1
|
Lipchick B, Guterres AN, Chen HY, Zundell DM, Del Aguila S, Reyes-Uribe PI, Tirado Y, Basu S, Yin X, Kossenkov AV, Lu Y, Mills GB, Liu Q, Goldman AR, Murphy ME, Speicher DW, Villanueva J. Selective abrogation of S6K2 identifies lipid homeostasis as a survival vulnerability in MAPK inhibitor-resistant NRAS-mutant melanoma. Sci Transl Med 2025; 17:eadp8913. [PMID: 39908352 DOI: 10.1126/scitranslmed.adp8913] [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: 04/18/2024] [Revised: 09/17/2024] [Accepted: 01/06/2025] [Indexed: 02/07/2025]
Abstract
Although oncogenic NRAS activates mitogen-activated protein kinase (MAPK) signaling, inhibition of the MAPK pathway is not therapeutically efficacious in NRAS-mutant (NRASMUT) tumors. Here, we report that selectively silencing the ribosomal protein S6 kinase 2 (S6K2) while preserving the activity of S6K1 perturbs lipid metabolism, enhances fatty acid unsaturation, and triggers lethal lipid peroxidation in NRASMUT melanoma cells that are resistant to MAPK inhibition. S6K2 depletion induces endoplasmic reticulum stress and peroxisome proliferator-activated receptor α (PPARα) activation, triggering cell death selectively in MAPK inhibitor-resistant melanoma. We found that combining PPARα agonists and polyunsaturated fatty acids phenocopied the effects of S6K2 abrogation, blocking tumor growth in both patient-derived xenografts and immunocompetent murine melanoma models. Collectively, our study establishes S6K2 and its effector subnetwork as promising targets for NRASMUT melanomas that are resistant to global MAPK pathway inhibitors.
Collapse
Affiliation(s)
- Brittany Lipchick
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Adam N Guterres
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Hsin-Yi Chen
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Delaine M Zundell
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Segundo Del Aguila
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Patricia I Reyes-Uribe
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Yulissa Tirado
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Subhasree Basu
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Xiangfan Yin
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Andrew V Kossenkov
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Yiling Lu
- Department of Systems Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gordon B Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Sciences University, Portland, OR 97239, USA
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Aaron R Goldman
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
- Proteomics and Metabolomics Core Facility, Wistar Institute, Philadelphia, PA 19104 USA
| | - Maureen E Murphy
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - David W Speicher
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| | - Jessie Villanueva
- Molecular and Cellular Oncogenesis Program, Wistar Institute, Philadelphia, PA 19104, USA
| |
Collapse
|
2
|
Yayli G, Tokofsky A, Nayar U. The intersection of the HER2-low subtype with endocrine resistance: the role of interconnected signaling pathways. Front Oncol 2024; 14:1461190. [PMID: 39650068 PMCID: PMC11621065 DOI: 10.3389/fonc.2024.1461190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 10/29/2024] [Indexed: 12/11/2024] Open
Abstract
Since its introduction in the 1970s, endocrine therapy that targets the estrogen receptor alpha (ERα) signaling pathway has had tremendous success in the clinic in estrogen receptor positive (ER+) breast cancer. However, resistance to endocrine therapy eventually develops in virtually all patients with metastatic disease. Endocrine resistance is a primary unaddressed medical need for ER+ metastatic breast cancer patients. It has been shown that tumors become resistant through various mechanisms, converging on the acquisition of genetic alterations of ER, components of the MAP kinase pathway, or transcription factors (TFs). For instance, mutations in the human epidermal growth factor receptor-2 (HER2) lead to complete resistance to all current endocrine therapies including aromatase inhibitors, selective estrogen receptor modulators, and selective estrogen receptor degraders, as well as cross-resistance to CDK4/6 inhibitors (CDK4/6is). Emerging evidence points to an intriguing connection between endocrine-resistant tumors and the HER2-low subtype. Specifically, recent studies and our analysis of a publicly available breast cancer dataset both indicate that metastatic ER+ breast cancer with endocrine resistance conferred through acquired genetic alterations can often be classified as HER2-low rather than HER2-0/HER2-negative. Limited data suggest that acquired endocrine resistance can also be accompanied by a subtype switch. Therefore, we suggest that there is an underappreciated association between the HER2-low subtype and endocrine resistance. In this perspective piece, we explore the evidence linking the HER2-low subtype with the various pathways to endocrine resistance and suggest that there are signaling networks in HER2-low tumors that intersect endocrine resistance and can be effectively targeted.
Collapse
Affiliation(s)
- Gizem Yayli
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Alexa Tokofsky
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
| | - Utthara Nayar
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, United States
- Oncology, Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
3
|
Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, Eng H, Nair MG, Makvandi P, Geoerger B, Kulke MH, Baird RD, Prabhu JS, Carbone D, Pecoraro C, Teh DBL, Sethi G, Cavalieri V, Lin KH, Javidi-Sharifi NR, Toska E, Davids MS, Brown JR, Diana P, Stebbing J, Fruman DA, Kumar AP. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer 2023; 22:138. [PMID: 37596643 PMCID: PMC10436543 DOI: 10.1186/s12943-023-01827-6] [Citation(s) in RCA: 651] [Impact Index Per Article: 325.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023] Open
Abstract
The PI3K/AKT/mTOR (PAM) signaling pathway is a highly conserved signal transduction network in eukaryotic cells that promotes cell survival, cell growth, and cell cycle progression. Growth factor signalling to transcription factors in the PAM axis is highly regulated by multiple cross-interactions with several other signaling pathways, and dysregulation of signal transduction can predispose to cancer development. The PAM axis is the most frequently activated signaling pathway in human cancer and is often implicated in resistance to anticancer therapies. Dysfunction of components of this pathway such as hyperactivity of PI3K, loss of function of PTEN, and gain-of-function of AKT, are notorious drivers of treatment resistance and disease progression in cancer. In this review we highlight the major dysregulations in the PAM signaling pathway in cancer, and discuss the results of PI3K, AKT and mTOR inhibitors as monotherapy and in co-administation with other antineoplastic agents in clinical trials as a strategy for overcoming treatment resistance. Finally, the major mechanisms of resistance to PAM signaling targeted therapies, including PAM signaling in immunology and immunotherapies are also discussed.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Aaron S C Foo
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
| | - Hiu Y Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - Kenneth C H Yap
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119077, Singapore
| | - William Jacot
- Department of Medical Oncology, Institut du Cancer de Montpellier, Inserm U1194, Montpellier University, Montpellier, France
| | - Robert H Jones
- Cardiff University and Velindre Cancer Centre, Museum Avenue, Cardiff, CF10 3AX, UK
| | - Huiyan Eng
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bangalore, 560034, India
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China
| | - Birgit Geoerger
- Department of Pediatric and Adolescent Oncology, Gustave Roussy Cancer Center, Inserm U1015, Université Paris-Saclay, Paris, France
| | - Matthew H Kulke
- Section of Hematology and Medical Oncology, Boston University and Boston Medical Center, Boston, MA, USA
| | - Richard D Baird
- Cancer Research UK Cambridge Centre, Hills Road, Cambridge, CB2 0QQ, UK
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bangalore, 560034, India
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Camilla Pecoraro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Daniel B L Teh
- Departments of Ophthalmology and Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, and Neurobiology Programme, National University of Singapore, Singapore, Singapore
| | - Gautam Sethi
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
| | - Vincenzo Cavalieri
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kevin H Lin
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | | | - Eneda Toska
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Justin Stebbing
- Division of Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0NN, UK
| | - David A Fruman
- Department of Molecular Biology and Biochemistry, University of California, 216 Sprague Hall, Irvine, CA, USA
| | - Alan P Kumar
- Department of Surgery, National University Hospital Singapore, National University of Singapore, Singapore, Singapore.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
| |
Collapse
|
4
|
Yang H, Zhou X, Fu D, Le C, Wang J, Zhou Q, Liu X, Yuan Y, Ding K, Xiao Q. Targeting RAS mutants in malignancies: successes, failures, and reasons for hope. Cancer Commun (Lond) 2023; 43:42-74. [PMID: 36316602 PMCID: PMC9859734 DOI: 10.1002/cac2.12377] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 07/15/2022] [Accepted: 10/13/2022] [Indexed: 01/22/2023] Open
Abstract
RAS genes are the most frequently mutated oncogenes and play critical roles in the development and progression of malignancies. The mutation, isoform (KRAS, HRAS, and NRAS), position, and type of substitution vary depending on the tissue types. Despite decades of developing RAS-targeted therapies, only small subsets of these inhibitors are clinically effective, such as the allele-specific inhibitors against KRASG12C . Targeting the remaining RAS mutants would require further experimental elucidation of RAS signal transduction, RAS-altered metabolism, and the associated immune microenvironment. This study reviews the mechanisms and efficacy of novel targeted therapies for different RAS mutants, including KRAS allele-specific inhibitors, combination therapies, immunotherapies, and metabolism-associated therapies.
