1
|
Briseño-Díaz P, Schnoor M, Bello-Ramirez M, Correa-Basurto J, Rojo-Domínguez A, Arregui L, Vega L, Núñez-González E, Palau-Hernández LA, Parra-Torres CG, García Córdova OM, Zepeda-Castilla E, Torices-Escalante E, Domínguez-Camacho L, Xoconostle-Cazares B, Meraz-Ríos MA, Delfín-Azuara S, Carrión-Estrada DA, Villegas-Sepúlveda N, Hernández-Rivas R, Thompson-Bonilla MDR, Vargas M. Synergistic effect of antagonists to KRas4B/PDE6 molecular complex in pancreatic cancer. Life Sci Alliance 2023; 6:e202302019. [PMID: 37813486 PMCID: PMC10561825 DOI: 10.26508/lsa.202302019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 09/20/2023] [Accepted: 09/25/2023] [Indexed: 10/12/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) has the worst prognosis among all human cancers as it is highly resistant to chemotherapy. K-Ras mutations usually trigger the development and progression of PDAC. We hypothesized that compounds stabilizing the KRas4B/PDE6δ complex could serve as PDAC treatments. Using in silico approaches, we identified the small molecules C14 and P8 that reduced K-Ras activation in primary PDAC cells. Importantly, C14 and P8 significantly prevented tumor growth in patient-derived xenotransplants. Combined treatment with C14 and P8 strongly increased cytotoxicity in PDAC cell lines and primary cultures and showed strong synergistic antineoplastic effects in preclinical murine PDAC models that were superior to conventional therapeutics without causing side effects. Mechanistically, C14 and P8 reduced tumor growth by inhibiting AKT and ERK signaling downstream of K-RAS leading to apoptosis, specifically in PDAC cells. Thus, combined treatment with C14 and P8 may be a superior pharmaceutical strategy to improve the outcome of PDAC.
Collapse
Affiliation(s)
- Paola Briseño-Díaz
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Michael Schnoor
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Martiniano Bello-Ramirez
- Laboratory of Molecular Modeling and Drug Design of the Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - Jose Correa-Basurto
- Laboratory of Molecular Modeling and Drug Design of the Higher School of Medicine, National Polytechnic Institute, Mexico City, Mexico
| | - Arturo Rojo-Domínguez
- Department of Natural Sciences, Metropolitan Autonomous University, Mexico City, Mexico
| | - Leticia Arregui
- Department of Natural Sciences, Metropolitan Autonomous University, Mexico City, Mexico
| | - Libia Vega
- https://ror.org/009eqmr18 Toxicology Department, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City, Mexico
| | - Enrique Núñez-González
- Department of Surgical Oncology and General Surgery, Hospital 1 de Octubre, ISSSTE, Mexico City, Mexico
| | | | | | | | - Ernesto Zepeda-Castilla
- Department of Surgical Oncology and General Surgery, Hospital 1 de Octubre, ISSSTE, Mexico City, Mexico
| | - Eduardo Torices-Escalante
- Department of Surgical Oncology and General Surgery, Hospital 1 de Octubre, ISSSTE, Mexico City, Mexico
| | - Leticia Domínguez-Camacho
- Department of Surgical Oncology and General Surgery, Hospital 1 de Octubre, ISSSTE, Mexico City, Mexico
| | - Beatriz Xoconostle-Cazares
- https://ror.org/009eqmr18 Department of Biotechnology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Marco Antonio Meraz-Ríos
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Sandra Delfín-Azuara
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Dayan Andrea Carrión-Estrada
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Nicolas Villegas-Sepúlveda
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | - Rosaura Hernández-Rivas
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| | | | - Miguel Vargas
- https://ror.org/009eqmr18 Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), México City, Mexico
| |
Collapse
|
2
|
Cura Daball P, Tröger H, Daum S. [Long-term response in advanced pancreatic adenocarcinoma - a case report and literature review]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2022; 60:1510-1516. [PMID: 34905798 DOI: 10.1055/a-1695-3528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Pancreatic cancer is still considered one of the most aggressive types of cancer and is associated with a very poor prognosis although there have been improvements in diagnostics and chemotherapy regimes in recent years. A cure can only be achieved through complete resection which is only possible when diagnosed at a very early stage, though this is rarely the case. We report on a patient with stage IV adenocarcinoma of the pancreas in which several therapeutically actionable mutations could be detected and discuss new options of targeted therapies. CASE REPORT A patient in his 50s was diagnosed with metastatic adenocarcinoma of the pancreas. The patient showed an excellent response to platinum-based chemotherapy with FOLFIRINOX. When a germline mutation in the BRCA-2 gene could be identified, he took part in the POLO-study receiving a maintenance therapy with the PARP-Inhibitor Olaparib. Due to a relapse, 2nd and 3rd line chemotherapy regimens were applied with Gemcitabine combined with Nab-Paclitaxel and later with Erlotinib. Although an activating mutation in the KRAS-gene could be detected as well, the patient rejected further experimental treatment. CONCLUSION Identifying predictive factors and specific targetable mutations in patients with advanced pancreatic cancer is needed to be able to apply more individual and specific therapies in order to improve outcomes.
Collapse
Affiliation(s)
- Paola Cura Daball
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| | - Hanno Tröger
- Medizinische Klinik 1, St Joseph Krankenhaus Berlin-Tempelhof, Berlin, Germany
| | - Severin Daum
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité Universitätsmedizin Berlin Campus Benjamin Franklin, Berlin, Germany
| |
Collapse
|
3
|
Alshememry AK, Alsaleh NB, Alkhudair N, Alzhrani R, Alshamsan A. Recent nanotechnology advancements to treat multidrug-resistance pancreatic cancer: Pre-clinical and clinical overview. Front Pharmacol 2022; 13:933457. [PMID: 36091785 PMCID: PMC9449524 DOI: 10.3389/fphar.2022.933457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/29/2022] [Indexed: 11/13/2022] Open
Abstract
Pancreatic cancer (PC) remains one of the most lethal and incurable forms of cancer and has a poor prognosis. One of the significant therapeutic challenges in PC is multidrug resistance (MDR), a phenomenon in which cancer cells develop resistance toward administered therapy. Development of novel therapeutic platforms that could overcome MDR in PC is crucial for improving therapeutic outcomes. Nanotechnology is emerging as a promising tool to enhance drug efficacy and minimize off-target responses via passive and/or active targeting mechanisms. Over the past decade, tremendous efforts have been made to utilize nanocarriers capable of targeting PC cells while minimizing off-target effects. In this review article, we first give an overview of PC and the major molecular mechanisms of MDR, and then we discuss recent advancements in the development of nanocarriers used to overcome PC drug resistance. In doing so, we explore the developmental stages of this research in both pre-clinical and clinical settings. Lastly, we discuss current challenges and gaps in the literature as well as potential future directions in the field.
