1
|
Liu L, Mao Y, Guo L, Li C, Wang Y. Advances in adjuvant therapy for operable N2 non-small cell lung cancer: a narrative review. Front Oncol 2025; 14:1523743. [PMID: 39906659 PMCID: PMC11790610 DOI: 10.3389/fonc.2024.1523743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) is still the disease with the highest incidence rate among malignant tumors, in which NSCLC under N2 stage has obvious survival differences among different patients due to its high heterogeneity. For NSCLC under this stage, the current treatment options are: preoperative neoadjuvant therapy, surgical treatment, postoperative adjuvant chemotherapy, postoperative adjuvant radiotherapy (PORT), Postoperative adjuvant targeted therapy and postoperative adjuvant immunotherapy. Whether postoperative adjuvant radiotherapy is routinely administered to patients with pN2 remains controversial in clinical application. Meanwhile, the booming development of adjuvant targeted therapy and adjuvant immunotherapy also provides newer therapeutic options for the prognosis of postoperative pN2 stage NSCLC, and some new markers will guide the adaptive application of immune drugs in the future. This article analyzes the current stage of therapeutic advances in operable stage N2 non-small cell lung cancer, and discusses in detail in this article the therapeutic controversy of postoperative adjuvant radiotherapy in pN2 stage non-small cell lung cancer, so as to explore a more reasonable treatment mode for future patients with stage N2 non-small cell lung cancer.
Collapse
Affiliation(s)
| | | | | | | | - Yiqian Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
2
|
Sheikh SR, Klesse LJ, Mangum R, Bui A, Siegel BI, Abdelbaki MS, Patel NJ. The role of MEK inhibition in pediatric low-grade gliomas. Front Oncol 2024; 14:1503894. [PMID: 39759151 PMCID: PMC11695311 DOI: 10.3389/fonc.2024.1503894] [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: 09/29/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumors in children. Many patients with unresectable tumors experience recurrence or long-term sequelae from standard chemotherapeutics. This mini-review explores the emerging role of MEK inhibitors in the management of pLGGs, highlighting their potential to transform current treatment paradigms. We review the molecular basis for therapeutic MEK inhibition in the context of pLGG, provide an evidence base for the use of the major MEK inhibitors currently available in the market for pLGG, and review the challenges in the use of MEKi inhibitors in this population.
Collapse
Affiliation(s)
- Shehryar R. Sheikh
- Department of Neurosurgery, Cleveland Clinic, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, OH, United States
| | - Laura J. Klesse
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Ross Mangum
- Center for Cancer and Blood Disorders, Phoenix Children’s Hospital, Phoenix, AZ, United States
| | - Ashley Bui
- Department of Pediatrics, Division of Hematology/Oncology, UT Southwestern Medical Center, Dallas, TX, United States
| | - Benjamin I. Siegel
- Brain Tumor Institute, Children’s National Hospital, Washington, DC, United States
- Division of Neurology, Children’s National Hospital, Washington, DC, United States
| | - Mohamed S. Abdelbaki
- Division of Hematology and Oncology, Department of Pediatrics, School of Medicine, Washington University, St. Louis, MO, United States
| | - Neha J. Patel
- Department of Pediatric Hematology-Oncology and Blood and Marrow Transplant, Cleveland Clinic, Cleveland, OH, United States
| |
Collapse
|
3
|
Bortolot M, Torresan S, De Carlo E, Bertoli E, Stanzione B, Del Conte A, Spina M, Bearz A. Navigating Therapeutic Challenges in BRAF-Mutated NSCLC: Non-V600 Mutations, Immunotherapy, and Overcoming Resistance. Int J Mol Sci 2024; 25:12972. [PMID: 39684685 DOI: 10.3390/ijms252312972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/27/2024] [Accepted: 11/30/2024] [Indexed: 12/18/2024] Open
Abstract
Although rare in non-small cell lung cancer (NSCLC), BRAF mutations present considerable therapeutic challenges. While the use of BRAF and MEK inhibitor combinations has significantly improved survival outcomes in patients with BRAF V600E mutations, no targeted therapies are currently available for class II and III mutations, leaving the optimal treatment strategy and prognosis for these patients uncertain. Additionally, despite immunotherapy typically showing limited benefit in patients with other activating genomic alterations, it appears to deliver comparable efficacy in BRAF-mutated NSCLC, emerging as a potentially viable treatment option, particularly in patients with a history of smoking. However, resistance to BRAF pathway inhibitors is inevitable, leading to disease progression, and a well-defined strategy to overcome these resistance mechanisms is lacking. This review aims to explore the critical challenges in the management of BRAF-mutated NSCLC, providing a comprehensive summary of the current evidence and highlighting ongoing clinical trials that aim to address these critical gaps.
Collapse
Affiliation(s)
- Martina Bortolot
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Sara Torresan
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
4
|
Stoff R, Markovic SN, McWilliams RR, Kottschade LA, Montane HN, Dimou A, Dudek AZ, Tan W, Dronca RS, Seetharam M, Chen R, Block MS. Real-world evidence on efficacy and toxicity of targeted therapy in older melanoma patients treated in a tertiary-hospital setting. Melanoma Res 2024; 34:510-518. [PMID: 39207855 PMCID: PMC11524625 DOI: 10.1097/cmr.0000000000000997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Melanoma is the deadliest form of skin cancer. The median age at diagnosis is 66. While most patients are treated with immunotherapy, the use of targeted therapy is a valid alternative for patients whose tumors harbor a BRAF or c-KIT driver mutation. These agents, while effective, come with a variety of side effects which limit their use, especially in older patients. We sought to assess the efficacy and toxicity of these agents in older melanoma patients. Melanoma patients over 65 treated with BRAF/MEK or c-KIT inhibitors were retrospectively identified, and their data were analyzed for treatment efficacy and toxicity. All data were compared using the Chi-square test for categorical comparisons and the Kruskal-Wallis method for median comparisons. One hundred and sixteen patients were identified. One hundred and six patients were treated with BRAF/MEK inhibitors. The assessed response rate (RR) was 83% and was comparable across different subgroups, including advanced line patients and those with a more aggressive disease. The median progression free survival (PFS) was 7.9 months, and the median overall survival (OS) was 15.7 months. Twenty-seven percent experienced grade 3-4 toxicity leading to a 24% treatment discontinuation rate. Another 10 patients were treated with the c-KIT inhibitor imatinib, for whom the assessed RR was 55%. The median PFS was 4.3 months, and the median OS was 22.6 months. Forty percent needed dose reductions, yet none had to stop treatment due to adverse effects. The use of targeted therapy in older patients is effective yet challenging due to toxicity. Deploying mitigation strategies can help maximizing their usefulness.