Collapse
Affiliation(s)
- Hang Yang
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
| | - Xinyi Zhou
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
| | - Dongliang Fu
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
| | - Chenqin Le
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
| | - Jiafeng Wang
- Department of Pharmacology and Department of Gastroenterology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058P. R. China
| | - Quan Zhou
- Department of Cell BiologySchool of Basic Medical SciencesZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Xiangrui Liu
- Department of Pharmacology and Department of Gastroenterology of the Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Ying Yuan
- Department of Medical Oncologythe Second Affiliated Hospital of Zhejiang University School of MedicineHangzhouZhejiang310058P. R. China
| | - Kefeng Ding
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| | - Qian Xiao
- Department of Colorectal Surgery and OncologyKey Laboratory of Cancer Prevention and InterventionMinistry of EducationThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiang310009P. R. China
| |
Collapse
|
5
|
Zhao MH, Wu AW. Targeting KRAS G12C mutations in colorectal cancer. Gastroenterol Rep (Oxf) 2022; 11:goac083. [PMID: 36632627 PMCID: PMC9825714 DOI: 10.1093/gastro/goac083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/02/2022] [Accepted: 12/06/2022] [Indexed: 01/09/2023] Open
Abstract
With the advent of Kirsten rat sarcoma viral oncogene homologue G12C (KRAS G12C) inhibitors, RAS is no longer considered undruggable. For the suppression of RAS, new therapeutic approaches have been suggested. However, current clinical studies have indicated therapeutic resistance after short-lived tumour suppression. According to preclinical studies, this might be associated with acquired genetic alterations, reactivation of downstream pathways, and stimulation for upstream signalling. In this review, we aimed to summarize current approaches for combination therapy to alleviate resistance to KRAS G12C inhibitors in colorectal cancer with a focus on the mechanisms of therapeutic resistance. We also analysed the relationship between various mechanisms and therapeutic resistance.
Collapse
Affiliation(s)
- Ming-He Zhao
- Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education; Unit III, Gastrointestinal Cancer Center, Peking University Cancer Hospital & Institute, Beijing, P. R. China
| | - Ai-Wen Wu
- Corresponding author. Key Laboratory of Carcinogenesis and Translational Research, Ministry of Education, Gastrointestinal Cancer Center, Unit III, Peking University Cancer Hospital & Institute, No. 52 Fucheng Rd, Haidian District, Beijing 100142, China. Tel/Fax: +86-10-88196981;
| |
Collapse
|
6
|
Prabhash K, Saha S, Joshi A, Noronha V, Patil V, Menon N, Singh A, Shetty O, Mittal N, Chandrani P, Chougule A. NRAS mutation in differentiated thyroid cancer. CANCER RESEARCH, STATISTICS, AND TREATMENT 2022. [DOI: 10.4103/crst.crst_296_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023] Open
|
7
|
Rio-Vilariño A, del Puerto-Nevado L, García-Foncillas J, Cebrián A. Ras Family of Small GTPases in CRC: New Perspectives for Overcoming Drug Resistance. Cancers (Basel) 2021; 13:3757. [PMID: 34359657 PMCID: PMC8345156 DOI: 10.3390/cancers13153757] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer remains among the cancers with the highest incidence, prevalence, and mortality worldwide. Although the development of targeted therapies against the EGFR and VEGFR membrane receptors has considerably improved survival in these patients, the appearance of resistance means that their success is still limited. Overactivation of several members of the Ras-GTPase family is one of the main actors in both tumour progression and the lack of response to cytotoxic and targeted therapies. This fact has led many resources to be devoted over the last decades to the development of targeted therapies against these proteins. However, they have not been as successful as expected in their move to the clinic so far. In this review, we will analyse the role of these Ras-GTPases in the emergence and development of colorectal cancer and their relationship with resistance to targeted therapies, as well as the status and new advances in the design of targeted therapies against these proteins and their possible clinical implications.
Collapse
Affiliation(s)
| | | | - Jesús García-Foncillas
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| | - Arancha Cebrián
- Translational Oncology Division, Hospital Universitario Fundación Jimenez Diaz, 28040 Madrid, Spain; (A.R.-V.); (L.d.P.-N.)
| |
Collapse
|
8
|
Iksen, Pothongsrisit S, Pongrakhananon V. Targeting the PI3K/AKT/mTOR Signaling Pathway in Lung Cancer: An Update Regarding Potential Drugs and Natural Products. Molecules 2021; 26:4100. [PMID: 34279440 PMCID: PMC8271933 DOI: 10.3390/molecules26134100] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 07/02/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is one of the most common cancers and has a high mortality rate. Due to its high incidence, the clinical management of the disease remains a major challenge. Several reports have documented a relationship between the phosphatidylinositol-3-kinase (PI3K)/ protein kinase B (AKT)/ mammalian target of rapamycin (mTOR) pathway and lung cancer. The recognition of this pathway as a notable therapeutic target in lung cancer is mainly due to its central involvement in the initiation and progression of the disease. Interest in using natural and synthetic medications to target these signaling pathways has increased in recent years, with promising results in vitro, in vivo, and in clinical trials. In this review, we focus on the current understanding of PI3K/AKT/mTOR signaling in tumor development. In addition to the signaling pathway, we highlighted the therapeutic potential of recently developed PI3K/AKT/mTOR inhibitors based on preclinical and clinical trials.
Collapse
Affiliation(s)
- Iksen
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.); (S.P.)
- Department of Pharmacy, Sekolah Tinggi Ilmu Kesehatan Senior Medan, Medan 20131, Indonesia
| | - Sutthaorn Pothongsrisit
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.); (S.P.)
| | - Varisa Pongrakhananon
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand; (I.); (S.P.)
- Preclinical Toxicity and Efficacy Assessment of Medicines and Chemicals Research Cluster, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
9
|
Garcia-Alvarez A, Ortiz C, Muñoz-Couselo E. Current Perspectives and Novel Strategies of NRAS-Mutant Melanoma. Onco Targets Ther 2021; 14:3709-3719. [PMID: 34135599 PMCID: PMC8202735 DOI: 10.2147/ott.s278095] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Melanoma is the deadliest cutaneous cancer. Activating mutations in NRAS are found in 20% of melanomas. NRAS-mutant melanoma is more aggressive and, therefore, has poorer outcomes, compared to non-NRAS-mutant melanoma. Despite promising preclinical data, to date immune checkpoint inhibitors remain the standard of care for locally advanced unresectable or metastatic NRAS melanoma. Data for efficacy of immunotherapy for NRAS melanoma mainly come from retrospective cohorts with divergent conclusions. MEK inhibitors have been the most developed targeted therapy approach. Although associated with an increase in progression-free survival, MEK inhibitors do not provide any benefit in terms of overall survival. Combination strategies with PI3K-AKT-mTOR pathway and CDK4/6 inhibitors seem to increase MEK inhibitors' benefit. Nevertheless, results from clinical trials are still prelaminar. A greater comprehension of the biology and intracellular interactions of NRAS-mutant melanoma will outline novel impactful strategies which could improve prognosis of these subgroup of patients.
Collapse
Affiliation(s)
- Alejandro Garcia-Alvarez
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Carolina Ortiz
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Eva Muñoz-Couselo
- Vall d’Hebron University Hospital, Medical Oncology Department, Melanoma and Other Skin Tumors Unit, Vall Hebron Institute of Oncology (VHIO), Barcelona, 08035, Spain
| |
Collapse
|
10
|
Patelli G, Tosi F, Amatu A, Mauri G, Curaba A, Patanè DA, Pani A, Scaglione F, Siena S, Sartore-Bianchi A. Strategies to tackle RAS-mutated metastatic colorectal cancer. ESMO Open 2021; 6:100156. [PMID: 34044286 PMCID: PMC8167159 DOI: 10.1016/j.esmoop.2021.100156] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/18/2022] Open
Abstract
The RAS oncogene is among the most commonly mutated in cancer. RAS mutations are identified in about half of patients diagnosed with metastatic colorectal cancer (mCRC), conferring poor prognosis and lack of response to anti-epidermal growth factor receptor (EGFR) antibodies. In the last decades, several investigational attempts failed in directly targeting RAS mutations, thus RAS was historically regarded as 'undruggable'. Recently, novel specific KRASG12C inhibitors showed promising results in different solid tumors, including mCRC, renewing interest in this biomarker as a target. In this review, we discuss different strategies of RAS targeting in mCRC, according to literature data in both clinical and preclinical settings. We recognized five main strategies focusing on those more promising: direct RAS targeting, targeting the mitogen-activated protein kinase (MAPK) pathway, harnessing RAS through immunotherapy combinations, RAS targeting through metabolic pathways, and finally other miscellaneous approaches. Direct KRASG12C inhibition is emerging as the most promising strategy in mCRC as well as in other solid malignancies. However, despite good disease control rates, tumor response and duration of response are still limited in mCRC. At this regard, combinational approaches with anti-epidermal growth factor receptor drugs or checkpoint inhibitors have been proposed to enhance treatment efficacy, based on encouraging results achieved in preclinical studies. Besides, concomitant therapies increasing metabolic stress are currently under evaluation and expected to also provide remarkable results in RAS codon mutations apart from KRASG12C. In conclusion, based on hereby reported efforts of translational research, RAS mutations should no longer be regarded as 'undruggable' and future avenues are now opening for translation in the clinic in mCRC.
Collapse
Affiliation(s)
- G Patelli
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - F Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - A Amatu
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - G Mauri
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Curaba
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - D A Patanè
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Pani
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - F Scaglione
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Clinical Pharmacology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - S Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy
| | - A Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy.
| |
Collapse
|
11
|
Mishra R, Patel H, Alanazi S, Kilroy MK, Garrett JT. PI3K Inhibitors in Cancer: Clinical Implications and Adverse Effects. Int J Mol Sci 2021; 22:3464. [PMID: 33801659 PMCID: PMC8037248 DOI: 10.3390/ijms22073464] [Citation(s) in RCA: 173] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 02/07/2023] Open
Abstract
The phospatidylinositol-3 kinase (PI3K) pathway is a crucial intracellular signaling pathway which is mutated or amplified in a wide variety of cancers including breast, gastric, ovarian, colorectal, prostate, glioblastoma and endometrial cancers. PI3K signaling plays an important role in cancer cell survival, angiogenesis and metastasis, making it a promising therapeutic target. There are several ongoing and completed clinical trials involving PI3K inhibitors (pan, isoform-specific and dual PI3K/mTOR) with the goal to find efficient PI3K inhibitors that could overcome resistance to current therapies. This review focuses on the current landscape of various PI3K inhibitors either as monotherapy or in combination therapies and the treatment outcomes involved in various phases of clinical trials in different cancer types. There is a discussion of the drug-related toxicities, challenges associated with these PI3K inhibitors and the adverse events leading to treatment failure. In addition, novel PI3K drugs that have potential to be translated in the clinic are highlighted.