Collapse
Affiliation(s)
- Abdullah K. Alshememry
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nasser B. Alsaleh
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Nora Alkhudair
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Rami Alzhrani
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- Nanobiotechnology Unit, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
- *Correspondence: Aws Alshamsan,
| |
Collapse
|
4
|
KRAS-Mutant Non-Small-Cell Lung Cancer: From Past Efforts to Future Challenges. Int J Mol Sci 2022; 23:ijms23169391. [PMID: 36012655 PMCID: PMC9408881 DOI: 10.3390/ijms23169391] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/12/2022] Open
Abstract
KRAS is the most frequently mutated oncogene identified in human cancers. Despite the numerous efforts to develop effective specific inhibitors against KRAS, this molecule has remained "undruggable" for decades. The development of direct KRAS inhibitors, such as sotorasib, the first FDA-approved drug targeting KRAS G12C, or adagrasib, was made possible with the discovery of a small pocket in the binding switch II region of KRAS G12C. However, a new challenge is represented by the necessity to overcome resistance mechanisms to KRAS inhibitors. Another area to be explored is the potential role of co-mutations in the selection of the treatment strategy, particularly in the setting of immune checkpoint inhibitors. The aim of this review was to analyze the state-of-the-art of KRAS mutations in non-small-cell lung cancer by describing the biological structure of KRAS and exploring the clinical relevance of KRAS as a prognostic and predictive biomarker. We reviewed the different treatment approaches, focusing on the novel therapeutic strategies for the treatment of KRAS-mutant lung cancers.
Collapse
|
5
|
Tarannum M, Vivero-Escoto JL. Nanoparticle-based therapeutic strategies targeting major clinical challenges in pancreatic cancer treatment. Adv Drug Deliv Rev 2022; 187:114357. [PMID: 35605679 DOI: 10.1016/j.addr.2022.114357] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Revised: 04/11/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest cancers due to its aggressiveness and the challenges for early diagnosis and treatment. Recently, nanotechnology has demonstrated relevant strategies to overcome some of the major clinical issues in the treatment of PDAC. This review is focused on the pathological hallmarks of PDAC and the impact of nanotechnology to find solutions. It describes the use of nanoparticle-based systems designed for the delivery of chemotherapeutic agents and combinatorial alternatives that address the chemoresistance associated with PDAC, the development of combination therapies targeting the molecular heterogeneity in PDAC, the investigation of novel therapies dealing with the improvement of immunotherapy and handling the desmoplastic stroma in PDAC by remodeling the tumor microenvironment. A special section is dedicated to the design of nanoparticles for unique non-traditional modalities that could be promising in the future for the improvement in the dismal prognosis of PDAC.
Collapse
|
6
|
Xu J, Pu Y, Lin R, Xiao S, Fu Y, Wang T. PEAC: An Ultrasensitive and Cost-Effective MRD Detection System in Non-small Cell Lung Cancer Using Plasma Specimen. Front Med (Lausanne) 2022; 9:822200. [PMID: 35308511 PMCID: PMC8928926 DOI: 10.3389/fmed.2022.822200] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/14/2022] [Indexed: 12/25/2022] Open
Abstract
Circulating tumor DNA (ctDNA), a tumor-derived fraction of cell-free DNA (cfDNA), has emerged as a promising marker in targeted therapy, immunotherapy, and minimal residual disease (MRD) monitoring in postsurgical patients. However, ctDNA level in early-stage cancers and postsurgical patients is very low, which posed many technical challenges to improve the detection rate and sensitivity, especially in the clinical practice of MRD detection. These challenges usually include insufficient DNA input amount, limit of detection (LOD), and high experimental costs. To resolve these challenges, we developed an ultrasensitive ctDNA MRD detection system in this study, namely PErsonalized Analysis of Cancer (PEAC), to simultaneously detect up to 37 mutations, which account for 70–80% non-small cell lung cancer (NSCLC) driver mutations from low plasma sample volume and enables LOD of 0.01% at a single-site level. We demonstrated the high performance achieved by PEAC on both cfDNA reference standards and clinical plasma samples from three NSCLC patient cohorts. For cfDNA reference standards, PEAC achieved a specificity of 99% and a sensitivity of 87% for the mutations at 0.01% allele fraction. In the second cohort, PEAC showed 100% concordance rate between ddPCR and Next-generation sequencing (NGS) among 29 samples. In the third cohort, 22 of 59 patients received EGFR TKI treatment. Among them, three in four patients identified low level actionable gene mutations only by PEAC had partial responses after targeted therapy, demonstrating high ctDNA detection ability of PEAC. Overall, the developed PEAC system can detect the majority of NSCLC driver mutations using 8–10 ml plasma samples, and has the advantages of high detection sensitivity and lower costs compared with the existing technologies such as ddPCR and NGS. These advantages make the PEAC system quite appropriate for ctDNA and MRD detection in early-stage NSCLC and postsurgical recurrence monitoring.