Collapse
Affiliation(s)
- Ronen Stoff
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Svetomir N. Markovic
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Robert R. McWilliams
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Lisa A. Kottschade
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Heather N. Montane
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Anastasios Dimou
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Arkadiusz Z. Dudek
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| | - Winston Tan
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Roxana S. Dronca
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Mahesh Seetharam
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Scottsdale, Arizona, USA
| | - Ruqin Chen
- Department of Hematology and Oncology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, Florida
| | - Matthew S. Block
- Department of Medical Oncology, Mayo Clinic Comprehensive Cancer Center, Rochester, Minnesota
| |
Collapse
|
5
|
Inoue H, Kuroda JI, Fujioka Y, Hata N, Mizoguchi M, Yoshii D, Sueyoshi H, Takeshima Y, Fujimoto K, Shinojima N, Sunami K, Mikami Y, Nakamura H, Mukasa A. Drug-resistant BRAF V600E-mutant recurrent pleomorphic xanthoastrocytoma, CNS WHO Grade 3 successfully resolved with incidental discontinuation of combined BRAF and MEK inhibitor therapy. Surg Neurol Int 2024; 15:417. [PMID: 39640311 PMCID: PMC11618650 DOI: 10.25259/sni_734_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 12/07/2024] Open
Abstract
Background Combination therapy with BRAF and MEK inhibitor holds promise for treating gliomas harboring the BRAF V600E mutation; however, the development of acquired resistance remains a challenge. Case Description We describe a case of repeated recurrent BRAF-mutant pleomorphic xanthoastrocytoma (central nervous system World Health Organization grade 3) treated with combination therapy with BRAF and MEK inhibitor. The patient received dabrafenib (BRAF inhibitor) and trametinib (MEK inhibitor); however, she developed resistance to the combination therapy. Remarkably, incidental drug discontinuation contributed to the disappearance of the resistant tumor. The same phenomenon was repeatedly observed after that. Genetic analysis demonstrated that the resistant tumor had BRAF V600E amplification; the resistant tumor remained BRAF→MEK→ERK pathway dependent, and drug resistance might be due to elevated BRAF V600E expression. We speculated that ERK1/2 signal extremes caused by the discontinuation of the combination therapy affected the resistant tumor survival. Conclusion This case study provides important insights into novel treatment strategies and their underlying mechanisms for gliomas with BRAF mutations.
Collapse
Affiliation(s)
- Hirotaka Inoue
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Jun-Ichiro Kuroda
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Yutaka Fujioka
- Department of Neurosurgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nobuhiro Hata
- Department of Neurosurgery, Oita University Faculty of Medicine, Yufu, Japan
| | | | - Daiki Yoshii
- Department of Diagnostic Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Sueyoshi
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Yuki Takeshima
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Kenji Fujimoto
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Naoki Shinojima
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Chuo-ku, Japan
| | - Yoshiki Mikami
- Department of Diagnostic Pathology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hideo Nakamura
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan
| | - Akitake Mukasa
- Department of Neurosurgery, Kumamoto University Hospital, Kumamoto, Japan
| |
Collapse
|
6
|
Smith-Uffen M, Park J, Parsonson A, Kiely BE, Vasista A. Estimating scenarios for survival time in patients with advanced melanoma receiving immunotherapy and targeted therapy. Oncologist 2024; 29:922-930. [PMID: 38768122 PMCID: PMC11546645 DOI: 10.1093/oncolo/oyae089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 04/15/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND We aim to provide survival scenario estimates for patients with advanced melanoma starting targeted therapies and immunotherapies. MATERIALS AND METHODS We sought randomized trials of targeted therapies and immunotherapies for advanced melanoma and recorded the following percentiles (represented survival scenario) from each overall survival (OS) curve: 90th (worst-case), 75th (lower-typical), 50th (median), 25th (upper-typical), and 10th (best-case). We tested whether these scenarios can be estimated for each OS curve by multiplying its median by 4 multiples: 0.25 (worst-case), 0.5 (lower-typical), 2 (upper-typical), and 3 (best-case). RESULTS We identified 15 trials with 8025 patients. For first-line combination targeted therapy treatment groups, the median (interquartile range, IQR) in months for each percentile was: 90th, 6.2 (6.0-6.5); 75th, 11.3 (11.3-11.4); and median, 24.4 (23.5-25.3). For the first-line combination immunotherapy treatment group, the percentiles in months were: 90th, 3.9 (2.8-4.5); 75th, 13.4 (10.1-15.4), median 73 (not applicable). In targeted therapy groups, simple multiples of the median OS were accurate for estimating the 90th percentile in 80%; 75th percentile in 40%; 25th percentile in 100%. In immunotherapy groups, these multiples were accurate at 0% for the 90th percentile, and 43% for the 75th percentile. The 90th percentile (worst-case scenario) was better estimated as 1/6× median OS, and the 75th percentile (lower-typical) as 1/3× median OS. CONCLUSIONS Simple multiples of the median OS are a useful framework to estimate scenarios for survival for patients receiving targeted therapies, not immunotherapy. Longer follow-up is required to estimate upper-typical and best-case scenarios.