Collapse
Affiliation(s)
| | | | | | | | - Joan T. Garrett
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267-0514, USA; (R.M.); (H.P.); (S.A.); (M.K.K.)
| |
Collapse
|
12
|
Organismal roles for the PI3Kα and β isoforms: their specificity, redundancy or cooperation is context-dependent. Biochem J 2021; 478:1199-1225. [DOI: 10.1042/bcj20210004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/16/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023]
Abstract
PI3Ks are important lipid kinases that produce phosphoinositides phosphorylated in position 3 of the inositol ring. There are three classes of PI3Ks: class I PI3Ks produce PIP3 at plasma membrane level. Although D. melanogaster and C. elegans have only one form of class I PI3K, vertebrates have four class I PI3Ks called isoforms despite being encoded by four different genes. Hence, duplication of these genes coincides with the acquisition of coordinated multi-organ development. Of the class I PI3Ks, PI3Kα and PI3Kβ, encoded by PIK3CA and PIK3CB, are ubiquitously expressed. They present similar putative protein domains and share PI(4,5)P2 lipid substrate specificity. Fifteen years after publication of their first isoform-selective pharmacological inhibitors and genetically engineered mouse models (GEMMs) that mimic their complete and specific pharmacological inhibition, we review the knowledge gathered in relation to the redundant and selective roles of PI3Kα and PI3Kβ. Recent data suggest that, further to their redundancy, they cooperate for the integration of organ-specific and context-specific signal cues, to orchestrate organ development, physiology, and disease. This knowledge reinforces the importance of isoform-selective inhibitors in clinical settings.
Collapse
|
13
|
Ryabaya OO, Abramov IS, Khochenkov DA, Akasov R, Sholina NV, Prokofieva AA. Rapamycin synergizes the cytotoxic effects of MEK inhibitor binimetinib and overcomes acquired resistance to therapy in melanoma cell lines in vitro. Invest New Drugs 2021; 39:987-1000. [PMID: 33683500 DOI: 10.1007/s10637-021-01089-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 02/15/2021] [Indexed: 12/14/2022]
Abstract
Objective The problem of drug resistance to BRAF-targeted therapy often occurs in melanoma treatment. Activation of PI3K/AKT/mTOR signaling pathway is one of the mechanisms of acquired resistance and a potential target for treatment. In the current research, we investigated that dual inhibition of mTOR and MEK synergistically reduced the viability of melanoma cells in vitro. Methods A combination of rapamycin (a macrolide immunosuppressant, mTOR inhibitor) and binimetinib (an anti-cancer small molecule, selective inhibitor of MEK) was studied using a panel of melanoma cell lines, including patient-derived cells. Results It was found, that combinatorial therapy of rapamycin (250 nM) and binimetinib (2 μM) resulted in 25% of cell viability compared to either rapamycin (85%) or binimetinib alone (50%) for A375 and vemurafenib-resistant Mel IL/R cells. The suppressed activation of mTOR and MEK by combined rapamycin and binimetinib treatment was confirmed using Western blot assay. Cell death occured via the apoptosis pathway; however, the combination treatment significantly increased the apoptosis only for Mel IL/R cells. The enhanced cytotoxic effect was also associated with enhanced cell cycle arrest in the G0/G1 phase. Conclusion In general, we provide the evidence that dual inhibition of mTOR and MEK could be promising for further preclinical investigations.
Collapse
Affiliation(s)
- Oxana O Ryabaya
- Department of the Experimental Diagnostic and Tumor Therapy N.N., Bloknin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow, 115478, Russia.
| | - Ivan S Abramov
- Center of Strategical Planning, Moscow, Russia, 10-1 Pogodinskaya Street, Moscow, 119121, Russia
| | - Dmitry A Khochenkov
- Department of the Experimental Diagnostic and Tumor Therapy N.N., Bloknin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow, 115478, Russia.,Togliatti State University, Belorusskaya str. 14, Togliatti, 445020, Russia
| | - Roman Akasov
- Institute of Molecular Medicine Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, Moscow, 119991, Russia.,Department of Biomaterials and Biotechnologies, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, 117997, Russia.,Federal Scientific Research Center «Crystallography and Photonics», Russian Academy of Sciences, 17a Butlerova st, Moscow, 117997, Russia
| | - Nataly V Sholina
- Department of the Experimental Diagnostic and Tumor Therapy N.N., Bloknin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow, 115478, Russia.,Institute of Molecular Medicine Sechenov First Moscow State Medical University, 8-2 Trubetskaya Street, Moscow, 119991, Russia
| | - Anastasia A Prokofieva
- Department of the Experimental Diagnostic and Tumor Therapy N.N., Bloknin National Medical Research Center of Oncology, 24 Kashirskoe Shosse, Moscow, 115478, Russia
| |
Collapse
|
14
|
Dunnett-Kane V, Nicola P, Blackhall F, Lindsay C. Mechanisms of Resistance to KRAS G12C Inhibitors. Cancers (Basel) 2021; 13:E151. [PMID: 33466360 PMCID: PMC7795113 DOI: 10.3390/cancers13010151] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/17/2022] Open
Abstract
KRAS is one of the most common human oncogenes, but concerted efforts to produce direct inhibitors have largely failed, earning KRAS the title of "undruggable". Recent efforts to produce subtype specific inhibitors have been more successful, and several KRASG12C inhibitors have reached clinical trials, including adagrasib and sotorasib, which have shown early evidence of efficacy in patients. Lessons from other inhibitors of the RAS pathway suggest that the effect of these drugs will be limited in vivo by the development of drug resistance, and pre-clinical studies of G12C inhibitors have identified evidence of this. In this review we discuss the current evidence for G12C inhibitors, the mechanisms of resistance to G12C inhibitors and potential approaches to overcome them. We discuss possible targets of combination therapy, including SHP2, receptor tyrosine kinases, downstream effectors and PD1/PDL1, and review the ongoing clinical trials investigating these inhibitors.
Collapse
Affiliation(s)
- Victoria Dunnett-Kane
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester M23 9LT, UK;
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (P.N.); (F.B.)
| | - Pantelis Nicola
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (P.N.); (F.B.)
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester M13 9PL, UK
| | - Fiona Blackhall
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (P.N.); (F.B.)
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester M13 9PL, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| | - Colin Lindsay
- Department of Medical Oncology, The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (P.N.); (F.B.)
- Division of Molecular and Clinical Cancer Sciences, University of Manchester, Manchester M13 9PL, UK
- Cancer Research UK Lung Cancer Centre of Excellence, Manchester M20 4BX, UK
| |
Collapse
|
15
|
Ruffinelli JC, Santos Vivas C, Sanz-Pamplona R, Moreno V. New advances in the clinical management of RAS and BRAF mutant colorectal cancer patients. Expert Rev Gastroenterol Hepatol 2021; 15:65-79. [PMID: 32946312 DOI: 10.1080/17474124.2021.1826305] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION In colorectal carcinogenesis, genetic alterations in RAS and BRAF oncogenes play an important role for cancer initiation and/or progression and represent a key focus in the search for targeted therapies. Despite many years of research and a great amount of studies, until very recently this pathway was considered extremely hard to downregulate to obtain a significant clinical impact in colorectal cancer patients. But better times are coming with the advent of new promising drugs and combinations strategies. AREAS COVERED In this review, we go over the biological characteristics of the MAPK pathway in colorectal tumors, while illustrating the clinical correlation of RAS and BRAF mutations, particularly its prognostic and predictive value. We also present newly data about recent improvements in the treatment strategy for patients harboring these types of tumors. EXPERT COMMENTARY With great advances in the knowledge of molecular basis of RAS and BRAF mutant colorectal cancer in conjunction with biotechnology development and the constant effort for improvement, in the near future many new therapeutic options would be available for the management of this group of patient with dismal prognosis.
Collapse
Affiliation(s)
- Jose Carlos Ruffinelli
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain
| | - Cristina Santos Vivas
- Department of Medical Oncology, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Consortium for Biomedical Research in Oncology (CIBERONC) , Barcelona, Spain.,Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona , Barcelona, Spain
| | - Rebeca Sanz-Pamplona
- Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) , Barcelona, Spain
| | - Victor Moreno
- Colorectal Cancer Group, ONCOBELL Program, Institut De Recerca Biomedica De Bellvitge (IDIBELL) , Barcelona, Spain.,Department of Clinical Sciences, Faculty of Medicine and Health Sciences, University of Barcelona , Barcelona, Spain.,Unit of Biomarkers and Susceptibility, Oncology Data Analytics Program (ODAP, Catalan Institute of Oncology (ICO), L'Hospitalet De Llobregat , Barcelona, Spain.,Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP) , Barcelona, Spain
| |
Collapse
|
16
|
Horizontal Combination of MEK and PI3K/mTOR Inhibition in BRAF Mutant Tumor Cells with or without Concomitant PI3K Pathway Mutations. Int J Mol Sci 2020; 21:ijms21207649. [PMID: 33081092 PMCID: PMC7589607 DOI: 10.3390/ijms21207649] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 09/22/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
The RAS/RAF and PI3K/Akt pathways play a key regulatory role in cancer and are often hit by oncogenic mutations. Despite molecular targeting, the long-term success of monotherapy is often hampered by de novo or acquired resistance. In the case of concurrent mutations in both pathways, horizontal combination could be a reasonable approach. In our study, we investigated the MEK inhibitor selumetinib and PI3K/mTOR dual inhibitor BEZ235 alone and in combination in BRAF-only mutant and BRAF + PI3K/PTEN double mutant cancer cells using short- and long-term 2D viability assays, spheroid assays, and immunoblots. In the 2D assays, selumetinib was more effective on BRAF-only mutant lines when compared to BRAF + PI3K/PTEN double mutants. Furthermore, combination therapy had an additive effect in most of the lines while synergism was observed in two of the double mutants. Importantly, in the SW1417 BRAF + PI3K double mutant cells, synergism was also confirmed in the spheroid and in the in vivo model. Mechanistically, p-Akt level decreased only in the SW1417 cell line after combination treatment. In conclusion, the presence of concurrent mutations alone did not predict a stronger response to combination treatment. Therefore, additional investigations are warranted to identify predictive factors that can select patients who can benefit from the horizontal combinational inhibition of these two pathways.