Collapse
Affiliation(s)
- Jianping Xu
- Department of Medical Oncology, Cancer Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Peking Union Medical College, Beijing, China
- *Correspondence: Jianping Xu
| | - Yue Pu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Rui Lin
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Shanshan Xiao
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Yingxue Fu
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
| | - Tao Wang
- Department of Research and Development, Hangzhou Repugene Technology Co., Ltd., Hangzhou, China
- Tao Wang
| |
Collapse
|
7
|
Vaclova T, Chakraborty A, Sherwood J, Ross S, Carroll D, Barrett JC, Downward J, de Bruin EC. Concomitant KRAS mutations attenuate sensitivity of non-small cell lung cancer cells to KRAS G12C inhibition. Sci Rep 2022; 12:2699. [PMID: 35177674 PMCID: PMC8854729 DOI: 10.1038/s41598-022-06369-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/17/2022] [Indexed: 12/17/2022] Open
Abstract
The development of covalent inhibitors against KRAS G12C represents a major milestone in treatment of RAS-driven cancers, especially in non-small cell lung cancer (NSCLC), where KRAS G12C is one of the most common oncogenic driver. Here we investigated if additional KRAS mutations co-occur with KRAS G12C (c.34G>T) in NSCLC tumours and if such mutation co-occurrence affects cellular response to G12C-specific inhibitors. Analysis of a large cohort of NSCLC patients whose tumours harboured KRAS mutations revealed co-occurring KRAS mutations in up to 8% of tumours with the KRAS c.34G>T mutation. KRAS c.35G>T was the most frequently co-occurring mutation, and could occur on the same allele (in cis) translating to a single mutant KRAS G12F protein, or on the other allele (in trans), translating to separate G12C and G12V mutant proteins. Introducing KRAS c.35G>T in trans in the KRAS G12C lung cancer model NCI-H358, as well as the co-occurrence in cis in the KRAS G12F lung cancer model NCI-H2291 led to cellular resistance to the G12C-specific inhibitor AZ’8037 due to continuing active MAPK and PI3K cascades in the presence of the inhibitor. Overall, our study provides a comprehensive assessment of co-occurring KRAS mutations in NSCLC and in vitro evidence of the negative impact of co-occurring KRAS mutations on cellular response to G12C inhibitors, highlighting the need for a comprehensive KRAS tumour genotyping for optimal patient selection for treatment with a KRAS G12C inhibitor.
Collapse
Affiliation(s)
- Tereza Vaclova
- Translational Medicine, Oncology, AstraZeneca, Cambridge, CB4 0WG, UK
| | | | - James Sherwood
- Precision Medicine and Biosamples, BioPharmaceutical, AstraZeneca, Cambridge, CB4 0WG, UK
| | - Sarah Ross
- Bioscience, Oncology, AstraZeneca, Cambridge, CB2 0RE, UK
| | - Danielle Carroll
- Translational Medicine, Oncology, AstraZeneca, Cambridge, CB4 0WG, UK
| | - J Carl Barrett
- Translational Medicine, Oncology, AstraZeneca, Waltham, MA, 02451, USA
| | - Julian Downward
- Oncogene Biology, Francis Crick Institute, London, NW1 1AT, UK
| | - Elza C de Bruin
- Translational Medicine, Oncology, AstraZeneca, Cambridge, CB4 0WG, UK.
| |
Collapse
|
8
|
Molina-Arcas M, Samani A, Downward J. Drugging the Undruggable: Advances on RAS Targeting in Cancer. Genes (Basel) 2021; 12:899. [PMID: 34200676 PMCID: PMC8228461 DOI: 10.3390/genes12060899] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
Around 20% of all malignancies harbour activating mutations in RAS isoforms. Despite this, there is a deficiency of RAS-targeting agents licensed for therapeutic use. The picomolar affinity of RAS for GTP, and the lack of suitable pockets for high-affinity small-molecule binding, precluded effective therapies despite decades of research. Recently, characterisation of the biochemical properties of KRAS-G12C along with discovery of its 'switch-II pocket' have allowed development of effective mutant-specific inhibitors. Currently seven KRAS-G12C inhibitors are in clinical trials and sotorasib has become the first one to be granted FDA approval. Here, we discuss historical efforts to target RAS directly and approaches to target RAS effector signalling, including combinations that overcome limitations of single-agent targeting. We also review pre-clinical and clinical evidence for the efficacy of KRAS-G12C inhibitor monotherapy followed by an illustration of combination therapies designed to overcome primary resistance and extend durability of response. Finally, we briefly discuss novel approaches to targeting non-G12C mutant isoforms.
Collapse
Affiliation(s)
| | - Amit Samani
- Oncogene Biology Laboratory, Francis Crick Institute, London NW1 1AT, UK;
- Department of Medical Oncology, Imperial College Healthcare NHS Trust, London W2 1NY, UK
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick Institute, London NW1 1AT, UK;
- Lung Cancer Group, Institute of Cancer Research, London SW3 6JB, UK
| |
Collapse
|
9
|
Alzhrani R, Alsaab HO, Vanamal K, Bhise K, Tatiparti K, Barari A, Sau S, Iyer AK. Overcoming the Tumor Microenvironmental Barriers of Pancreatic Ductal Adenocarcinomas for Achieving Better Treatment Outcomes. ADVANCED THERAPEUTICS 2021; 4:2000262. [PMID: 34212073 PMCID: PMC8240487 DOI: 10.1002/adtp.202000262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Indexed: 02/06/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly aggressive disease with the lowest survival rate among all solid tumors. The lethality of PDAC arises from late detection and propensity of the tumor to metastasize and develop resistance against chemo and radiation therapy. A highly complex tumor microenvironment composed of dense stroma, immune cells, fibroblast, and disorganized blood vessels, is the main obstacle to current PDAC therapy. Despite the tremendous success of immune checkpoint inhibitors (ICIs) in cancers, PDAC remains one of the poorest responders of ICIs therapy. The immunologically "cold" phenotype of PDAC is attributed to the low mutational burden, high infiltration of myeloid-derived suppressor cells and T-regs, contributing to a significant immunotherapy resistance mechanism. Thus, the development of innovative strategies for turning immunologically "cold" tumor into "hot" ones is an unmet need to improve the outcome of PDAC ICIs therapies. Other smart strategies, such as nanomedicines, sonic Hedgehog inhibitor, or smoothened inhibitor, are discussed to enhance chemotherapeutic agents' efficiency by disrupting the PDAC stroma. This review highlights the current challenges and various preclinical and clinical strategies to overcome current PDAC therapy difficulties, thus significantly advancing PDAC research knowledge.