Collapse
Affiliation(s)
| | - John Park
- Department of Medical Oncology, Nepean Cancer Care Centre, Kingswood, NSW, Australia
| | - Andrew Parsonson
- Department of Medical Oncology, Nepean Cancer Care Centre, Kingswood, NSW, Australia
| | - Belinda E Kiely
- NHMRC Clinical trials Centre, University of Sydney, Camperdown, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
| | - Anuradha Vasista
- Department of Medical Oncology, Nepean Cancer Care Centre, Kingswood, NSW, Australia
- Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- The Crown Princess Mary Cancer Centre, Westmead Hospital, Westmead, NSW, Australia
| |
Collapse
|
7
|
Coelho MA, Strauss ME, Watterson A, Cooper S, Bhosle S, Illuzzi G, Karakoc E, Dinçer C, Vieira SF, Sharma M, Moullet M, Conticelli D, Koeppel J, McCarten K, Cattaneo CM, Veninga V, Picco G, Parts L, Forment JV, Voest EE, Marioni JC, Bassett A, Garnett MJ. Base editing screens define the genetic landscape of cancer drug resistance mechanisms. Nat Genet 2024; 56:2479-2492. [PMID: 39424923 PMCID: PMC11549056 DOI: 10.1038/s41588-024-01948-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 09/13/2024] [Indexed: 10/21/2024]
Abstract
Drug resistance is a principal limitation to the long-term efficacy of cancer therapies. Cancer genome sequencing can retrospectively delineate the genetic basis of drug resistance, but this requires large numbers of post-treatment samples to nominate causal variants. Here we prospectively identify genetic mechanisms of resistance to ten oncology drugs from CRISPR base editing mutagenesis screens in four cancer cell lines using a guide RNA library predicted to install 32,476 variants in 11 cancer genes. We identify four functional classes of protein variants modulating drug sensitivity and use single-cell transcriptomics to reveal how these variants operate through distinct mechanisms, including eliciting a drug-addicted cell state. We identify variants that can be targeted with alternative inhibitors to overcome resistance and functionally validate an epidermal growth factor receptor (EGFR) variant that sensitizes lung cancer cells to EGFR inhibitors. Our variant-to-function map has implications for patient stratification, therapy combinations and drug scheduling in cancer treatment.
Collapse
Affiliation(s)
- Matthew A Coelho
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.
- Cancer Genome Editing, Wellcome Sanger Institute, Hinxton, UK.
- Open Targets, Cambridge, UK.
| | - Magdalena E Strauss
- EMBL-European Bioinformatics Institute, Cambridge, UK
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
- Department of Mathematics and Statistics, University of Exeter, Exeter, UK
| | - Alex Watterson
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Sarah Cooper
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
| | - Shriram Bhosle
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | | | - Emre Karakoc
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Cansu Dinçer
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Sara F Vieira
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Mamta Sharma
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Marie Moullet
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Daniela Conticelli
- Department of Oncology, University of Turin, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy
| | - Jonas Koeppel
- Generative and Synthetic Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Katrina McCarten
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
| | - Chiara M Cattaneo
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Experimental Hematology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vivien Veninga
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Gabriele Picco
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK
- Open Targets, Cambridge, UK
| | - Leopold Parts
- Generative and Synthetic Genomics, Wellcome Sanger Institute, Hinxton, UK
| | | | - Emile E Voest
- Department of Immunology and Molecular Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
- Oncode Institute, Utrecht, the Netherlands
| | - John C Marioni
- EMBL-European Bioinformatics Institute, Cambridge, UK
- Cancer Research UK, Cambridge Institute, University of Cambridge, Cambridge, UK
- Genentech, South San Francisco, CA, USA
| | - Andrew Bassett
- Gene Editing and Cellular Research and Development, Wellcome Sanger Institute, Hinxton, UK
| | - Mathew J Garnett
- Translational Cancer Genomics, Wellcome Sanger Institute, Hinxton, UK.
- Open Targets, Cambridge, UK.
| |
Collapse
|
8
|
Schmidlin K, Apodaca S, Newell D, Sastokas A, Kinsler G, Geiler-Samerotte K. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. eLife 2024; 13:RP94144. [PMID: 39255191 PMCID: PMC11386965 DOI: 10.7554/elife.94144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Kara Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Sam Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Daphne Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Alexander Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| | - Grant Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, United States
| | - Kerry Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, United States
- School of Life Sciences, Arizona State University, Tempe, United States
| |
Collapse
|
9
|
Nakano E, Takahashi A, Ogata D, Namikawa K, Yamazaki N. Real-world efficacy and safety of BRAF-targeted therapy for patients with advanced melanoma: A single-center retrospective study in Japan. J Dermatol 2024; 51:1199-1207. [PMID: 38716655 DOI: 10.1111/1346-8138.17263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/10/2024] [Accepted: 04/21/2024] [Indexed: 09/03/2024]
Abstract
Most clinical trials investigating targeted therapies for patients harboring BRAF V600 mutations have included mostly White patients, and data for Asian patients are scarce. Although there are several retrospective studies in Japanese patients, they have investigated only the dabrafenib + trametinib regimen, and have had a short follow-up period. We conducted a single-center retrospective study to update previous studies and compare the outcomes with those in White patients. We analyzed 89 patients who received dabrafenib + trametinib or encorafenib + binimetinib, including 11 who received both treatment regimens. The overall response rate was 79.8%, with complete response in 25 patients (28.1%) and partial response in 45 patients (51.7%). The median progression-free survival was 13.7 months, and the median overall survival was 32.9 months. The 3-year progression-free and overall survival rates were 31.8% and 47.9%, respectively. Although the two regimens showed no significant differences in efficacy, their safety profiles differed, as reported in clinical trials. Therefore, the most frequent adverse event associated with the dabrafenib + trametinib regimen was pyrexia (61.3%) and that of encorafenib + binimetinib was blurred vision (32.0%). Switching directly to another targeted therapy after progressive disease showed no clinical response; however, rechallenge followed by immune checkpoint inhibitor therapy showed a certain response. As a prognostic factor, performance status was associated with progression-free survival, and performance status, serum lactate dehydrogenase level, and dose interruption were associated with overall survival in the multivariate analysis. Real-world data on targeted therapy for patients with melanoma in Japan suggest that both dabrafenib + trametinib and encorafenib + binimetinib show similar efficacy and safety in Asian and White patients.