Collapse
|
17
|
Foster J, Freidlin B, Korn EL, Smith M. Evaluation of the contribution of randomised cancer clinical trials evaluating agents without documented single-agent activity. ESMO Open 2020; 5:e000871. [PMID: 33122353 PMCID: PMC7597487 DOI: 10.1136/esmoopen-2020-000871] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND With the development of targeted agents, the approach to combination cancer therapy has evolved to focus on identifying ways in which pathway inhibition by one agent may enhance the activity of other agents. In theory, this implies that under this new paradigm, agents are no longer required to show single-agent activity, as the pathway inhibited by the targeted agent may only have a therapeutic effect when given with other agents. This raises the question of the extent to which anticancer agents without single-agent activity can contribute to effective combination regimens. PATIENTS AND METHODS We reviewed outcomes of randomised phase 2 combination trials sponsored by the National Cancer Institute Cancer Therapy Evaluation Program that were activated in 2008 to 2017 and noted the single-agent activity of the experimental agents. RESULTS Fifty-three trials were identified, and 50 had available results: 7 (14%), 15 (30%) and 28 (56%) had experimental agents with single-agent activity classified as active, inactive and indeterminate, respectively. Thirteen per cent (95% CI=1.7% to 40.5%) of trials evaluating inactive agents and 11.6% (95% CI=3.9% to 25.1%) of trials evaluating agents without known single-agent activity (pooled inactive and indeterminate) were positive, compared with 42.9% (95% CI=9.9% to 81.6%) for agents with single-agent activity. CONCLUSIONS Incorporating agents without documented single-agent activity into treatment regimens is unlikely to produce meaningful improvements in activity unless there is compelling biological rationale. This finding has important implications for the prioritisation of anticancer agents for combination testing, and for the allocation of clinical trial resources.
Collapse
Affiliation(s)
- Jared Foster
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA.
| | - Boris Freidlin
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - E L Korn
- Biometric Research Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| | - Malcolm Smith
- Cancer Therapy Evaluation Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
18
|
Darici S, Alkhaldi H, Horne G, Jørgensen HG, Marmiroli S, Huang X. Targeting PI3K/Akt/mTOR in AML: Rationale and Clinical Evidence. J Clin Med 2020; 9:E2934. [PMID: 32932888 PMCID: PMC7563273 DOI: 10.3390/jcm9092934] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Revised: 09/07/2020] [Accepted: 09/10/2020] [Indexed: 12/12/2022] Open
Abstract
Acute myeloid leukemia (AML) is a highly heterogeneous hematopoietic malignancy characterized by excessive proliferation and accumulation of immature myeloid blasts in the bone marrow. AML has a very poor 5-year survival rate of just 16% in the UK; hence, more efficacious, tolerable, and targeted therapy is required. Persistent leukemia stem cell (LSC) populations underlie patient relapse and development of resistance to therapy. Identification of critical oncogenic signaling pathways in AML LSC may provide new avenues for novel therapeutic strategies. The phosphatidylinositol-3-kinase (PI3K)/Akt and the mammalian target of rapamycin (mTOR) signaling pathway, is often hyperactivated in AML, required to sustain the oncogenic potential of LSCs. Growing evidence suggests that targeting key components of this pathway may represent an effective treatment to kill AML LSCs. Despite this, accruing significant body of scientific knowledge, PI3K/Akt/mTOR inhibitors have not translated into clinical practice. In this article, we review the laboratory-based evidence of the critical role of PI3K/Akt/mTOR pathway in AML, and outcomes from current clinical studies using PI3K/Akt/mTOR inhibitors. Based on these results, we discuss the putative mechanisms of resistance to PI3K/Akt/mTOR inhibition, offering rationale for potential candidate combination therapies incorporating PI3K/Akt/mTOR inhibitors for precision medicine in AML.
Collapse
Affiliation(s)
- Salihanur Darici
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Hazem Alkhaldi
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Gillian Horne
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Heather G. Jørgensen
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| | - Sandra Marmiroli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, 41124 Modena, Italy;
| | - Xu Huang
- Haemato-Oncology/Systems Medicine Group, Paul O’Gorman Leukaemia Research Centre, University of Glasgow, Glasgow G12 0ZD, UK; (H.A.); (G.H.); (H.G.J.)
| |
Collapse
|
19
|
Vanni I, Tanda ET, Dalmasso B, Pastorino L, Andreotti V, Bruno W, Boutros A, Spagnolo F, Ghiorzo P. Non-BRAF Mutant Melanoma: Molecular Features and Therapeutical Implications. Front Mol Biosci 2020; 7:172. [PMID: 32850962 PMCID: PMC7396525 DOI: 10.3389/fmolb.2020.00172] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 07/03/2020] [Indexed: 02/06/2023] Open
Abstract
Melanoma is one of the most aggressive tumors of the skin, and its incidence is growing worldwide. Historically considered a drug resistant disease, since 2011 the therapeutic landscape of melanoma has radically changed. Indeed, the improved knowledge of the immune system and its interactions with the tumor, and the ever more thorough molecular characterization of the disease, has allowed the development of immunotherapy on the one hand, and molecular target therapies on the other. The increased availability of more performing technologies like Next-Generation Sequencing (NGS), and the availability of increasingly large genetic panels, allows the identification of several potential therapeutic targets. In light of this, numerous clinical and preclinical trials are ongoing, to identify new molecular targets. Here, we review the landscape of mutated non-BRAF skin melanoma, in light of recent data deriving from Whole-Exome Sequencing (WES) or Whole-Genome Sequencing (WGS) studies on melanoma cohorts for which information on the mutation rate of each gene was available, for a total of 10 NGS studies and 992 samples, focusing on available, or in experimentation, targeted therapies beyond those targeting mutated BRAF. Namely, we describe 33 established and candidate driver genes altered with frequency greater than 1.5%, and the current status of targeted therapy for each gene. Only 1.1% of the samples showed no coding mutations, whereas 30% showed at least one mutation in the RAS genes (mostly NRAS) and 70% showed mutations outside of the RAS genes, suggesting potential new roads for targeted therapy. Ongoing clinical trials are available for 33.3% of the most frequently altered genes.
Collapse
Affiliation(s)
- Irene Vanni
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | | | - Bruna Dalmasso
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Lorenza Pastorino
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Virginia Andreotti
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - William Bruno
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| | - Andrea Boutros
- Medical Oncology, IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | | | - Paola Ghiorzo
- Genetics of Rare Cancers, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Genetics of Rare Cancers, Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
| |
Collapse
|
20
|
Abstract
RAS (KRAS, NRAS and HRAS) is the most frequently mutated gene family in cancers, and, consequently, investigators have sought an effective RAS inhibitor for more than three decades. Even 10 years ago, RAS inhibitors were so elusive that RAS was termed 'undruggable'. Now, with the success of allele-specific covalent inhibitors against the most frequently mutated version of RAS in non-small-cell lung cancer, KRASG12C, we have the opportunity to evaluate the best therapeutic strategies to treat RAS-driven cancers. Mutation-specific biochemical properties, as well as the tissue of origin, are likely to affect the effectiveness of such treatments. Currently, direct inhibition of mutant RAS through allele-specific inhibitors provides the best therapeutic approach. Therapies that target RAS-activating pathways or RAS effector pathways could be combined with these direct RAS inhibitors, immune checkpoint inhibitors or T cell-targeting approaches to treat RAS-mutant tumours. Here we review recent advances in therapies that target mutant RAS proteins and discuss the future challenges of these therapies, including combination strategies.
Collapse
|
21
|
Dilly A, Honick BD, Lee YJ, Bartlett DL, Choudry HA. Rational application of targeted therapeutics in mucinous colon/appendix cancers with positive predictive factors. Cancer Med 2020; 9:1753-1767. [PMID: 31958897 PMCID: PMC7050077 DOI: 10.1002/cam4.2847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/26/2022] Open
Abstract
Molecular‐targeted therapies have demonstrated disappointing results against most advanced solid cancers. This may largey be attributed to irrational drug use against unselected cancers. We investigated the efficacy of dual MEK‐PI3K drug therapy against KRAS mutated mucin 2 (MUC2)‐secreting LS174T cells and patient‐derived ex vivo and in vivo models of KRAS mutated mucinous colon/appendix cancers. These tumors demonstrate unique phenotypic and genotypic features that likely predict sensitivity to this targeted co‐therapy. Co‐treatment with MEK inhibitor (trametinib) and PI3K inhibitor (pictilisib)‐induced synergistic cytotoxicity and intrinsic mitochondrial‐mediated apoptosis in LS174T cells and tumor explants in vitro. Dual drug therapy also induced endoplasmic reticulum stress (ERS)‐associated proteins (GRP78/BiP, ATF4, and CHOP). However, CHOP knock‐down assays demonstrated that mitochondrial‐mediated apoptosis in LS174T cells was not ERS‐dependent. Dual drug therapy also significantly decreased MUC2 expression, MUC2 post‐translational modification (palmitoylation) and secretion in LS174T cells, suggesting a simultaneous cytotoxic and mucin suppressive mechanism of action. We also demonstrated effective mucinous tumor growth suppression in ex vivo epithelial organoid (colonoid) cultures and in in vivo intraperitoneal patient‐derived xenograft models derived from mucinous colon/appendix cancer. These promising preclinical data support a role for dual MEK‐PI3K inhibitor therapy in mucinous colon/appendix cancers. We postulate that mucinous KRAS mutated cancers are especially vulnerable to this co‐treatment based on their unique phenotypic and genotypic characteristics.