Collapse
Affiliation(s)
- Rami Alzhrani
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Hashem O. Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Taif University, Taif 21944, Saudi Arabia
| | - Kushal Vanamal
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
| | - Ketki Bhise
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
| | - Katyayani Tatiparti
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
| | - Ayatakshi Barari
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
| | - Samaresh Sau
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
| | - Arun K. Iyer
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit 48201, United States
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University, School of Medicine, Detroit, MI, United States
| |
Collapse
|
10
|
Noordhof AL, Damhuis RAM, Hendriks LEL, de Langen AJ, Timens W, Venmans BJW, van Geffen WH. Prognostic impact of KRAS mutation status for patients with stage IV adenocarcinoma of the lung treated with first-line pembrolizumab monotherapy. Lung Cancer 2021; 155:163-169. [PMID: 33838467 DOI: 10.1016/j.lungcan.2021.04.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/29/2021] [Accepted: 04/01/2021] [Indexed: 12/23/2022]
Abstract
OBJECTIVES Monotherapy with pembrolizumab is the preferred first-line treatment for metastatic non-small cell lung cancer with programmed death-ligand 1 (PD-L1) expression ≥50 %, without targetable oncogenic drivers. Although targeted therapies are in development for patients with specific Kirsten rat sarcoma (KRAS) mutations, these are not available in daily care yet. It is not clear whether there is a difference in survival on first-line pembrolizumab for patients with a high PD-L1 status with or without a KRAS mutation. We aim to compare this survival based on real-world data. MATERIALS AND METHODS This is a real-world retrospective population-based study using data from the Netherlands Cancer Registry. We selected patients with stage IV lung adenocarcinoma with PD-L1 expression ≥50 % diagnosed between January 2017 and December 2018, treated with first-line pembrolizumab. Patients with EGFR mutations, ALK translocations or ROS1 rearrangements were excluded. The primary outcome parameter was overall survival. RESULTS 388 (57 %) of 595 patients had a KRAS mutation. KRAS was seen more frequently in women than in men (65 % versus 49 % respectively, p < 0.001). The median overall survival was 19.2 months versus 16.8 months for patients with and without KRAS mutation, respectively (p = 0.86). Multivariable analysis revealed WHO performance score, number of organs with metastases and PD-L1 percentage as independent prognostic factors. KRAS mutation status had no prognostic influence (hazard ratio = 1.03, 95 % CI 0.83-1.29). CONCLUSION The survival of KRAS mutated versus KRAS wild-type lung adenocarcinoma patients, treated with first-line pembrolizumab monotherapy, is similar, suggesting that KRAS has no prognostic value with respect to treatment with pembrolizumab.
Collapse
Affiliation(s)
- A L Noordhof
- Department of Respiratory Medicine, Medical Center Leeuwarden, Henri Dunantweg 2, 8934 AD, Leeuwarden, the Netherlands
| | - R A M Damhuis
- Department of Research, Comprehensive Cancer Organization, Plesmanlaan 121, 1066 CX, Utrecht, the Netherlands
| | - L E L Hendriks
- Department of Respiratory Medicine, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX, Maastricht, the Netherlands
| | - A J de Langen
- Department of Thoracic Oncology, Netherlands Cancer Institute, NA 1007 MB, Amsterdam, the Netherlands
| | - W Timens
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, 9713 GZ, Groningen, the Netherlands
| | - B J W Venmans
- Department of Respiratory Medicine, Medical Center Leeuwarden, Henri Dunantweg 2, 8934 AD, Leeuwarden, the Netherlands
| | - W H van Geffen
- Department of Respiratory Medicine, Medical Center Leeuwarden, Henri Dunantweg 2, 8934 AD, Leeuwarden, the Netherlands.
| |
Collapse
|
11
|
Madhyastha N, Samantha SK, Dittakavi S, Markose M, Mallurwar SR, Zainuddin M, Mullangi R. Validated HPLC‐MS/MS method for quantitation of AMG 510, a KRAS G12C inhibitor, in mouse plasma and its application to a pharmacokinetic study in mice. Biomed Chromatogr 2021; 35:e5043. [DOI: 10.1002/bmc.5043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/28/2020] [Accepted: 11/30/2020] [Indexed: 11/08/2022]
Affiliation(s)
| | | | | | - Meenu Markose
- Drug Metabolism and Pharmacokinetics Jubilant Biosys Ltd Bangalore India
| | | | - Mohd Zainuddin
- Drug Metabolism and Pharmacokinetics Jubilant Biosys Ltd Bangalore India
| | - Ramesh Mullangi
- Drug Metabolism and Pharmacokinetics Jubilant Biosys Ltd Bangalore India
| |
Collapse
|
12
|
Abstract
Lung cancer is the leading cause of cancer mortality. It is classified into different histologic subtypes, including adenocarcinoma, squamous carcinoma, and large cell carcinoma (commonly referred as non-small cell lung cancer) and small cell lung cancer. Comprehensive molecular characterization of lung cancer has expanded our understanding of the cellular origins and molecular pathways affected in each of these subtypes. Many of these genetic alterations represent potential therapeutic targets for which drugs are constantly under development. This article discusses the molecular characteristics of the main lung cancer subtypes and discusses the current guidelines and novel targeted therapies, including checkpoint immunotherapy.
Collapse
Affiliation(s)
- Roberto Ruiz-Cordero
- Department of Pathology, University of California San Francisco, 1825 4th Street Room L2181A, San Francisco, CA 94158, USA.
| | - Walter Patrick Devine
- Department of Pathology, University of California San Francisco, 1600 Divisadero Street Room B-620, San Francisco, CA 94115, USA
| |
Collapse
|
13
|
Kim D, Xue JY, Lito P. Targeting KRAS(G12C): From Inhibitory Mechanism to Modulation of Antitumor Effects in Patients. Cell 2020; 183:850-859. [PMID: 33065029 PMCID: PMC7669705 DOI: 10.1016/j.cell.2020.09.044] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/24/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023]
Abstract
KRAS mutations are among the most common genetic alterations in lung, colorectal, and pancreatic cancers. Direct inhibition of KRAS oncoproteins has been a long-standing pursuit in precision oncology, one established shortly after the discovery of RAS mutations in human cancer cells nearly 40 years ago. Recent advances in medicinal chemistry have established inhibitors targeting KRAS(G12C), a mutation found in ∼13% of lung adenocarcinomas and, at a lower frequency, in other cancers. Preclinical studies describing their discovery and mechanism of action, coupled with emerging clinical data from patients treated with these drugs, have sparked a renewed enthusiasm in the study of KRAS and its therapeutic potential. Here, we discuss how these advances are reshaping the fundamental aspects of KRAS oncoprotein biology and the strides being made toward improving patient outcomes in the clinic.
