 27710, USA
| | - Michael R Shafique
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | | | - Samer Sansil
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Anthony Neuger
- Cancer Pharmacokinetics & Pharmacodynamics Core, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Hilal Ozakinci
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Theresa A Boyle
- Department of Pathology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jongphil Kim
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Eric B Haura
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| | - Jhanelle E Gray
- Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr., Tampa, FL 33612, USA
| |
Collapse
|
8
|
YÜCE M, GÜMÜŞKAPTAN Ç, ÇON AH, YAZICI F. Conjugated Linoleic Acid strengthens the apoptotic effect of low-dose cisplatin in A549 cells by inducing Bcl-2 downregulation. Prostaglandins Other Lipid Mediat 2023; 166:106731. [PMID: 37001725 DOI: 10.1016/j.prostaglandins.2023.106731] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/22/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023]
Abstract
One of the chemotherapeutic agents widely used in the treatment of non-small cell lung cancer (NSCLC) is cisplatin. However, the resistance of cancer cells to cisplatin and additionally serious side effects from cisplatin limit its use. Conjugated linoleic acid (CLA) has been shown to suppress the development of carcinogenesis in vitro and in vivo studies and has antitumoral activity in many cancers. The study aimed to investigate the potential effect of using cisplatin, the first-line treatment for NSCLC, in combination with CLA to increase its efficacy in low-dose use. MTT cytotoxicity assay was performed to determine the effects of CLA in combination with cisplatin on cell viability of NSCLC cell lines. The apoptotic effect of this combination on NSCLC cell lines and cell cycle distribution was determined by flow cytometry. At the same time, apoptosis and cell cycle-related gene expression levels were determined by Real-Time PCR. Combination treatment of low-dose cisplatin with CLA resulted in a significant decrease in cell viability compared to cisplatin alone, and an increase in the rate of apoptotic cells was observed. While cisplatin caused G1 phase arrest in cancer cells, there was an increase in cell percentages in S and G2 phases after combined application with CLA. In high-dose cisplatin administration, it was observed that the efficiency of the decrease in anti-apoptotic BCL2 expression related to resistance to chemotherapeutic agents was less than that of low-dose cisplatin administration. Combined administration of high-dose cisplatin with CLA significantly recovered BCL2 downregulation.
Collapse
|
9
|
Yang Y, Zhang H, Huang S, Chu Q. KRAS Mutations in Solid Tumors: Characteristics, Current Therapeutic Strategy, and Potential Treatment Exploration. J Clin Med 2023; 12:jcm12020709. [PMID: 36675641 PMCID: PMC9861148 DOI: 10.3390/jcm12020709] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 12/21/2022] [Accepted: 12/22/2022] [Indexed: 01/18/2023] Open
Abstract
Kristen rat sarcoma (KRAS) gene is one of the most common mutated oncogenes in solid tumors. Yet, KRAS inhibitors did not follow suit with the development of targeted therapy, for the structure of KRAS has been considered as being implausible to target for decades. Chemotherapy was the initial recommended therapy for KRAS-mutant cancer patients, which was then replaced by or combined with immunotherapy. KRAS G12C inhibitors became the most recent breakthrough in targeted therapy, with Sotorasib being approved by the Food and Drug Administration (FDA) based on its significant efficacy in multiple clinical studies. However, the subtypes of the KRAS mutations are complex, and the development of inhibitors targeting non-G12C subtypes is still at a relatively early stage. In addition, the monotherapy of KRAS inhibitors has accumulated possible resistance, acquiring the exploration of combination therapies or next-generation KRAS inhibitors. Thus, other non-target, conventional therapies have also been considered as being promising. Here in this review, we went through the characteristics of KRAS mutations in cancer patients, and the prognostic effect that it poses on different therapies and advanced therapeutic strategy, as well as cutting-edge research on the mechanisms of drug resistance, tumor development, and the immune microenvironment.
Collapse
|
10
|
Drugging KRAS: current perspectives and state-of-art review. J Hematol Oncol 2022; 15:152. [PMID: 36284306 PMCID: PMC9597994 DOI: 10.1186/s13045-022-01375-4] [Citation(s) in RCA: 71] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/10/2022] Open
Abstract
After decades of efforts, we have recently made progress into targeting KRAS mutations in several malignancies. Known as the ‘holy grail’ of targeted cancer therapies, KRAS is the most frequently mutated oncogene in human malignancies. Under normal conditions, KRAS shuttles between the GDP-bound ‘off’ state and the GTP-bound ‘on’ state. Mutant KRAS is constitutively activated and leads to persistent downstream signaling and oncogenesis. In 2013, improved understanding of KRAS biology and newer drug designing technologies led to the crucial discovery of a cysteine drug-binding pocket in GDP-bound mutant KRAS G12C protein. Covalent inhibitors that block mutant KRAS G12C were successfully developed and sotorasib was the first KRAS G12C inhibitor to be approved, with several more in the pipeline. Simultaneously, effects of KRAS mutations on tumour microenvironment were also discovered, partly owing to the universal use of immune checkpoint inhibitors. In this review, we discuss the discovery, biology, and function of KRAS in human malignancies. We also discuss the relationship between KRAS mutations and the tumour microenvironment, and therapeutic strategies to target KRAS. Finally, we review the current clinical evidence and ongoing clinical trials of novel agents targeting KRAS and shine light on resistance pathways known so far.