Collapse
MESH Headings
- Animals
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Appendiceal Neoplasms/genetics
- Appendiceal Neoplasms/pathology
- Appendiceal Neoplasms/therapy
- Appendix/cytology
- Appendix/pathology
- Appendix/surgery
- Cell Line, Tumor
- Chemotherapy, Adjuvant/methods
- Colon/cytology
- Colon/pathology
- Colon/surgery
- Colonic Neoplasms/genetics
- Colonic Neoplasms/pathology
- Colonic Neoplasms/therapy
- Drug Synergism
- Endoplasmic Reticulum Chaperone BiP
- Female
- Humans
- Indazoles/pharmacology
- Indazoles/therapeutic use
- Intestinal Mucosa/cytology
- Intestinal Mucosa/pathology
- Intestinal Mucosa/surgery
- Mice
- Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors
- Molecular Targeted Therapy/methods
- Mucin-2/metabolism
- Mutation
- Neoplasms, Cystic, Mucinous, and Serous/genetics
- Neoplasms, Cystic, Mucinous, and Serous/pathology
- Neoplasms, Cystic, Mucinous, and Serous/therapy
- Phosphatidylinositol 3-Kinases/metabolism
- Primary Cell Culture
- Protein Kinase Inhibitors/pharmacology
- Protein Kinase Inhibitors/therapeutic use
- Proto-Oncogene Proteins p21(ras)/genetics
- Pyridones/pharmacology
- Pyridones/therapeutic use
- Pyrimidinones/pharmacology
- Pyrimidinones/therapeutic use
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Ashokkumar Dilly
- Department of SurgeryUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Brendon D. Honick
- Department of SurgeryUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Yong J. Lee
- Department of SurgeryUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - David L. Bartlett
- Department of SurgeryUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| | - Haroon A. Choudry
- Department of SurgeryUniversity of Pittsburgh Medical CenterPittsburghPAUSA
| |
Collapse
|
22
|
Westin SN, Sill MW, Coleman RL, Waggoner S, Moore KN, Mathews CA, Martin LP, Modesitt SC, Lee S, Ju Z, Mills GB, Schilder RJ, Fracasso PM, Birrer MJ, Aghajanian C. Safety lead-in of the MEK inhibitor trametinib in combination with GSK2141795, an AKT inhibitor, in patients with recurrent endometrial cancer: An NRG Oncology/GOG study. Gynecol Oncol 2019; 155:420-428. [PMID: 31623857 DOI: 10.1016/j.ygyno.2019.09.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/26/2019] [Accepted: 09/27/2019] [Indexed: 11/25/2022]
Abstract
OBJECTIVE We sought to determine safety and efficacy of the AKT inhibitor, GSK2141795, combined with the MEK inhibitor, trametinib, in endometrial cancer. METHODS Patients with measurable recurrent endometrial cancer were eligible. One to two prior cytotoxic regimens were allowed; prior use of a MEK or PI3K pathway inhibitor was excluded. Initial trial design consisted of a KRAS mutation stratified randomized phase II with a safety lead-in evaluating the combination. For the safety lead in, the previously recommended phase 2 dose (RP2D; trametinib 1.5 mg, GSK2141795 50 mg) was chosen for Dose Level 1 (DL1). RESULTS Of 26 enrolled patients, 14 were treated on DL1 and 12 were treated on DL-1 (trametinib 1.5 mg, GSK2141795 25 mg). Most common histologies were endometrioid (58%) and serous (27%). Four of 25 (16%) patients were KRAS mutant. Dose limiting toxicities (DLTs) were assessed during cycle 1. DL1 had 8 DLTs (hypertension (n = 2), mucositis (2), rash (2), dehydration, stroke/acute kidney injury). DL1 was deemed non-tolerable so DL-1 was explored. DL-1 had no DLTs. Sixty-five percent of patients had ≥ grade 3 toxicity. There were no responses in DL1 (0%, 90%CI 0-15%) and 1 response in DL-1 (8.3%, 90%CI 0.4-33.9%). Proportion PFS at 6 months for DL1 is 14%, and 25% for DL-1. CONCLUSION The combination of trametinib and GSK2141795 had high levels of toxicity in endometrial cancer at the previously RP2D but was tolerable at a reduced dose. Due to insufficient preliminary efficacy at a tolerable dose, the Phase II study was not initiated.
Collapse
Affiliation(s)
- Shannon N Westin
- Department of Gynecologic Oncology, University of Texas M. D Anderson Cancer Center, USA.
| | - Michael W Sill
- NRG Oncology Statistics and Data Management Center Buffalo Office, Roswell Park Cancer Institute, USA.
| | - Robert L Coleman
- Department of Gynecologic Oncology, University of Texas M. D Anderson Cancer Center, USA.
| | - Steven Waggoner
- Department of Gynecologic Oncology, Case Western Reserve University, USA.
| | - Kathleen N Moore
- Department of Gynecologic Oncology, University of Oklahoma Health Sciences Center, Stephenson Cancer Center, USA.
| | - Cara A Mathews
- Department of Gynecologic Oncology, Women & Infants Hospital, USA.
| | - Lainie P Martin
- Department of Hematology/Oncology, Fox Chase Cancer Center, USA.
| | - Susan C Modesitt
- Director of Gynecologic Oncology Division, University of Virginia, USA.
| | - Sanghoon Lee
- Department of Medicine and the UVA Cancer Center, University of Virginia, USA.
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas M. D Anderson Cancer Center, USA.
| | - Gordon B Mills
- Department of Medicine and the UVA Cancer Center, University of Virginia, USA.
| | - Russell J Schilder
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, USA.
| | - Paula M Fracasso
- Department of Systems Biology, University of Texas M.D Anderson Cancer Center, USA.
| | | | - Carol Aghajanian
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical College, USA.
| |
Collapse
|
23
|
Liu JF, Gray KP, Wright AA, Campos S, Konstantinopoulos PA, Peralta A, MacNeill K, Morrissey S, Whalen C, Dillon D, Matulonis UA. Results from a single arm, single stage phase II trial of trametinib and GSK2141795 in persistent or recurrent cervical cancer. Gynecol Oncol 2019; 154:95-101. [PMID: 31118140 DOI: 10.1016/j.ygyno.2019.05.003] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/24/2022]
Abstract
BACKGROUND Improved treatment for advanced cervical cancer is needed; currently, treatment options include combined chemotherapy and bevacizumab or pembrolizumab monotherapy for PD-L1 positive disease. PIK3CA and KRAS mutations have been reported in cervical cancers; this study therefore tested dual inhibition of PI3K and RAS signaling by combining the MEK inhibitor trametinib and the AKT inhibitor GSK2141795 in recurrent cervical cancer. METHODS This was an investigator-initiated phase II study combining trametinib and GSK2141795 in patients with recurrent cervical cancer. Primary endpoint was best tumor response; secondary endpoints included progression free survival, overall survival, and safety assessment. Translational objectives included characterization of molecular alterations in PI3K and RAS signaling pathway genes. RESULTS Planned accrual was 35 patients; 14 patients were enrolled and received at least one dose of study drug before the study was terminated due to discontinuation of GSK2141795 development. There were no confirmed responses; 1 patient had an unconfirmed PR, 8 had stable disease, 3 had progression as best response, and 2 were unevaluable. Toxicities were mostly grade 1 and 2, although 57% of patients experienced grade 3/4 adverse events and 50% patients required a dose reduction. CONCLUSIONS The combination of trametinib and GSK2141795 was feasible but required dose holds and modifications for adverse events; however, anti-cancer activity was minimal, even in patients with PI3K or RAS pathway alterations. Although the study was terminated early after GSK2141795 development was halted, the findings in these 14 patients do not support further development of this combination in cervical cancer.