Collapse
Affiliation(s)
- Eiji Nakano
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Takahashi
- Department of Dermatologic Oncology, National Cancer Center Hospital East, Chiba, Japan
| | - Dai Ogata
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Kenjiro Namikawa
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Department of Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
10
|
Wang M, Sullivan RJ, Mooradian MJ. Toxicities from BRAF and MEK Inhibitors: Strategies to Maximize Therapeutic Success. Curr Oncol Rep 2024; 26:934-944. [PMID: 38850505 DOI: 10.1007/s11912-024-01544-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2024] [Indexed: 06/10/2024]
Abstract
PURPOSE OF REVIEW This report highlights several of the recent therapeutic advancements in the treatment of BRAF-mutant tumors, discusses the most common adverse events observed with BRAF-targeted agents, and suggests strategies to manage and mitigate treatment-related toxicities. RECENT FINDINGS BRAF and MEK inhibitors represent a significant advancement in the treatment of BRAF-mutated malignancies with data across tumor types demonstrating the anti-tumor efficacy of dual MAPK inhibition. Although these agents have a reasonable toxicity profile, variable side effects across organ systems can develop. The discovery of activating BRAF mutations and subsequent development of BRAF and MEK inhibitors has transformed the treatment algorithms of BRAF-mutant malignancies. With increased application of these targeted regimens, identification and prompt management of their unique adverse events are crucial.
Collapse
Affiliation(s)
- Mike Wang
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
| | - Ryan J Sullivan
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Meghan J Mooradian
- Division of Medical Oncology, Massachusetts General Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
11
|
Dong S, Wang Z, Zhang JT, Yan B, Zhang C, Gao X, Sun H, Li YS, Yan HH, Tu HY, Liu SYM, Gong Y, Gao W, Huang J, Liao RQ, Lin JT, Ke EE, Xu Z, Zhang X, Xia X, Li AN, Liu SY, Pan Y, Yang JJ, Zhong WZ, Yi X, Zhou Q, Yang XN, Wu YL. Circulating Tumor DNA-Guided De-Escalation Targeted Therapy for Advanced Non-Small Cell Lung Cancer: A Nonrandomized Controlled Trial. JAMA Oncol 2024; 10:932-940. [PMID: 38869865 DOI: 10.1001/jamaoncol.2024.1779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
Importance Uninterrupted targeted therapy until disease progression or intolerable toxic effects is currently the routine therapy for advanced non-small cell lung cancer (NSCLC) involving driver gene variations. However, drug resistance is inevitable. Objective To assess the clinical feasibility of adaptive de-escalation tyrosine kinase inhibitor (TKI) treatment guided by circulating tumor DNA (ctDNA) for achieving complete remission after local consolidative therapy (LCT) in patients with advanced NSCLC. Design, Setting, and Participants This prospective nonrandomized controlled trial was conducted at a single center from June 3, 2020, to July 19, 2022, and included 60 patients with advanced NSCLC with driver variations without radiologically detectable disease after TKI and LCT. The median (range) follow-up time was 19.2 (3.8-29.7) months. Data analysis was conducted from December 15, 2022, to May 10, 2023. Intervention Cessation of TKI treatment and follow-up every 3 months. Treatment was restarted in patients with progressive disease (defined by the Response Evaluation Criteria in Solid Tumors 1.1 criteria), detectable ctDNA, or elevated carcinoembryonic antigen (CEA) levels, whichever manifested first, and treatment ceased if all indicators were negative during follow-up surveillance. Main Outcomes and Measures Progression-free survival (PFS). Secondary end points were objective response rate, time to next treatment, and overall survival. Results Among the total study sample of 60 participants (median [range] age, 55 [21-75] years; 33 [55%] were female), the median PFS was 18.4 (95% CI, 12.6-24.2) months and the median (range) total treatment break duration was 9.1 (1.5-28.1) months. Fourteen patients (group A) remained in TKI cessation with a median (range) treatment break duration of 20.3 (6.8-28.1) months; 31 patients (group B) received retreatment owing to detectable ctDNA and/or CEA and had a median PFS of 20.2 (95% CI, 12.9-27.4) months with a median (range) total treatment break duration of 8.8 (1.5-20.6) months; and 15 patients (group C) who underwent retreatment with TKIs due to progressive disease had a median PFS of 5.5 (95% CI, 1.5-7.2) months. For all participants, the TKI retreatment response rate was 96%, the median time to next treatment was 29.3 (95% CI, 25.3-35.2) months, and the data for overall survival were immature. Conclusions and Relevance The findings of this nonrandomized controlled trial suggest that this adaptive de-escalation TKI strategy for patients with NSCLC is feasible in those with no lesions after LCT and a negative ctDNA test result. This might provide a de-escalation treatment strategy guided by ctDNA for the subset of patients with advanced NSCLC. Trial Registration ClinicalTrials.gov Identifier: NCT03046316.