Collapse
|
11
|
Guo Q, Liu L, Chen Z, Fan Y, Zhou Y, Yuan Z, Zhang W. Current treatments for non-small cell lung cancer. Front Oncol 2022; 12:945102. [PMID: 36033435 PMCID: PMC9403713 DOI: 10.3389/fonc.2022.945102] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Despite improved methods of diagnosis and the development of different treatments, mortality from lung cancer remains surprisingly high. Non-small cell lung cancer (NSCLC) accounts for the large majority of lung cancer cases. Therefore, it is important to review current methods of diagnosis and treatments of NSCLC in the clinic and preclinic. In this review, we describe, as a guide for clinicians, current diagnostic methods and therapies (such as chemotherapy, chemoradiotherapy, targeted therapy, antiangiogenic therapy, immunotherapy, and combination therapy) for NSCLC.
Collapse
Affiliation(s)
- Qianqian Guo
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Liwei Liu
- Department of Pharmacy, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zelong Chen
- Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Artificial Intelligence and IoT Smart Medical Engineering Research Center of Henan Province, Zhengzhou, China
| | - Yannan Fan
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
| | - Yang Zhou
- Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s Hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Ziqiao Yuan
- Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wenzhou Zhang, ; Ziqiao Yuan,
| | - Wenzhou Zhang
- Department of Pharmacy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou University, Zhengzhou, China
- *Correspondence: Wenzhou Zhang, ; Ziqiao Yuan,
| |
Collapse
|
12
|
Limagne E, Nuttin L, Thibaudin M, Jacquin E, Aucagne R, Bon M, Revy S, Barnestein R, Ballot E, Truntzer C, Derangère V, Fumet JD, Latour C, Rébé C, Bellaye PS, Kaderbhaï CG, Spill A, Collin B, Callanan MB, Lagrange A, Favier L, Coudert B, Arnould L, Ladoire S, Routy B, Joubert P, Ghiringhelli F. MEK inhibition overcomes chemoimmunotherapy resistance by inducing CXCL10 in cancer cells. Cancer Cell 2022; 40:136-152.e12. [PMID: 35051357 DOI: 10.1016/j.ccell.2021.12.009] [Citation(s) in RCA: 127] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 10/21/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022]
Abstract
Chemotherapy with anti PD-1/PD-L1 antibodies has become the standard of care for patients with metastatic non-small cell lung cancer (mNSCLC). Using lung tumor models, where pemetrexed and cisplatin (PEM/CDDP) chemotherapy remains unable to synergize with immune checkpoint inhibitors (ICIs), we linked the failure of this treatment with its inability to induce CXCL10 expression and CD8+ T cell recruitment. Using drug screening, we showed that combining a MEK inhibitor (MEKi) with PEM/CDDP triggers CXCL10 secretion by cancer cells and CD8+ T cell recruitment, sensitizing it to ICIs. PEM/CDDP plus a MEKi promotes optineurin (OPTN)-dependent mitophagy, resulting in CXCL10 production in a mitochondrial DNA- and TLR9-dependent manner. TLR9 or autophagy/mitophagy inhibition abolishes the anti-tumor efficacy of PEM/CDDP plus MEKi/anti-PD-L1 therapy. In human NSCLCs, high OPTN, TLR9, and CXCL10 expression is associated with a better response to ICIs. Our results underline the role of TLR9- and OPTN-dependent mitophagy in enhancing chemoimmunotherapy efficacy.
Collapse
Affiliation(s)
- Emeric Limagne
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France.