Collapse
Affiliation(s)
- Joyce F Liu
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America.
| | - Kathryn P Gray
- Department of Data Science, Dana-Farber Cancer Institute Boston, MA, United States of America
| | - Alexi A Wright
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Susana Campos
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Panagiotis A Konstantinopoulos
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Ariana Peralta
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Kimberley MacNeill
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Stephanie Morrissey
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Christin Whalen
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States of America
| | - Ursula A Matulonis
- Division of Gynecologic Oncology, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, United States of America
| |
Collapse
|
24
|
Shapiro GI, LoRusso P, Kwak E, Pandya S, Rudin CM, Kurkjian C, Cleary JM, Pilat MJ, Jones S, de Crespigny A, Fredrickson J, Musib L, Yan Y, Wongchenko M, Hsieh HJ, Gates MR, Chan IT, Bendell J. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Invest New Drugs 2019; 38:419-432. [PMID: 31020608 DOI: 10.1007/s10637-019-00776-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Accepted: 04/01/2019] [Indexed: 12/21/2022]
Abstract
Purpose We investigated the combination of the MEK inhibitor, cobimetinib, and the pan-PI3K inhibitor, pictilisib, in an open-label, phase Ib study. Experimental Design Patients with advanced solid tumors were enrolled in 3 dose escalation schedules: (1) both agents once-daily for 21-days-on 7-days-off ("21/7"); (2) intermittent cobimetinib and 21/7 pictilisib ("intermittent"); or (3) both agents once-daily for 7-days-on 7-days-off ("7/7"). Starting doses for the 21/7, intermittent, and 7/7 schedules were 20/80, 100/130, and 40/130 mg of cobimetinib/pictilisib, respectively. Nine indication-specific expansion cohorts interrogated the recommended phase II dose and schedule. Results Of 178 enrollees (dose escalation: n = 98), 177 patients were dosed. The maximum tolerated doses for cobimetinib/pictilisib (mg) were 40/100, 125/180, and not reached, for the 21/7, intermittent, and 7/7 schedules, respectively. Six dose-limiting toxicities included grade 3 (G3) elevated lipase, G4 elevated creatine phosphokinase, and G3 events including fatigue concurrent with a serious adverse event (SAE) of diarrhea, decreased appetite, and SAEs of hypersensitivity and dehydration. Common drug-related adverse events included nausea, fatigue, vomiting, decreased appetite, dysgeusia, rash, and stomatitis. Pharmacokinetic parameters of the drugs used in combination were unaltered compared to monotherapy exposures. Confirmed partial responses were observed in patients with BRAF-mutant melanoma (n = 1) and KRAS-mutant endometrioid adenocarcinoma (n = 1). Eighteen patients remained on study ≥6 months. Biomarker data established successful blockade of MAP kinase (MAPK) and PI3K pathways. The metabolic response rate documented by FDG-PET was similar to that observed with cobimetinib monotherapy. Conclusions Cobimetinib and pictilisib combination therapy in patients with solid tumors had limited tolerability and efficacy.
Collapse
Affiliation(s)
- Geoffrey I Shapiro
- Dana-Farber Cancer Institute, Mayer 446, 450 Brookline Avenue, Boston, MA, 02215, USA.
| | | | - Eunice Kwak
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Susan Pandya
- Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Carla Kurkjian
- Stephenson Cancer Center University of Oklahoma, Oklahoma City, OK, USA
| | - James M Cleary
- Dana-Farber Cancer Institute, Mayer 446, 450 Brookline Avenue, Boston, MA, 02215, USA
| | | | - Suzanne Jones
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| | | | | | - Luna Musib
- Genentech, Inc., South San Francisco, CA, USA
| | - Yibing Yan
- Genentech, Inc., South San Francisco, CA, USA
| | | | | | | | - Iris T Chan
- Genentech, Inc., South San Francisco, CA, USA
| | - Johanna Bendell
- Sarah Cannon Research Institute/Tennessee Oncology, Nashville, TN, USA
| |
Collapse
|
25
|
Maiti A, Naqvi K, Kadia TM, Borthakur G, Takahashi K, Bose P, Daver NG, Patel A, Alvarado Y, Ohanian M, DiNardo CD, Cortes JE, Jabbour EJ, Garcia-Manero G, Kantarjian HM, Ravandi F. Phase II Trial of MEK Inhibitor Binimetinib (MEK162) in RAS-mutant Acute Myeloid Leukemia. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2019; 19:142-148.e1. [PMID: 30635233 PMCID: PMC11852403 DOI: 10.1016/j.clml.2018.12.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 11/17/2018] [Accepted: 12/12/2018] [Indexed: 01/25/2023]
Abstract
BACKGROUND Relapsed and refractory (R/R) acute myeloid leukemia (AML) continues to be a therapeutic challenge with poor outcomes. Dysregulation of the mitogen-activated protein (MAP) kinase/extracellular-signal regulated kinase (ERK) pathway frequently occurs in AML and myelodysplastic syndrome (MDS). Preclinical studies and early-phase trials have shown promise for MAP-ERK kinase (MEK) inhibition in AML. We evaluated the safety and efficacy of the MEK 1/2 inhibitor binimetinib in advanced myeloid malignancies. PATIENTS AND METHODS Nineteen patients with R/R AML and MDS, who were not candidates for intensive chemotherapy or with disease resistance or intolerance to standard treatment were enrolled in the present phase II study of binimetinib dosed twice daily continuously in 28-day cycles. RESULTS The median age of the cohort was 64 years (range, 31-85 years). These patients had received a median of 3 previous lines of therapy (range, 1-6). The median bone marrow blast percentage was 49% (range, 2%-94%), and 14 patients had RAS mutations. The patients received a median of 2 cycles (range, 1-4 cycles) of binimetinib and received treatment for a median duration of 1.2 months (range, 0.1-3.4 months). Sixteen patients (84%) received the 45-mg twice daily dose. The most common grade 3/4 treatment-emergent adverse events were hypokalemia (6%), hypotension (6%), lung infection (6%), and febrile neutropenia (6%). No treatment-related deaths occurred. One of the 13 evaluable patients (8%) achieved a complete response with incomplete blood count recovery lasting 2.1 months. The other 12 patients (92%) did not have a response. Six patients could not be evaluated. CONCLUSION Binimetinib had tolerable safety profile with a minimal response in RAS-mutant AML. Future studies should focus on better patient selection and synergistic combination therapies involving MEK inhibition.
Collapse
Affiliation(s)
- Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX; Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kiran Naqvi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ami Patel
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Maro Ohanian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jorge E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias J Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
26
|
Clinical update on K-Ras targeted therapy in gastrointestinal cancers. Crit Rev Oncol Hematol 2018; 130:78-91. [DOI: 10.1016/j.critrevonc.2018.07.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Revised: 07/24/2018] [Accepted: 07/31/2018] [Indexed: 12/11/2022] Open
|
27
|
KRAS-Mutant non-small cell lung cancer: From biology to therapy. Lung Cancer 2018; 124:53-64. [PMID: 30268480 DOI: 10.1016/j.lungcan.2018.07.013] [Citation(s) in RCA: 209] [Impact Index Per Article: 29.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 07/09/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022]
Abstract
In patients with non-small cell lung cancer (NSCLC), the most frequent oncogene driver mutation in Western countries is Kirsten rat sarcoma viral oncogene homolog (KRAS), and KRAS-mutant NSCLC is associated with smoking. There are various sources of biological heterogeneity of KRAS-mutant NSCLC, including different genotypes that may be associated with specific clinical outcomes, the presence of other co-mutations that exhibit different biological features and drug sensitivity patterns, and mutant allelic content. The efficacy of chemotherapy in patients with KRAS-mutant NSCLC is generally poor and numerous novel therapeutic strategies have been developed. These approaches include targeting KRAS membrane associations, targeting downstream signalling pathways, the use of KRAS synthetic lethality, direct targeting of KRAS, and immunotherapy. Of these, immunotherapy may be one of the most promising treatment approaches for patients with KRAS-mutant NSCLC. Recent data also suggest the potential for distinct efficacy of immunotherapy according to the presence of other co-mutations. In view of the biological heterogeneity of KRAS-mutant NSCLC, treatment will likely need to be individualised and, in future, may require the use of rational combinations of treatment, many of which are currently under investigation.
Collapse
|
28
|
Cho M, Gong J, Frankel P, Synold TW, Lim D, Chung V, Chao J, Li D, Chen Y, Sentovich S, Melstrom K, Singh G, Luevanos E, Fakih M. A phase I clinical trial of binimetinib in combination with FOLFOX in patients with advanced metastatic colorectal cancer who failed prior standard therapy. Oncotarget 2017; 8:79750-79760. [PMID: 29108355 PMCID: PMC5668088 DOI: 10.18632/oncotarget.19336] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 06/30/2017] [Indexed: 12/20/2022] Open
Abstract
Background This was a first in-human, open-label, dose-escalation phase I study conducted to evaluate the maximum tolerated dose (MTD), safety, and efficacy of the combination of oral binimetinib and FOLFOX. Materials and Methods Patients with metastatic colorectal cancer (mCRC) who progressed on prior standard therapies received twice daily binimetinib continuously or intermittently with FOLFOX. Dose-limiting toxicities (DLTs) were assessed in the first 2 cycles of study treatment. Pharmacokinetic (PK) analysis of 5-FU and oxaliplatin was performed at the MTD in an expanded 6 patient cohort. Results Twenty-six patients were enrolled and assessed for safety. In the dose-escalation phase, no DLTs were noted in all binimetinib dosing schedules and the MTD of binimetinib in with FOLFOX was 45 mg orally twice daily. There were no significant differences in the PKs of 5-FU or oxaliplatin with or without binimetinib. Continuous dosing of binimetinib produced SD at 2 months in 9 of 13 evaluable patients and a median PFS of 3.5 months. Nine of 10 patients had PD at 2 months on the intermittent arm. Conclusions Oral binimetinib and FOLFOX has a manageable toxicity profile and showed some evidence of antitumor activity in heavily pretreated mCRC patients.
Collapse
Affiliation(s)
- May Cho
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Jun Gong
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Paul Frankel
- Department of Statistics, City of Hope National Medical Center, Duarte, CA, USA
| | - Timothy W Synold
- Department of Cancer Biology, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Dean Lim
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Vincent Chung
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Joseph Chao
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Daneng Li
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Yuan Chen
- Department of Molecular Medicine, Beckman Research Institute of City of Hope, Duarte,CA, USA
| | - Stephen Sentovich
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Kurt Melstrom
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Gagandeep Singh
- Department of Surgical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Eloise Luevanos
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| | - Marwan Fakih
- Department of Medical Oncology, City of Hope National Medical Center, Duarte, CA, USA
| |
Collapse
|
29
|
Bahrami A, Hassanian SM, ShahidSales S, Farjami Z, Hasanzadeh M, Anvari K, Aledavood A, Maftouh M, Ferns GA, Khazaei M, Avan A. Targeting RAS signaling pathway as a potential therapeutic target in the treatment of colorectal cancer. J Cell Physiol 2017; 233:2058-2066. [PMID: 28262927 DOI: 10.1002/jcp.25890] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 03/02/2017] [Indexed: 12/19/2022]
Abstract
The V-Ki-ras2 Kirsten rat sarcoma viral oncogene homolog (KRAS) is frequently dysregulated in colorectal cancer (CRC). It is involved in the modulation of several downstream effectors, that include: Raf/Mek/Erk, PI3K/Akt, RalGDS/p38MAPK, and Rac/Rho, and thereby influences tumorigenesis, the invasive behaviors of tumor cell, and resistance to therapy. There is growing evidence exploring the use of drugs that target these pathways in the treatment of CRC. Cetuximab has been approved for CRC patients without a KRAS mutation, or for EGFR-expressing metastatic CRC, although some of the patients have a mutation of KRAS and NRAS. This review summarizes the recent knowledge about the therapeutic potential of targeting RAS with particular emphasis on recent preclinical and clinical studies in treatment of CRC.