Collapse
Affiliation(s)
- Dongsung Kim
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Jenny Yaohua Xue
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA
| | - Piro Lito
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer, New York, NY, USA; Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program, New York, NY, USA; Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
14
|
Giustini NP, Jeong AR, Buturla J, Bazhenova L. Advances in Treatment of Locally Advanced or Metastatic Non-Small Cell Lung Cancer: Targeted Therapy. Clin Chest Med 2020; 41:223-235. [PMID: 32402358 DOI: 10.1016/j.ccm.2020.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The treatment of metastatic non-small cell lung cancer (NSCLC) is constantly evolving. Although the advent of immunotherapy has played an important role in the treatment of patients with NSCLC, the identification of driver mutations and the subsequent specific treatment of these targets often lead to durable responses while maintaining quality of life. This review delves into targeted therapies available for epidermal growth factor receptor, anaplastic lymphoma kinase, ROS1, neurotrophic tropomyosin receptor kinase, and BRAF- mutated NSCLC patients, as well as other mutations with promising novel drugs under clinical investigation.
Collapse
Affiliation(s)
- Nicholas P Giustini
- UCSD Moores Cancer Center, 3855 Health Sciences Drive MC #0987, La Jolla, CA 92093-0829, USA.
| | - Ah-Reum Jeong
- UCSD Moores Cancer Center, 3855 Health Sciences Drive MC #0987, La Jolla, CA 92093-0829, USA
| | - James Buturla
- UCSD Moores Cancer Center, 3855 Health Sciences Drive MC #0987, La Jolla, CA 92093-0829, USA
| | - Lyudmila Bazhenova
- UCSD Moores Cancer Center, 3855 Health Sciences Drive MC #0987, La Jolla, CA 92093-0829, USA
| |
Collapse
|
15
|
Abstract
Aberrations in rat sarcoma (RAS) viral oncogene are the most prevalent and best-known genetic alterations identified in human cancers. Indeed, RAS drives tumorigenesis as one of the downstream effectors of EGFR activation, regulating cellular switches and functions and triggering intracellular signaling cascades such as the MAPK and PI3K pathways. Of the three RAS isoforms expressed in human cells, all of which were linked to tumorigenesis more than three decades ago, KRAS is the most frequently mutated. In particular, point mutations in KRAS codon 12 are present in up to 80% of KRAS-mutant malignancies. Unfortunately, there are no approved KRAS-targeted agents, despite decades of research and development. Recently, a revolutionary strategy to use covalent allosteric inhibitors that target a shallow pocket on the KRAS surface has provided new impetus for renewed drug development efforts, specifically against KRASG12C. These inhibitors, such as AMG 510 and MRTX849, show promise in early-phase studies. Nevertheless, combination strategies that target resistance mechanisms have become vital in the war against KRAS-mutant tumors.
Collapse
Affiliation(s)
- Kyaw Z Thein
- Division of Hematology and Medical Oncology, Oregon Health and Science University/Knight Cancer Institute, Portland, Oregon 97239, USA;
| | - Amadeo B Biter
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; ,
| | - David S Hong
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA; ,
| |
Collapse
|
16
|
Upadhya A, Yadav KS, Misra A. Targeted drug therapy in non-small cell lung cancer: Clinical significance and possible solutions-Part I. Expert Opin Drug Deliv 2020; 18:73-102. [PMID: 32954834 DOI: 10.1080/17425247.2021.1825377] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Non-small cell lung cancer (NSCLC) comprises of 84% of all lung cancer cases. The treatment options for NSCLC at advanced stages are chemotherapy and radiotherapy. Chemotherapy involves conventional nonspecific chemotherapeutics, and targeted-protein/receptor-specific small molecule inhibitors. Biologically targeted therapies such as an antibody-based immunotherapy have been approved in combination with conventional therapeutics. Approved targeted chemotherapy is directed against the kinase domains of mutated cellular receptors such as epidermal growth factor receptor (EGFR), anaplastic lymphoma kinases (ALK), neurotrophic receptor kinases (NTRK) and against downstream signaling molecules such as BRAF (v-raf murine sarcoma viral oncogene homolog B1). Approved biologically targeted therapy involves the use of anti-angiogenesis antibodies and antibodies against immune checkpoints. AREAS COVERED The rationale for the employment of targeted therapeutics and the resistance that may develop to therapy are discussed. Novel targeted therapeutics in clinical trials are also included. EXPERT OPINION Molecular and histological profiling of a given tumor specimen to determine the aberrant onco-driver is a must before deciding a targeted therapeutic regimen for the patient. Periodic monitoring of the patients response to a given therapeutic regimen is also mandatory so that any semblance of resistance to therapy can be deciphered and the regimen may be accordingly altered.
Collapse
Affiliation(s)
- Archana Upadhya
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Khushwant S Yadav
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| | - Ambikanandan Misra
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS , Mumbai, Maharashtra, India
| |
Collapse
|
17
|
Wang Y, Lakoma A, Zogopoulos G. Building towards Precision Oncology for Pancreatic Cancer: Real-World Challenges and Opportunities. Genes (Basel) 2020; 11:E1098. [PMID: 32967105 PMCID: PMC7563487 DOI: 10.3390/genes11091098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 02/06/2023] Open
Abstract
The advent of next-generation sequencing (NGS) has provided unprecedented insight into the molecular complexity of pancreatic ductal adenocarcinoma (PDAC). This has led to the emergence of biomarker-driven treatment paradigms that challenge empiric treatment approaches. However, the growth of sequencing technologies is outpacing the development of the infrastructure required to implement precision oncology as routine clinical practice. Addressing these logistical barriers is imperative to maximize the clinical impact of molecular profiling initiatives. In this review, we examine the evolution of precision oncology in PDAC, spanning from germline testing for cancer susceptibility genes to multi-omic tumor profiling. Furthermore, we highlight real-world challenges to delivering precision oncology for PDAC, and propose strategies to improve the generation, interpretation, and clinical translation of molecular profiling data.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Surgery, McGill University, Montreal, QC H4A 3J1, Canada; (Y.W.); (A.L.)
- Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - Anna Lakoma
- Department of Surgery, McGill University, Montreal, QC H4A 3J1, Canada; (Y.W.); (A.L.)
- Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| | - George Zogopoulos
- Department of Surgery, McGill University, Montreal, QC H4A 3J1, Canada; (Y.W.); (A.L.)