Collapse
Affiliation(s)
- Song Dong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zhen Wang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jia-Tao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Bingfa Yan
- Geneplus-Beijing Institute, Beijing, China
| | - Chao Zhang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xuan Gao
- Geneplus-Beijing Institute, Beijing, China
| | - Hao Sun
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yang-Si Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hong-Hong Yan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hai-Yan Tu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Si-Yang Maggie Liu
- Department of Hematology, First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| | - Yuhua Gong
- Geneplus-Beijing Institute, Beijing, China
| | - Wei Gao
- Geneplus-Beijing Institute, Beijing, China
| | - Jie Huang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Ri-Qiang Liao
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jun-Tao Lin
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - E-E Ke
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zelong Xu
- Geneplus-Beijing Institute, Beijing, China
| | - Xue Zhang
- Geneplus-Beijing Institute, Beijing, China
| | | | - An-Na Li
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Si-Yang Liu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi Pan
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jin-Ji Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Wen-Zhao Zhong
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Xin Yi
- Geneplus-Beijing Institute, Beijing, China
| | - Qing Zhou
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| | - Xue-Ning Yang
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yi-Long Wu
- Guangdong Lung Cancer Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- Chinese Thoracic Oncology Group, Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Donzé C, Leenhardt F, Vinches M, Eberlé MC, Fersing C. Clinical Pharmacy Initiatives Contribute to the Excellent Efficacy of the Dabrafenib/Trametinib Combination for Iodine-Refractory Thyroid Carcinoma: A Case Report. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1037. [PMID: 39064466 PMCID: PMC11278742 DOI: 10.3390/medicina60071037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/09/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024]
Abstract
A 76-year-old female patient presented with an iodine-refractory papillary thyroid carcinoma (PTC), diagnosed eight years earlier, with several lymph node recurrences requiring successive surgeries. Fluorodeoxyglucose ([18F]FDG) positron emission tomography/computed tomography (PET/CT) imaging revealed a new unresectable loco-regional recurrence. The patient was diagnosed with a somatic BRAF V600E mutation. Therefore, dabrafenib and trametinib combination therapy was introduced and closely monitored by a dedicated multidisciplinary team, involving pharmaceutical consultations. As early as six weeks after treatment initiation, the patient reported multiple adverse events (AEs) to the clinical pharmacy team, who provided advice on resolving AEs or improving tolerance. Close interprofessional collaboration among healthcare workers involved in the care pathway allowed for the identification of the most opportune times for temporary suspension of treatment (four suspensions over seven months) or dose reduction (two reductions over 3.5 months). This resulted in a total treatment duration (one year) longer than the average times reported in the literature. The patient showed a rapid and excellent response to treatment immediately after initiation, culminating in a complete metabolic response assessed by [18F]FDG PET/CT imaging at nine months. Twenty-five months after treatment discontinuation, the disease remained controlled. Overall, dabrafenib and trametinib combination could offer excellent outcomes in selected patients with refractory BRAF-mutated PTC, with additional clinical pharmacy initiatives allowing for the optimized management of AEs and prolonged treatment periods.
Collapse
Affiliation(s)
- Charlotte Donzé
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
| | - Fanny Leenhardt
- Pharmacy Department, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, University of Montpellier, Institut Régional du Cancer de Montpellier (ICM), 34090 Montpellier, France
| | - Marie Vinches
- Medical Oncology Department, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
| | - Marie-Claude Eberlé
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
| | - Cyril Fersing
- Nuclear Medicine Department, Institut Régional du Cancer de Montpellier (ICM), University of Montpellier, 34298 Montpellier, France
- IBMM, Univ Montpellier, CNRS, ENSCM, 34293 Montpellier, France
| |
Collapse
|
13
|
Schmidlin, Apodaca, Newell, Sastokas, Kinsler, Geiler-Samerotte. Distinguishing mutants that resist drugs via different mechanisms by examining fitness tradeoffs. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.17.562616. [PMID: 37905147 PMCID: PMC10614906 DOI: 10.1101/2023.10.17.562616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
There is growing interest in designing multidrug therapies that leverage tradeoffs to combat resistance. Tradeoffs are common in evolution and occur when, for example, resistance to one drug results in sensitivity to another. Major questions remain about the extent to which tradeoffs are reliable, specifically, whether the mutants that provide resistance to a given drug all suffer similar tradeoffs. This question is difficult because the drug-resistant mutants observed in the clinic, and even those evolved in controlled laboratory settings, are often biased towards those that provide large fitness benefits. Thus, the mutations (and mechanisms) that provide drug resistance may be more diverse than current data suggests. Here, we perform evolution experiments utilizing lineage-tracking to capture a fuller spectrum of mutations that give yeast cells a fitness advantage in fluconazole, a common antifungal drug. We then quantify fitness tradeoffs for each of 774 evolved mutants across 12 environments, finding these mutants group into 6 classes with characteristically different tradeoffs. Their unique tradeoffs may imply that each group of mutants affects fitness through different underlying mechanisms. Some of the groupings we find are surprising. For example, we find some mutants that resist single drugs do not resist their combination, while others do. And some mutants to the same gene have different tradeoffs than others. These findings, on one hand, demonstrate the difficulty in relying on consistent or intuitive tradeoffs when designing multidrug treatments. On the other hand, by demonstrating that hundreds of adaptive mutations can be reduced to a few groups with characteristic tradeoffs, our findings may yet empower multidrug strategies that leverage tradeoffs to combat resistance. More generally speaking, by grouping mutants that likely affect fitness through similar underlying mechanisms, our work guides efforts to map the phenotypic effects of mutation.
Collapse
Affiliation(s)
- Schmidlin
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Apodaca
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Newell
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Sastokas
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| | - Kinsler
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA
| | - Geiler-Samerotte
- Biodesign Center for Mechanisms of Evolution, Arizona State University, Tempe, AZ
- School of Life Sciences, Arizona State University, Tempe AZ
| |
Collapse
|
14
|