Collapse
Affiliation(s)
- Afsane Bahrami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Medical Biochemistry, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodabeh ShahidSales
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Farjami
- Department of Modern Sciences and Technologies, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Malihe Hasanzadeh
- Department of Gynecology Oncology, Woman Health Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Anvari
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Aledavood
- Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mina Maftouh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, UK
| | - Majid Khazaei
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Cancer Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
30
|
Tomasini P, Walia P, Labbe C, Jao K, Leighl NB. Targeting the KRAS Pathway in Non-Small Cell Lung Cancer. Oncologist 2016; 21:1450-1460. [PMID: 27807303 PMCID: PMC5153335 DOI: 10.1634/theoncologist.2015-0084] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 07/29/2016] [Indexed: 12/19/2022] Open
Abstract
: Lung cancer remains the leading cause of cancer-related deaths worldwide. However, significant progress has been made individualizing therapy based on molecular aberrations (e.g., EGFR, ALK) and pathologic subtype. KRAS is one of the most frequently mutated genes in non-small cell lung cancer (NSCLC), found in approximately 30% of lung adenocarcinomas, and is thus an appealing target for new therapies. Although no targeted therapy has yet been approved for the treatment of KRAS-mutant NSCLC, there are multiple potential therapeutic approaches. These may include direct inhibition of KRAS protein, inhibition of KRAS regulators, alteration of KRAS membrane localization, and inhibition of effector molecules downstream of mutant KRAS. This article provides an overview of the KRAS pathway in lung cancer and related therapeutic strategies under investigation. IMPLICATIONS FOR PRACTICE The identification of oncogene-addicted cancers and specific inhibitors has revolutionized non-small cell lung cancer (NSCLC) treatment and outcomes. One of the most commonly mutated genes in adenocarcinoma is KRAS, found in approximately 30% of lung adenocarcinomas, and thus it is an appealing target for new therapies. This review provides an overview of the KRAS pathway and related targeted therapies under investigation in NSCLC. Some of these agents may play a key role in KRAS-mutant NSCLC treatment in the future.
Collapse
Affiliation(s)
- Pascale Tomasini
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Preet Walia
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Catherine Labbe
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Kevin Jao
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| | - Natasha B Leighl
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Stjepanovic N, Velazquez-Martin J, Bedard P. Ocular toxicities of MEK inhibitors and other targeted therapies. Ann Oncol 2016; 27:998-1005. [DOI: 10.1093/annonc/mdw100] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 02/17/2016] [Indexed: 12/11/2022] Open
|
32
|
Posch C, Vujic I, Monshi B, Sanlorenzo M, Weihsengruber F, Rappersberger K, Ortiz-Urda S. Searching for the Chokehold of NRAS Mutant Melanoma. J Invest Dermatol 2016; 136:1330-1336. [PMID: 27160069 DOI: 10.1016/j.jid.2016.03.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/01/2016] [Accepted: 03/08/2016] [Indexed: 12/19/2022]
Abstract
Up to 18% of melanomas harbor mutations in the neuroblastoma rat-sarcoma homolog (NRAS). Yet, decades of research aimed to interfere with oncogenic RAS signaling have been largely disappointing and have not resulted in meaningful clinical outputs. Recent advances in disease modeling, structural biology, and an improved understanding of RAS cycling as well as RAS signaling networks have renewed hope for developing strategies to selectively block hyperactive RAS function. This review discusses direct and indirect blocking of activated RAS with a focus on current and potential future therapeutic approaches for NRAS mutant melanoma.
Collapse
Affiliation(s)
- Christian Posch
- Department of Dermatology, Mt. Zion Cancer Research Center, University of California San Francisco, San Francisco, California, USA; Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Department of Dermatology, The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna, Vienna, Austria.
| | - Igor Vujic
- Department of Dermatology, Mt. Zion Cancer Research Center, University of California San Francisco, San Francisco, California, USA; Department of Dermatology, The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna, Vienna, Austria
| | - Babak Monshi
- Department of Dermatology, The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna, Vienna, Austria
| | - Martina Sanlorenzo
- Department of Dermatology, Mt. Zion Cancer Research Center, University of California San Francisco, San Francisco, California, USA; Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Felix Weihsengruber
- Department of Dermatology, The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna, Vienna, Austria
| | - Klemens Rappersberger
- Department of Dermatology, The Rudolfstiftung Hospital, Academic Teaching Hospital, Medical University Vienna, Vienna, Austria
| | - Susana Ortiz-Urda
- Department of Dermatology, Mt. Zion Cancer Research Center, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
33
|
Woodfield SE, Zhang L, Scorsone KA, Liu Y, Zage PE. Binimetinib inhibits MEK and is effective against neuroblastoma tumor cells with low NF1 expression. BMC Cancer 2016; 16:172. [PMID: 26925841 PMCID: PMC4772351 DOI: 10.1186/s12885-016-2199-z] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Accepted: 02/17/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Novel therapies are needed for children with high-risk and relapsed neuroblastoma. We hypothesized that MAPK/ERK kinase (MEK) inhibition with the novel MEK1/2 inhibitor binimetinib would be effective in neuroblastoma preclinical models. METHODS Levels of total and phosphorylated MEK and extracellular signal-regulated kinase (ERK) were examined in primary neuroblastoma tumor samples and in neuroblastoma cell lines by Western blot. A panel of established neuroblastoma tumor cell lines was treated with increasing concentrations of binimetinib, and their viability was determined using MTT assays. Western blot analyses were performed to examine changes in total and phosphorylated MEK and ERK and to measure apoptosis in neuroblastoma tumor cells after binimetinib treatment. NF1 protein levels in neuroblastoma cell lines were determined using Western blot assays. Gene expression of NF1 and MEK1 was examined in relationship to neuroblastoma patient outcomes. RESULTS Both primary neuroblastoma tumor samples and cell lines showed detectable levels of total and phosphorylated MEK and ERK. IC50 values for cells sensitive to binimetinib ranged from 8 nM to 1.16 μM, while resistant cells did not demonstrate any significant reduction in cell viability with doses exceeding 15 μM. Sensitive cells showed higher endogenous expression of phosphorylated MEK and ERK. Gene expression of NF1, but not MEK1, correlated with patient outcomes in neuroblastoma, and NF1 protein expression also correlated with responses to binimetinib. CONCLUSIONS Neuroblastoma tumor cells show a range of sensitivities to the novel MEK inhibitor binimetinib. In response to binimetinib, sensitive cells demonstrated complete loss of phosphorylated ERK, while resistant cells demonstrated either incomplete loss of ERK phosphorylation or minimal effects on MEK phosphorylation, suggesting alternative mechanisms of resistance. NF1 protein expression correlated with responses to binimetinib, supporting the use of NF1 as a biomarker to identify patients that may respond to MEK inhibition. MEK inhibition therefore represents a potential new therapeutic strategy for neuroblastoma.
Collapse
Affiliation(s)
- Sarah E Woodfield
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.
| | - Linna Zhang
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.
| | - Kathleen A Scorsone
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA.
| | - Yin Liu
- Department of Neurobiology and Anatomy, The University of Texas Medical School, Houston, TX, USA. .,Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA.
| | - Peter E Zage
- Department of Pediatrics, Section of Hematology-Oncology, Baylor College of Medicine, Houston, TX, USA. .,Texas Children's Cancer Center, Houston, TX, USA.
| |
Collapse
|
34
|
Hudson K, Hancox UJ, Trigwell C, McEwen R, Polanska UM, Nikolaou M, Morentin Gutierrez P, Avivar-Valderas A, Delpuech O, Dudley P, Hanson L, Ellston R, Jones A, Cumberbatch M, Cosulich SC, Ward L, Cruzalegui F, Green S. Intermittent High-Dose Scheduling of AZD8835, a Novel Selective Inhibitor of PI3Kα and PI3Kδ, Demonstrates Treatment Strategies for PIK3CA-Dependent Breast Cancers. Mol Cancer Ther 2016; 15:877-89. [PMID: 26839307 DOI: 10.1158/1535-7163.mct-15-0687] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 01/25/2016] [Indexed: 11/16/2022]
Abstract
The PIK3CA gene, encoding the p110α catalytic unit of PI3Kα, is one of the most frequently mutated oncogenes in human cancer. Hence, PI3Kα is a target subject to intensive efforts in identifying inhibitors and evaluating their therapeutic potential. Here, we report studies with a novel PI3K inhibitor, AZD8835, currently in phase I clinical evaluation. AZD8835 is a potent inhibitor of PI3Kα and PI3Kδ with selectivity versus PI3Kβ, PI3Kγ, and other kinases that preferentially inhibited growth in cells with mutant PIK3CA status, such as in estrogen receptor-positive (ER(+)) breast cancer cell lines BT474, MCF7, and T47D (sub-μmol/L GI50s). Consistent with this, AZD8835 demonstrated antitumor efficacy in corresponding breast cancer xenograft models when dosed continuously. In addition, an alternative approach of intermittent high-dose scheduling (IHDS) was explored given our observations that higher exposures achieved greater pathway inhibition and induced apoptosis. Indeed, using IHDS, monotherapy AZD8835 was able to induce tumor xenograft regression. Furthermore, AZD8835 IHDS in combination with other targeted therapeutic agents further enhanced antitumor activity (up to 92% regression). Combination partners were prioritized on the basis of our mechanistic insights demonstrating signaling pathway cross-talk, with a focus on targeting interdependent ER and/or CDK4/6 pathways or alternatively a node (mTOR) in the PI3K-pathway, approaches with demonstrated clinical benefit in ER(+) breast cancer patients. In summary, AZD8835 IHDS delivers strong antitumor efficacy in a range of combination settings and provides a promising alternative to continuous dosing to optimize the therapeutic index in patients. Such schedules merit clinical evaluation. Mol Cancer Ther; 15(5); 877-89. ©2016 AACR.