- Research Institute of the McGill University Health Centre, McGill University, Montreal, QC H4A 3J1, Canada
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada
| |
Collapse
|
18
|
Uprety D, Adjei AA. KRAS: From undruggable to a druggable Cancer Target. Cancer Treat Rev 2020; 89:102070. [DOI: 10.1016/j.ctrv.2020.102070] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/04/2020] [Accepted: 07/06/2020] [Indexed: 02/07/2023]
|
19
|
Kattan WE, Hancock JF. RAS Function in cancer cells: translating membrane biology and biochemistry into new therapeutics. Biochem J 2020; 477:2893-2919. [PMID: 32797215 PMCID: PMC7891675 DOI: 10.1042/bcj20190839] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 02/07/2023]
Abstract
The three human RAS proteins are mutated and constitutively activated in ∼20% of cancers leading to cell growth and proliferation. For the past three decades, many attempts have been made to inhibit these proteins with little success. Recently; however, multiple methods have emerged to inhibit KRAS, the most prevalently mutated isoform. These methods and the underlying biology will be discussed in this review with a special focus on KRAS-plasma membrane interactions.
Collapse
Affiliation(s)
- Walaa E. Kattan
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| | - John F. Hancock
- Department of Integrative Biology and Pharmacology, McGovern Medical School University of Texas Health Science Center at Houston, TX 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, TX 77030, USA
| |
Collapse
|
20
|
Yang SR, Schultheis AM, Yu H, Mandelker D, Ladanyi M, Büttner R. Precision medicine in non-small cell lung cancer: Current applications and future directions. Semin Cancer Biol 2020; 84:184-198. [PMID: 32730814 DOI: 10.1016/j.semcancer.2020.07.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/24/2020] [Accepted: 07/13/2020] [Indexed: 12/24/2022]
Abstract
Advances in biomarkers, targeted therapies, and immuno-oncology have transformed the clinical management of patients with advanced NSCLC. For oncogene-driven tumors, there are highly effective targeted therapies against EGFR, ALK, ROS1, BRAF, TRK, RET, and MET. In addition, investigational therapies for KRAS, NRG1, and HER2 have shown promising results and may become standard-of-care in the near future. In parallel, immune-checkpoint therapy has emerged as an indispensable treatment modality, especially for patients lacking actionable oncogenic drivers. While PD-L1 expression has shown modest predictive utility, biomarkers for immune-checkpoint inhibition in NSCLC have remained elusive and represent an area of active investigation. Given the growing importance of biomarkers, optimal utilization of small tissue biopsies and alternative genotyping methods using circulating cell-free DNA have become increasingly integrated into clinical practice. In this review, we will summarize the current landscape and emerging trends in precision medicine for patients with advanced NSCLC with a special focus on predictive biomarker testing.
Collapse
Affiliation(s)
- Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | | | - Helena Yu
- Memorial Sloan Kettering Cancer Center, Department of Medicine, United States
| | - Diana Mandelker
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Marc Ladanyi
- Memorial Sloan Kettering Cancer Center, Department of Pathology, United States
| | - Reinhard Büttner
- University Hospital of Cologne, Department of Pathology, Germany.
| |
Collapse
|
21
|
|
22
|
Cui W, Franchini F, Alexander M, Officer A, Wong HL, IJzerman M, Desai J, Solomon BJ. Real world outcomes in KRAS G12C mutation positive non-small cell lung cancer. Lung Cancer 2020; 146:310-317. [PMID: 32619782 DOI: 10.1016/j.lungcan.2020.06.030] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/21/2020] [Accepted: 06/24/2020] [Indexed: 01/06/2023]
Abstract
BACKGROUND KRAS mutations are found in 20-30 % of non-small cell lung cancers (NSCLC) and were traditionally considered undruggable. KRASG12C mutation confers sensitivity to KRASG12C covalent inhibitors, however its prognostic impact remains unclear. This study assesses the frequency, clinical features, prevalence of brain metastases and outcomes in KRASG12C NSCLC in a real-world setting. METHODS Patients enrolled in the prospective Thoracic Malignancies Cohort (TMC) between July 2012 to October 2019 with recurrent/metastatic non-squamous NSCLC, available KRAS results, and without EGFR/ALK/ROS1 gene aberrations, were selected. Data was extracted from TMC and patient records. Clinicopathologic features, treatment and overall survival (OS) was compared for KRAS wildtype (KRASWT) and KRAS mutated (KRASmut); and KRASG12C and other (KRASother) mutations. RESULTS Of 1386 NSCLC patients, 1040 were excluded: non-metastatic/recurrent (526); unknown KRAS status (356); ALK/EGFR/ROS1 positive (154); duplicate (4). Of 346 patients analysed, 144 (42 %) were KRASmut, of whom 65 (45 %) were KRASG12C. All patients with KRASG12C were active or ex-smokers, compared to 92 % of KRASother and 83 % of KRASWT. The prevalence of brain metastases during follow-up was similar between KRASmut and KRASWT (33 % vs 40 %, p = 0.17), and KRASG12C and KRASother (40 % vs 41 %, p = 0.74). The proportion of patients receiving one or multiple lines of systemic therapy was comparable. OS was similar between KRASmut and KRASWT (p = 0.54), and KRASG12C and KRASother (p = 0.39). CONCLUSION Patients with KRASmut and KRASWT, and KRASG12C and KRASother NSCLC have comparable clinical features, treatment and survival. While not prognostic, KRASG12C may be an important predictive biomarker as promising KRASG12C covalent inhibitors continue to be developed.
Collapse
Affiliation(s)
- Wanyuan Cui
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia.
| | - Fanny Franchini
- University of Melbourne, Centre for Health Policy and Centre for Cancer Research, Melbourne, VIC, 3000, Australia
| | - Marliese Alexander
- Pharmacy Department, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Ann Officer
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Hui-Li Wong
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Parkville, VIC, 3052, Australia
| | - Maarten IJzerman
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Pharmacy Department, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia
| | - Jayesh Desai
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Benjamin J Solomon
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, 3000, Australia; Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, 3010, Australia
| |
Collapse
|
23
|
Corsini EM, Ripley RT. Commentary: KRAS-mutant lung adenocarcinomas-a work in progress. J Thorac Cardiovasc Surg 2020; 163:e87-e88. [PMID: 32771234 DOI: 10.1016/j.jtcvs.2020.06.037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 06/08/2020] [Accepted: 06/09/2020] [Indexed: 01/15/2023]
Affiliation(s)
- Erin M Corsini
- Division of General Thoracic Surgery, The Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - R Taylor Ripley
- Division of General Thoracic Surgery, The Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
24
|
Optimal Management of Patients with Advanced NSCLC Harboring High PD-L1 Expression and Driver Mutations. Curr Treat Options Oncol 2020; 21:60. [PMID: 32588244 DOI: 10.1007/s11864-020-00750-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
OPINION STATEMENT Patients with stage IV or recurrent/metastatic non-small cell lung cancer (NSCLC) whose tumors harbor high PD-L1 expression and driver mutations with approved targeted treatments (EGFR, ALK, BRAFV600E, ROS1) should receive initial therapy with targeted therapy based on impressive clinical activity. PD-(L)1 inhibitors have demonstrated minimal activity in many driver mutation subsets including EGFR and ALK and appears to have more benefit in smoking-associated oncogenic drivers (KRAS, BRAF). For KRAS-driven tumors, co-mutations such as STK11/LKB1 are negative predictive markers of immunotherapy with or without chemotherapy. Therefore, driver mutations need to be evaluated before pursuing immunotherapy independent of PD-L1 expression level. Caution should be used with TKIs following or concurrent with immunotherapy owing to potentially increased toxicity. New immunotherapy combinations are needed especially for oncogene-driven tumors associated with never or light smoking history.