Adamopoulos C, Papavassiliou KA, Poulikakos PI, Papavassiliou AG. RAF and MEK Inhibitors in Non-Small Cell Lung Cancer. Int J Mol Sci 2024; 25:4633. [PMID: 38731852 PMCID: PMC11083651 DOI: 10.3390/ijms25094633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Lung cancer, despite recent advancements in survival rates, represents a significant global health burden. Non-small cell lung cancer (NSCLC), the most prevalent type, is driven largely by activating mutations in Kirsten rat sarcoma viral oncogene homologue (KRAS) and receptor tyrosine kinases (RTKs), and less in v-RAF murine sarcoma viral oncogene homolog B (BRAF) and mitogen-activated protein-kinase kinase (MEK), all key components of the RTK-RAS-mitogen-activated protein kinase (MAPK) pathway. Learning from melanoma, the identification of BRAFV600E substitution in NSCLC provided the rationale for the investigation of RAF and MEK inhibition as a therapeutic strategy. The regulatory approval of two RAF-MEK inhibitor combinations, dabrafenib-trametinib, in 2017, and encorafenib-binimetinib, in 2023, signifies a breakthrough for the management of BRAFV600E-mutant NSCLC patients. However, the almost universal emergence of acquired resistance limits their clinical benefit. New RAF and MEK inhibitors, with distinct biochemical characteristics, are in preclinical and clinical development. In this review, we aim to provide valuable insights into the current state of RAF and MEK inhibition in the management of NSCLC, fostering a deeper understanding of the potential impact on patient outcomes.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Kostas A. Papavassiliou
- First University Department of Respiratory Medicine, ‘Sotiria’ Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Poulikos I. Poulikakos
- Department of Oncological Sciences, Precision Immunology Institute, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
15
|
Hussain SA, Faisal H, Dy GK. Prolonged Disease Control Despite ALK Inhibitor Discontinuation in Advanced ALK-Positive NSCLC. Eur J Case Rep Intern Med 2024; 11:004527. [PMID: 38846658 PMCID: PMC11152215 DOI: 10.12890/2024_004527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/09/2024] Open
Abstract
Introduction EML4-ALK is an oncogenic driver, seen in around five per cent of advanced non-small-cell lung cancer (NSCLC) patients, which can be targeted with anaplastic lymphoma kinase tyrosine kinase inhibitors with great response rates. Disease flare refers to sudden rapid disease worsening on tyrosine kinase inhibitors (TKI) discontinuation, which is associated with shorter survival and worse outcomes. Here, we review cases previously published in the literature where patients developed disease flares, and contrast this with our patients who had prolonged survival despite TKI discontinuation. Case description We report three different patients with advanced ALK-positive NSCLC seen at our institute, who had EML4-ALK translocation variant 1 oncogenic driver on next-generation sequencing. They received treatment with several different ALK inhibitors before opting to discontinue TKI. They were able to come off TKI safely without developing disease flare and had prolonged survival. Discussion Shorter time to progression on TKI, presence of symptoms with disease progression or central nervous system/pleural metastasis have been previously linked with development of flare, although this was not seen in our case series. Tumour response at the time of treatment discontinuation, line of therapy, overall disease burden, fusion variant and co-alteration status can affect the prognosis of these patients after ALK TKI cessation. In particular, variant 1 and wild-type TP53 status may be a suitable patient population for dose optimisation strategies. Intermittent TKI dosing strategies may help to avoid acquiring resistance mutations and prevent long-term treatment toxicities. Conclusion It is important for clinicians to identify patients at risk for developing disease flare on TKI discontinuation to improve outcomes. Intermittent TKI dosing strategies require further investigation. LEARNING POINTS Patients who develop disease flare after cessation have poor survival and worse outcomes.Certain phenotypic and molecular characteristics of the tumour may help clinicians identify which patients are likely and which are unlikely to develop disease flare on TKI discontinuation.Advanced ALK-positive NSCLC with variant 1 and wild-type TP53 may be a suitable patient population for intermittent TKI dosing investigations.
Collapse
Affiliation(s)
- Syed Ather Hussain
- Department of Thoracic Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| | - Hafsa Faisal
- Department of Pulmonary Critical Care Medicine, University at Buffalo, Buffalo, USA
| | - Grace K. Dy
- Department of Thoracic Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, USA
| |
Collapse
|
16
|
Nelson BE, Roszik J, Ahmed J, Barretto CMN, Nardo M, Campbell E, Johnson AM, Piha-Paul SA, Oliva ICG, Weathers SP, Cabanillas M, Javle M, Meric-Bernstam F, Subbiah V. RAF inhibitor re-challenge therapy in BRAF-aberrant pan-cancers: the RE-RAFFLE study. Mol Cancer 2024; 23:64. [PMID: 38532456 PMCID: PMC10964523 DOI: 10.1186/s12943-024-01982-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/11/2024] [Indexed: 03/28/2024] Open
Abstract
Previous studies have shown the clinical benefit of rechallenging the RAF pathway in melanoma patients previously treated with BRAF inhibitors. 44 patients with multiple tumors harboring RAF alterations were rechallenged with a second RAF inhibitor, either as monotherapy or in combination with other therapies, after prior therapy with a first RAF inhibitor. This retrospective observational study results showed that rechallenging with RAFi(s) led to an overall response rate of 18.1% [PR in thyroid (1 anaplastic; 3 papillary), 1 ovarian, 2 melanoma, 1 cholangiocarcinoma, and 1 anaplastic astrocytoma]. The clinical benefit rate was 54.5%; more than 30% of patients had durable responses with PR and SD lasting > 6 months. The median progression-free survival on therapy with second RAF inhibitor in the rechallenge setting either as monotherapy or combination was shorter at 2.7 months (0.9-30.1 m) compared to 8.6 months (6.5-11.5 m) with RAF-1i. However, the median PFS with RAF-2i responders (PFS-2) improved at 12.8 months compared to 11.4 months with RAF-1i responders. The median OS from retreatment with RAF-2i was 15.5 months (11.1-30.8 m). Further prospective studies are needed to validate these results and expand targeted therapy options for RAF-aberrant cancers.
Collapse
Affiliation(s)
- Blessie Elizabeth Nelson
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Jason Roszik
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jibran Ahmed
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Carmelia Maria Noia Barretto
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mirella Nardo
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Erick Campbell
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Amber M Johnson
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarina A Piha-Paul
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Isabella C Glitza Oliva
- Department of Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiao-Pei Weathers
- Department of Neuro-Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Cabanillas
- Department of Endocrinology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Milind Javle
- Department of Gastrointestinal Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vivek Subbiah
- Early-Phase Drug Development, Sarah Cannon Research Institute, Nashville, TN, USA.
| |
Collapse
|
17
|
Timofeev O, Giron P, Lawo S, Pichler M, Noeparast M. ERK pathway agonism for cancer therapy: evidence, insights, and a target discovery framework. NPJ Precis Oncol 2024; 8:70. [PMID: 38485987 PMCID: PMC10940698 DOI: 10.1038/s41698-024-00554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/16/2024] [Indexed: 03/18/2024] Open
Abstract
At least 40% of human cancers are associated with aberrant ERK pathway activity (ERKp). Inhibitors targeting various effectors within the ERKp have been developed and explored for over two decades. Conversely, a substantial body of evidence suggests that both normal human cells and, notably to a greater extent, cancer cells exhibit susceptibility to hyperactivation of ERKp. However, this vulnerability of cancer cells remains relatively unexplored. In this review, we reexamine the evidence on the selective lethality of highly elevated ERKp activity in human cancer cells of varying backgrounds. We synthesize the insights proposed for harnessing this vulnerability of ERK-associated cancers for therapeutical approaches and contextualize these insights within established pharmacological cancer-targeting models. Moreover, we compile the intriguing preclinical findings of ERK pathway agonism in diverse cancer models. Lastly, we present a conceptual framework for target discovery regarding ERKp agonism, emphasizing the utilization of mutual exclusivity among oncogenes to develop novel targeted therapies for precision oncology.