Collapse
Affiliation(s)
- Kevin Hudson
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom.
| | - Urs J Hancox
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Cath Trigwell
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Robert McEwen
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Urszula M Polanska
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Myria Nikolaou
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | | | | | - Oona Delpuech
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Phillippa Dudley
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Lyndsey Hanson
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Rebecca Ellston
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Alys Jones
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Marie Cumberbatch
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Sabina C Cosulich
- AstraZeneca Pharmaceuticals, CRUK-CI Li Ka Shing Centre, Cambridge, United Kingdom
| | - Lara Ward
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Francisco Cruzalegui
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| | - Stephen Green
- AstraZeneca Pharmaceuticals, Oncology iMed, Macclesfield, Cheshire, United Kingdom
| |
Collapse
|
35
|
Caunt CJ, Sale MJ, Smith PD, Cook SJ. MEK1 and MEK2 inhibitors and cancer therapy: the long and winding road. Nat Rev Cancer 2015; 15:577-92. [PMID: 26399658 DOI: 10.1038/nrc4000] [Citation(s) in RCA: 436] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The role of the ERK signalling pathway in cancer is thought to be most prominent in tumours in which mutations in the receptor tyrosine kinases RAS, BRAF, CRAF, MEK1 or MEK2 drive growth factor-independent ERK1 and ERK2 activation and thence inappropriate cell proliferation and survival. New drugs that inhibit RAF or MEK1 and MEK2 have recently been approved or are currently undergoing late-stage clinical evaluation. In this Review, we consider the ERK pathway, focusing particularly on the role of MEK1 and MEK2, the 'gatekeepers' of ERK1/2 activity. We discuss their validation as drug targets, the merits of targeting MEK1 and MEK2 versus BRAF and the mechanisms of action of different inhibitors of MEK1 and MEK2. We also consider how some of the systems-level properties (intrapathway regulatory loops and wider signalling network connections) of the ERK pathway present a challenge for the success of MEK1 and MEK2 inhibitors, discuss mechanisms of resistance to these inhibitors, and review their clinical progress.
Collapse
Affiliation(s)
- Christopher J Caunt
- Department of Biology and Biochemistry, University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Matthew J Sale
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Paul D Smith
- AstraZeneca, Oncology iMed, Cancer Biosciences, Cancer Research UK, Li Ka Shing Centre, Cambridge Institute, Robinson Way, Cambridge CB2 0RE, UK
| | - Simon J Cook
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge CB22 3AT, UK
| |
Collapse
|
36
|
Successful use of next generation genomic sequencing (NGS)-directed therapy of clear cell carcinoma of the ovary (CCCO) with trametinib and metformin in a patient with chemotherapy-refractory disease. GYNECOLOGIC ONCOLOGY RESEARCH AND PRACTICE 2015; 2:4. [PMID: 27231564 PMCID: PMC4880882 DOI: 10.1186/s40661-015-0013-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 08/18/2015] [Indexed: 11/11/2022]
Abstract
Purpose Compared to other subtypes of epithelial ovarian cancer, clear cell carcinoma of the ovary bears an ominous reputation for chemotherapy resistance, increased relapse rate, and diminished survival. Among patients with distinct histopathologic subtypes, molecular analyses have identified a variety of known drivers of the malignant behavior, and depict a striking heterogeneity. Methods A patient with rapidly metastatic CCCO that was refractory to taxane, platinum, pemetrexed, and bevacizumab-based strategies underwent molecular profiling which disclosed dual MAPK and PI3K/AKT/mTOR pathway mutations. Results Combined targeted therapy with trametinib and metformin resulted in a dramatic disease regression without toxicity. Conclusion The case highlights the utility of precision medicine combining individual molecular diagnosis with rational therapeutic intervention with targeted agents.
Collapse
|
37
|
García-García C, Rivas MA, Ibrahim YH, Calvo MT, Gris-Oliver A, Rodríguez O, Grueso J, Antón P, Guzmán M, Aura C, Nuciforo P, Jessen K, Argilés G, Dienstmann R, Bertotti A, Trusolino L, Matito J, Vivancos A, Chicote I, Palmer HG, Tabernero J, Scaltriti M, Baselga J, Serra V. MEK plus PI3K/mTORC1/2 Therapeutic Efficacy Is Impacted by TP53 Mutation in Preclinical Models of Colorectal Cancer. Clin Cancer Res 2015; 21:5499-5510. [PMID: 26272063 DOI: 10.1158/1078-0432.ccr-14-3091] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 07/07/2015] [Indexed: 02/05/2023]
Abstract
PURPOSE PI3K pathway activation occurs in concomitance with RAS/BRAF mutations in colorectal cancer, limiting the sensitivity to targeted therapies. Several clinical studies are being conducted to test the tolerability and clinical activity of dual MEK and PI3K pathway blockade in solid tumors. EXPERIMENTAL DESIGN In the present study, we explored the efficacy of dual pathway blockade in colorectal cancer preclinical models harboring concomitant activation of the ERK and PI3K pathways. Moreover, we investigated if TP53 mutation affects the response to this therapy. RESULTS Dual MEK and mTORC1/2 blockade resulted in synergistic antiproliferative effects in cell lines bearing alterations in KRAS/BRAF and PIK3CA/PTEN. Although the on-treatment cell-cycle effects were not affected by the TP53 status, a marked proapoptotic response to therapy was observed exclusively in wild-type TP53 colorectal cancer models. We further interrogated two independent panels of KRAS/BRAF- and PIK3CA/PTEN-altered cell line- and patient-derived tumor xenografts for the antitumor response toward this combination of agents. A combination response that resulted in substantial antitumor activity was exclusively observed among the wild-type TP53 models (two out of five, 40%), but there was no such response across the eight mutant TP53 models (0%). Interestingly, within a cohort of 14 patients with colorectal cancer treated with these agents for their metastatic disease, two patients with long-lasting responses (32 weeks) had TP53 wild-type tumors. CONCLUSIONS Our data support that, in wild-type TP53 colorectal cancer cells with ERK and PI3K pathway alterations, MEK blockade results in potent p21 induction, preventing apoptosis to occur. In turn, mTORC1/2 inhibition blocks MEK inhibitor-mediated p21 induction, unleashing apoptosis. Clin Cancer Res; 21(24); 5499-510. ©2015 AACR.
Collapse
Affiliation(s)
- Celina García-García
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Martín A Rivas
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Yasir H Ibrahim
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - María Teresa Calvo
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Albert Gris-Oliver
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Olga Rodríguez
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Judit Grueso
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Pilar Antón
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Marta Guzmán
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Claudia Aura
- Molecular Pathology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Pathology Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | | | - Guillem Argilés
- Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Rodrigo Dienstmann
- Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Andrea Bertotti
- Department of Oncology, University of Torino School of Medicine, 10060 Candiolo, Torino, Italy.,Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Livio Trusolino
- Department of Oncology, University of Torino School of Medicine, 10060 Candiolo, Torino, Italy.,Translational Cancer Medicine, Candiolo Cancer Institute - FPO IRCCS, 10060 Candiolo, Torino, Italy
| | - Judit Matito
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Ana Vivancos
- Cancer Genomics Group, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Irene Chicote
- Stem Cells and Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Héctor G Palmer
- Stem Cells and Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| | - Josep Tabernero
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain.,Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain
| | - Maurizio Scaltriti
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - José Baselga
- Human Oncology & Pathogenesis Program, Memorial Sloan-Kettering Cancer Center, New York, NY 10065.,Breast Medicine Service, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10065
| | - Violeta Serra
- Experimental Therapeutics Laboratory, Vall d'Hebron Institute of Oncology (VHIO), 08035-Barcelona, Spain
| |
Collapse
|
38
|
Jokinen E, Koivunen JP. MEK and PI3K inhibition in solid tumors: rationale and evidence to date. Ther Adv Med Oncol 2015; 7:170-80. [PMID: 26673580 PMCID: PMC4406912 DOI: 10.1177/1758834015571111] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
PI3K-AKT-mTOR and Ras-Raf-MEK-ERK are the most commonly altered oncogenic pathways in solid malignancies. There has been a lot of enthusiasm to develop inhibitors to these pathways for cancer therapy. Unfortunately, the antitumor activities of single-agent therapies have generally been disappointing, excluding B-Raf mutant melanoma and renal cell cancer. Preclinical studies have suggested that concurrent targeting of the PI3K-AKT-mTOR and Ras-Raf-MEK-ERK pathways is an active combination in various solid malignancies. In the current work, we review the preclinical data of the PI3K and MEK dual targeting as a cancer therapy and the results of early-phase clinical trials, and propose future directions.
Collapse
Affiliation(s)
- E Jokinen
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland, PB20, 90029 OYS
| | - J P Koivunen
- Department of Oncology and Radiotherapy, Oulu University Hospital, Oulu, Finland
| |
Collapse
|