Collapse
|
25
|
Singh N. Lung cancer in India: The rewards of REWERDS (REal World Evidence from Retrospective Data analysiS). Curr Probl Cancer 2020; 44:100591. [PMID: 32505367 DOI: 10.1016/j.currproblcancer.2020.100591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 03/25/2020] [Indexed: 12/15/2022]
Affiliation(s)
- Navneet Singh
- Department of Pulmonary Medicine, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| |
Collapse
|
26
|
Chen R, Manochakian R, James L, Azzouqa AG, Shi H, Zhang Y, Zhao Y, Zhou K, Lou Y. Emerging therapeutic agents for advanced non-small cell lung cancer. J Hematol Oncol 2020; 13:58. [PMID: 32448366 PMCID: PMC7245927 DOI: 10.1186/s13045-020-00881-7] [Citation(s) in RCA: 170] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/23/2020] [Indexed: 01/06/2023] Open
Abstract
Advanced non-small cell lung cancer (NSCLC) is the most common type of lung cancer, with a poor prognosis and no known cure. Survival time is often short because of limited treatment options. Recent advances in targeted therapy and immunotherapy have changed the landscape for the treatment of advanced NSCLC. In the last 10 years, the US Food and Drug Administration (FDA) has approved more than 17 new medications for this devastating disease and more are coming. Molecular and immunogenic testing makes personalized medicine possible for patients with advanced NSCLC. The new medications provide promising efficacy and safety resulting in improved long-term survival for a significant number of patients. In this review, we summarize the recent advances in advanced/metastatic NSCLC therapeutics with a specific focus on first in-human or early-phase I/II clinical trials. These drugs either offer better alternatives to current standard drugs in the same class or are a completely new class of drugs with novel mechanisms of action. Advances are divided into (1) targeted agents, (2) antibody-drug conjugates, and (3) immunotherapies. Finally, we present a brief review of the emerging agents and ongoing clinical studies.
Collapse
MESH Headings
- Angiogenesis Inhibitors/therapeutic use
- Antineoplastic Agents, Immunological/therapeutic use
- Cancer Vaccines/therapeutic use
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Non-Small-Cell Lung/therapy
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Drugs, Investigational/therapeutic use
- Enzyme Inhibitors/therapeutic use
- Humans
- Immune Checkpoint Inhibitors/therapeutic use
- Immunoconjugates/therapeutic use
- Immunotherapy/trends
- Immunotherapy, Adoptive
- Lung Neoplasms/drug therapy
- Lung Neoplasms/pathology
- Lung Neoplasms/therapy
- Lymphocytes, Tumor-Infiltrating/transplantation
- Molecular Targeted Therapy
- Neoplasm Proteins/antagonists & inhibitors
- Oncogene Proteins, Fusion/antagonists & inhibitors
- Therapies, Investigational/trends
Collapse
Affiliation(s)
- Ruqin Chen
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Rami Manochakian
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Lauren James
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Abdel-Ghani Azzouqa
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Huashan Shi
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yan Zhang
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yujie Zhao
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Kexun Zhou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Yanyan Lou
- Division of Hematology and Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA.
| |
Collapse
|
27
|
Ruppert AM, Beau-Faller M, Debieuvre D, Ouafik L, Westeel V, Rouquette I, Mazières J, Bringuier PP, Monnet I, Escande F, Ricordel C, Merlio JP, Janicot H, Lemoine A, Foucher P, Poudenx M, Morin F, Langlais A, Souquet PJ, Barlesi F, Wislez M. Outcomes of Patients With Advanced NSCLC From the Intergroupe Francophone de Cancérologie Thoracique Biomarkers France Study by KRAS Mutation Subtypes. JTO Clin Res Rep 2020; 1:100052. [PMID: 34589947 PMCID: PMC8474404 DOI: 10.1016/j.jtocrr.2020.100052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022] Open
Abstract
Introduction KRAS mutations are detected in 20% to 30% of NSCLC. However, KRAS mutation subtypes may differently influence the outcome of patients with advanced NSCLC. Methods In the Biomarkers France study, 4894 KRAS mutations (26.2%) were detected in 4634 patients from the 17,664 enrolled patients with NSCLC. Survival and treatment data on noncurative stage III to IV NSCLC were available for 901 patients. First- and second-line treatment effects on progression-free survival and overall survival were analyzed according to the KRAS mutations subtype. Results Over 95% of patients with KRAS mutation were smokers or former smokers who were white (99.5%), presenting with adenocarcinoma (82.5%). The most common KRAS mutation subtype was G12C (374 patients; 41.5%), followed by G12V (168; 18.6%), G12D (131; 14.5%), G12A (62; 6.9%), G13C (45; 5.0%), G13D (31; 3.4%), and others (10; 1%). Approximately 21% of patients had transition mutation and 68.2% had a transversion mutation. G12D and transition mutations were predominant in never-smokers. The median overall survival for patients with KRAS-mutated NSCLC was 8.1 months (95% confidence interval [CI]: 7.5-9.5), without any differences according to the different KRAS subtypes mutations. The median progression-free survival was 4.6 months (95% CI: 4.2-5.1) for first-line treatment and 4.8 months (95% CI: 4.3-6.8) for second-line treatment, without any differences according to the different KRAS subtypes mutations. Conclusions KRAS mutation subtypes influenced neither treatment responses nor outcomes. The KRAS G12C mutation was detected in 41.5% of patients, who are now eligible for potent and specific G12C inhibitors.