Collapse
Affiliation(s)
- Oleg Timofeev
- Institute of Molecular Oncology, Member of the German Center for Lung Research (DZL), Philipps University, 35043, Marburg, Germany
| | - Philippe Giron
- Vrije Universiteit Brussel (VUB), Universitair Ziekenhuis Brussel (UZ Brussel), Clinical Sciences, Research group Genetics, Reproduction and Development, Centre for Medical Genetics, Laarbeeklaan 101, 1090, Brussels, Belgium
| | - Steffen Lawo
- CRISPR Screening Core Facility, Max Planck Institute for Biology of Ageing, 50931, Cologne, Germany
| | - Martin Pichler
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany
| | - Maxim Noeparast
- Translational Oncology, II. Med Clinics Hematology and Oncology, 86156, Augsburg, Germany.
| |
Collapse
|
18
|
Hanrahan AJ, Chen Z, Rosen N, Solit DB. BRAF - a tumour-agnostic drug target with lineage-specific dependencies. Nat Rev Clin Oncol 2024; 21:224-247. [PMID: 38278874 PMCID: PMC11857949 DOI: 10.1038/s41571-023-00852-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
In June 2022, the FDA granted Accelerated Approval to the BRAF inhibitor dabrafenib in combination with the MEK inhibitor trametinib for the treatment of adult and paediatric patients (≥6 years of age) with unresectable or metastatic BRAFV600E-mutant solid tumours, except for BRAFV600E-mutant colorectal cancers. The histology-agnostic approval of dabrafenib plus trametinib marks the culmination of two decades of research into the landscape of BRAF mutations in human cancers, the biochemical mechanisms underlying BRAF-mediated tumorigenesis, and the clinical development of selective RAF and MEK inhibitors. Although the majority of patients with BRAFV600E-mutant tumours derive clinical benefit from BRAF inhibitor-based combinations, resistance to treatment develops in most. In this Review, we describe the biochemical basis for oncogenic BRAF-induced activation of MAPK signalling and pan-cancer and lineage-specific mechanisms of intrinsic, adaptive and acquired resistance to BRAF inhibitors. We also discuss novel RAF inhibitors and drug combinations designed to delay the emergence of treatment resistance and/or expand the population of patients with BRAF-mutant cancers who benefit from molecularly targeted therapies.
Collapse
Affiliation(s)
- Aphrothiti J Hanrahan
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ziyu Chen
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Physiology, Biophysics & Systems Biology, Weill Cornell Graduate School of Medical Sciences, Cornell University, New York, NY, USA
| | - Neal Rosen
- Molecular Pharmacology Program, Sloan Kettering Institute for Cancer Research, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - David B Solit
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Weill Cornell Medical College, Cornell University, New York, NY, USA.
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
19
|
Fangusaro J, Jones DT, Packer RJ, Gutmann DH, Milde T, Witt O, Mueller S, Fisher MJ, Hansford JR, Tabori U, Hargrave D, Bandopadhayay P. Pediatric low-grade glioma: State-of-the-art and ongoing challenges. Neuro Oncol 2024; 26:25-37. [PMID: 37944912 PMCID: PMC10768984 DOI: 10.1093/neuonc/noad195] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023] Open
Abstract
The most common childhood central nervous system (CNS) tumor is pediatric low-grade glioma (pLGG), representing 30%-40% of all CNS tumors in children. Although there is high associated morbidity, tumor-related mortality is relatively rare. pLGG is now conceptualized as a chronic disease, underscoring the importance of functional outcomes and quality-of-life measures. A wealth of data has emerged about these tumors, including a better understanding of their natural history and their molecular drivers, paving the way for the use of targeted inhibitors. While these treatments have heralded tremendous promise, challenges remain about how to best optimize their use, and the long-term toxicities associated with these inhibitors remain unknown. The International Pediatric Low-Grade Glioma Coalition (iPLGGc) is a global group of physicians and scientists with expertise in pLGG focused on addressing key pLGG issues. Here, the iPLGGc provides an overview of the current state-of-the-art in pLGG, including epidemiology, histology, molecular landscape, treatment paradigms, survival outcomes, functional outcomes, imaging response, and ongoing challenges. This paper also serves as an introduction to 3 other pLGG manuscripts on (1) pLGG preclinical models, (2) consensus framework for conducting early-phase clinical trials in pLGG, and (3) pLGG resistance, rebound, and recurrence.