| | - Lisa Nuttin
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Marion Thibaudin
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Elise Jacquin
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; INSERM UMR-S 1193, Université Paris-Saclay, Châtenay-Malabry, France
| | - Romain Aucagne
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; CRISPR Innovative Genomics (CRIGEN) Platform, Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO), Dijon University Hospital, 21000 Dijon, France
| | - Marjorie Bon
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Solène Revy
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Robby Barnestein
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Elise Ballot
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Caroline Truntzer
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Valentin Derangère
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Jean-David Fumet
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Charlène Latour
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Cédric Rébé
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Pierre-Simon Bellaye
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Nuclear Medicine Unit, Preclinical Imagery and Radiotherapy Platform, Centre Georges-François Leclerc, 21000 Dijon, France
| | | | - Aodrenn Spill
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Bertrand Collin
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Nuclear Medicine Unit, Preclinical Imagery and Radiotherapy Platform, Centre Georges-François Leclerc, 21000 Dijon, France; Institut de Chimie Moléculaire de l'Université; de Bourgogne, UMR CNRS 6302, 21000, Dijon, France
| | - Mary B Callanan
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France; CRISPR Innovative Genomics (CRIGEN) Platform, Unit for Innovation in Genetics and Epigenetics in Oncology (IGEO), Dijon University Hospital, 21000 Dijon, France
| | - Aurélie Lagrange
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Laure Favier
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Bruno Coudert
- Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Laurent Arnould
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Unit of Pathology, Department of Biology and Pathology of the Tumors, Centre Georges-François Leclerc, 21000 Dijon, France
| | - Sylvain Ladoire
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France
| | - Bertrand Routy
- University of Montreal Research Center (CRCHUM), Montreal, QC, Canada
| | - Philippe Joubert
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Laval University, Quebec City, QC, Canada
| | - François Ghiringhelli
- University of Bourgogne Franche-Comté, 21000 Dijon, France; Department of Medical Oncology, Centre Georges-François Leclerc, 21000 Dijon, France; Cancer Biology Transfer Platform, Centre Georges-François Leclerc, Equipe Labellisée Ligue Contre le Cancer, 21000 Dijon, France; Centre de Recherche INSERM LNC-UMR1231, 21000 Dijon, France; Genetic and Immunology Medical Institute, Dijon, France.
| |
Collapse
|
13
|
Rodriguez BL, Gibbons DL. MEK inhibition invigorates chemoimmunotherapy by tumor mitophagy-induced CXCL10 expression. Cell Rep Med 2022; 3:100506. [PMID: 35106515 PMCID: PMC8784763 DOI: 10.1016/j.xcrm.2021.100506] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
A recent study by Limagne et al.1 in Cancer Cell demonstrates that addition of MEK inhibitor to standard-of-care platinum/pemetrexed promotes mitophagy-dependent CXCL10 expression via optineurin and TLR9. Tumor cell secretion of CXCL10 produces T cell recruitment and enhances immunotherapy efficacy.
Collapse
Affiliation(s)
- B Leticia Rodriguez
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
14
|
Ferrara MG, Stefani A, Pilotto S, Carbone C, Vita E, Di Salvatore M, D'Argento E, Sparagna I, Monaca F, Valente G, Vitale A, Piro G, Belluomini L, Milella M, Tortora G, Bria E. The Renaissance of KRAS Targeting in Advanced Non-Small-Cell Lung Cancer: New Opportunities Following Old Failures. Front Oncol 2022; 11:792385. [PMID: 35004317 PMCID: PMC8733471 DOI: 10.3389/fonc.2021.792385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/06/2021] [Indexed: 12/14/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) represents the perfect paradigm of ‘precision medicine’ due to its complex intratumoral heterogeneity. It is truly characterized by a range of molecular alterations that can deeply influence the natural history of this disease. Several molecular alterations have been found over time, paving the road to biomarker-driven therapy and radically changing the prognosis of ‘oncogene addicted’ NSCLC patients. Kirsten rat sarcoma (KRAS) mutations are present in up to 30% of NSCLC (especially in adenocarcinoma histotype) and have been identified decades ago. Since its discovery, its molecular characteristics and its marked affinity to a specific substrate have led to define KRAS as an undruggable alteration. Despite that, many attempts have been made to develop drugs capable of targeting KRAS signaling but, until a few years ago, these efforts have been unsuccessful. Comprehensive genomic profiling and wide-spectrum analysis of genetic alterations have only recently allowed to identify different types of KRAS mutations. This tricky step has finally opened new frontiers in the treatment approach of KRAS-mutant patients and might hopefully increase their prognosis and quality of life. In this review, we aim to highlight the most interesting aspects of (epi)genetic KRAS features, hoping to light the way to the state of art of targeting KRAS in NSCLC.
Collapse
Affiliation(s)
- Miriam Grazia Ferrara
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Alessio Stefani
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Sara Pilotto
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Carmine Carbone
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Emanuele Vita
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | | | - Ettore D'Argento
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Ileana Sparagna
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Federico Monaca
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Giustina Valente
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Antonio Vitale
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Geny Piro
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy
| | - Lorenzo Belluomini
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Michele Milella
- Section of Oncology, Department of Medicine, University of Verona School of Medicine and Verona University Hospital Trust, Verona, Italy
| | - Giampaolo Tortora
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| | - Emilio Bria
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Roma, Italy.,Section of Oncology, Department of Translational Medicine, Università Cattolica Del Sacro Cuore, Roma, Italy
| |
Collapse
|