Collapse
Affiliation(s)
- Anne-Marie Ruppert
- GRC n°04, Theranoscan, AP-HP, Groupe Hospitalier Sorbonne Université, Hôpital Tenon, Paris, France.,Department of Pneumology, AP-HP, Groupe Hospitalier Sorbonne Université, Hôpital Tenon, Paris, France
| | - Michèle Beau-Faller
- Laboratory of Biochemistry and Molecular Biology, Centre Hospitalier Universitaire de Strasbourg, Hôpital de Hautepierre, Strasbourg, France.,IRFAC UMR-S1113, Inserm, Université de Strasbourg, Strasbourg, France
| | - Didier Debieuvre
- Department of Pneumology, GHRMSA, Emile Muller Hospital, Mulhouse, France
| | - L'Houcine Ouafik
- Aix Marseille Univ, CNRS, INP, Inst Neurophysiopathol, Marseille, France.,Aix Marseille Univ, APHM, CHU Nord, Department of Tumor Biology, Marseille, France
| | - Virginie Westeel
- Centre Hospitalier Régional Universitaire de Besançon, Hôpital Jean Minjoz, Department of Pneumology, Besançon, France.,INSERM UMR 1098, Université de Bourgogne-Franche-Comté, Besançon, France
| | - Isabelle Rouquette
- Pathology Department, Centre Hospitalier Universitaire de Toulouse Institut Universitaire du Cancer de Toulouse, Oncopôle, Toulouse, France
| | - Julien Mazières
- Department of Pneumology, CHU de Toulouse, Université Paul Sabatier, Toulouse, France
| | - Pierre-Paul Bringuier
- Institut de Pathologie Multisite des Hospices Civils de Lyon Site Est and Université Claude Bernard Lyon 1, Lyon, France
| | - Isabelle Monnet
- Department of Pneumology, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Fabienne Escande
- Laboratory of Biochemistry and Molecular Biology, CHRU Lille, Lille, France
| | - Charles Ricordel
- Unité COSS INSERM U1242 - CEM - Université de Rennes, Rennes, France.,Department of Pneumology, CHU Rennes, Rennes, France
| | - Jean-Philippe Merlio
- Department of Tumor Biology, CHU and University of Bordeaux, INSERM U1053, Bordeaux, France
| | - Henri Janicot
- Department of Pneumology, CHU Clermont-Ferrand, Hôpital Gabriel Montpied, Clermont-Ferrand, France
| | - Antoinette Lemoine
- Department of Oncogenetics, AP-HP, Groupe Hospitalier APHP.Univeristé Pars-Saclay, Hôpital Paul Brousse, INSERM UMR-S 1193, Paris, France
| | - Pascal Foucher
- Department of Thoracic Oncology, University hospital Dijon-Bourgogne, Dijon, France
| | - Michel Poudenx
- Departement of Oncology, MédicaleCentre Antoine Lacassagne, Nice, France
| | - Franck Morin
- French Cooperative Thoracic Intergroup, Paris, France
| | | | - Pierre-Jean Souquet
- Department of Pneumology and Thoracic Oncology, Hospices Civils de Lyon, Centre Hospitalier Lyon-Sud, Pierre-Bénite, France
| | - Fabrice Barlesi
- Multidisciplinary Oncology & Therapeutic Innovations Department, Aix Marseille University, INSERM, CNRS, CRCM, APHM, Marseille, France
| | - Marie Wislez
- Department of Pneumology, Thoracic Oncology Unit, AP-HP, Groupe Hospitalier HUPC, Hôpital Cochin, Paris, France.,Centre de Recherche des Cordeliers, Université Paris Descartes, UMRS1138 « Complement, Inflammation and Cancer », Paris, France
| |
Collapse
|
28
|
Lau DK, Burge M, Roy A, Chau I, Haller DG, Shapiro JD, Peeters M, Pavlakis N, Karapetis CS, Tebbutt NC, Segelov E, Price TJ. Update on optimal treatment for metastatic colorectal cancer from the AGITG expert meeting: ESMO congress 2019. Expert Rev Anticancer Ther 2020; 20:251-270. [PMID: 32186929 DOI: 10.1080/14737140.2020.1744439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Introduction: Outcomes in metastatic colorectal cancer are improving, due to the tailoring of therapy enabled by better understanding of clinical behavior according to molecular subtype.Areas covered: A review of the literature and recent conference presentations was undertaken on the topic of systemic treatment of metastatic colorectal cancer. This review summarizes expert discussion of the current evidence for therapies in metastatic colorectal cancer (mCRC) based on molecular subgrouping.Expert opinion: EGFR-targeted and VEGF-targeted antibodies are now routinely incorporated into treatment strategies for mCRC. EGFR-targeted antibodies are restricted to patients with extended RAS wild-type profiles, with evidence that they should be further restricted to patients with left-sided tumors. Clinically distinct treatment pathways based on tumor RAS, BRAF, HER2 and MMR status, are now clinically applicable. Evidence suggests therapy for additional subgroups will soon be defined; the most advanced being for patients with KRAS G12 C mutation and gene TRK fusion defects.
Collapse
Affiliation(s)
- David K Lau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | - Matthew Burge
- Medical Oncology, Royal Brisbane Hospital, Brisbane, Australia.,University of Queensland, Brisbane, Australia
| | - Amitesh Roy
- Medical Oncology, Flinders Centre for Innovation in Cancer, Bedford Park, Australia
| | - Ian Chau
- GI and Lymphoma Unit, Royal Marsden NHS Foundation Trust, London and Surrey, UK
| | - Daniel G Haller
- Abramson Cancer Center at the Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jeremy D Shapiro
- Monash University, Melbourne, Australia.,Medical Oncology, Cabrini Medical Centre, Melbourne, Australia
| | - Marc Peeters
- Medical Oncology, University Hospital Antwerp, Edegem, Belgium
| | - Nick Pavlakis
- Medical Oncology, Royal North Shore Hospital, St Leonards, Australia.,Sydney University, Camperdown, Sydney, Australia
| | | | - Niall C Tebbutt
- Medical Oncology, Austin Health, Heidelberg, Australia.,Department of Surgery, University of Melbourne, Melbourne, Australia
| | - Eva Segelov
- Monash University, Melbourne, Australia.,Medical Oncology, Monash Medical Centre, Clayton, Australia
| | - Timothy J Price
- Medical Oncology, The Queen Elizabeth Hospital, Woodville, Australia
| |
Collapse
|