Collapse
Affiliation(s)
- Jason Fangusaro
- Department of Hematology and Oncology, Children’s Healthcare of Atlanta and Emory University, Atlanta, Georgia, USA
| | - David T Jones
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
| | - Roger J Packer
- Brain Tumor Institute, Daniel and Jennifer Gilbert Neurofibromatosis Institute, Neuroscience and Behavioral Medicine, Children’s National Medical Center, Washington, District of Columbia, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Till Milde
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Olaf Witt
- Translational Program, Hopp Children’s Cancer Center Heidelberg (KiTZ), Heidelberg, Germany
- German Cancer Research Center (DKFZ), University Hospital Heidelberg, Heidelberg, Germany
- Clinical Cooperation Unit Pediatric Oncology, German Cancer Research Center (DKFZ) and German Consortium for Translational Cancer Research (DKTK), Heidelberg, Germany
- KiTZ Clinical Trial Unit (ZIPO), Department of Pediatric Hematology, Oncology, Immunology and Pulmonology, Heidelberg University Hospital; National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Sabine Mueller
- Department of Neurological Surgery, University of California, San Francisco, California, USA
- Department of Pediatrics, University of California, San Francisco, California, USA
- Department of Neurology, University of California, San Francisco, California, USA
- Department of Oncology, University Children’s Hospital Zürich, Zürich, Switzerland
| | - Michael J Fisher
- Division of Oncology, The Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jordan R Hansford
- Michael Rice Centre for Hematology and Oncology, Women’s and Children’s Hospital, Adelaide, South Australia, Australia
- South Australia Health and Medical Research Institute, Adelaide, South Australia, Australia
- South Australia ImmunoGENomics Cancer Institute, University of Adelaide, Adelaide, South Australia, Australia
| | - Uri Tabori
- The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Darren Hargrave
- University College London Great Ormond Street Institute of Child Health, London, UK
| | - Pratiti Bandopadhayay
- Department of Pediatric Oncology, Dana-Farber/Boston Children’s Cancer and Blood Disorders Center, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Kozyra P, Pitucha M. Revisiting the Role of B-RAF Kinase as a Therapeutic Target in Melanoma. Curr Med Chem 2024; 31:2003-2020. [PMID: 37855341 DOI: 10.2174/0109298673258495231011065225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 07/19/2023] [Accepted: 08/17/2023] [Indexed: 10/20/2023]
Abstract
Malignant melanoma is the rarest but most aggressive and deadly skin cancer. Melanoma is the result of a malignant transformation of melanocytes, which leads to their uncontrolled proliferation. Mutations in the mitogen-activated protein kinase (MAPK) pathway, which are crucial for the control of cellular processes, such as apoptosis, division, growth, differentiation, and migration, are one of its most common causes. BRAF kinase, as one of the known targets of this pathway, has been known for many years as a prominent molecular target in melanoma therapy, and the following mini-review outlines the state-of-the-art knowledge regarding its structure, mutations and mechanisms.
Collapse
Affiliation(s)
- Paweł Kozyra
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL, 20093, Poland
| | - Monika Pitucha
- Independent Radiopharmacy Unit, Faculty of Pharmacy, Medical University of Lublin, Lublin, PL-20093, Poland
| |
Collapse
|
21
|
Derbal Y. Adaptive Control of Tumor Growth. Cancer Control 2024; 31:10732748241230869. [PMID: 38294947 PMCID: PMC10832444 DOI: 10.1177/10732748241230869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/04/2023] [Accepted: 01/15/2024] [Indexed: 02/02/2024] Open
Abstract
Cancer treatment optimizations select the most optimum combinations of drugs, sequencing schedules, and appropriate doses that would limit toxicity and yield an improved patient quality of life. However, these optimizations often lack an adequate consideration of cancer's near-infinite potential for evolutionary adaptation to therapeutic interventions. Adapting cancer therapy based on monitored tumor burden and clonal composition is an intuitively sound approach to the treatment of cancer as an inherently complex and adaptive system. The adaptation would be driven by clinical outcome setpoints embodying the aims to thwart therapeutic resistance and maintain a long-term management of the disease or even a cure. However, given the nonlinear, stochastic dynamics of tumor response to therapeutic interventions, adaptive therapeutic strategies may at least need a one-step-ahead prediction of tumor burden to maintain their control over tumor growth dynamics. The article explores the feasibility of adaptive cancer treatment driven by tumor state feedback assuming cell adaptive fitness to be the underlying source of phenotypic plasticity and pathway entropy as a biomarker of tumor growth trajectory. The exploration is undertaken using deterministic and stochastic models of tumor growth dynamics.
Collapse
Affiliation(s)
- Youcef Derbal
- Ted Rogers School of Information Technology Management, Toronto Metropolitan University, Toronto, ON, Canada
| |
Collapse
|
22
|
Maltas J, Killarney ST, Singleton KR, Strobl MAR, Washart R, Wood KC, Wood KB. Drug dependence in cancer is exploitable by optimally constructed treatment holidays. Nat Ecol Evol 2024; 8:147-162. [PMID: 38012363 PMCID: PMC10918730 DOI: 10.1038/s41559-023-02255-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 10/19/2023] [Indexed: 11/29/2023]
Abstract
Cancers with acquired resistance to targeted therapy can become simultaneously dependent on the presence of the targeted therapy drug for survival, suggesting that intermittent therapy may slow resistance. However, relatively little is known about which tumours are likely to become dependent and how to schedule intermittent therapy optimally. Here we characterized drug dependence across a panel of over 75 MAPK-inhibitor-resistant BRAFV600E mutant melanoma models at the population and single-clone levels. Melanocytic differentiated models exhibited a much greater tendency to give rise to drug-dependent progeny than their dedifferentiated counterparts. Mechanistically, acquired loss of microphthalmia-associated transcription factor in differentiated melanoma models drives ERK-JunB-p21 signalling to enforce drug dependence. We identified the optimal scheduling of 'drug holidays' using simple mathematical models that we validated across short and long timescales. Without detailed knowledge of tumour characteristics, we found that a simple adaptive therapy protocol can produce near-optimal outcomes using only measurements of total population size. Finally, a spatial agent-based model showed that optimal schedules derived from exponentially growing cells in culture remain nearly optimal in the context of tumour cell turnover and limited environmental carrying capacity. These findings may guide the implementation of improved evolution-inspired treatment strategies for drug-dependent cancers.
Collapse
Affiliation(s)
- Jeff Maltas
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA
| | - Shane T Killarney
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | | | - Maximilian A R Strobl
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH, USA
| | - Rachel Washart
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA.
| | - Kevin B Wood
- Department of Biophysics, University of Michigan, Ann Arbor, MI, USA.
- Department of Physics, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
23
|
Dayimu A, Gupta A, Matin RN, Nobes J, Board R, Payne M, Rao A, Fusi A, Danson S, Eccles B, Carser J, Brown CO, Steven N, Bhattacharyya M, Brown E, Gonzalez M, Highley M, Pickering L, Kumar S, Waterston A, Burghel G, Demain L, Baker E, Wulff J, Qian W, Twelves S, Middleton M, Corrie P. A randomised phase 2 study of intermittent versus continuous dosing of dabrafenib plus trametinib in patients with BRAF V600 mutant advanced melanoma (INTERIM). Eur J Cancer 2024; 196:113455. [PMID: 38029480 DOI: 10.1016/j.ejca.2023.113455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/07/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND BRAF+MEK inhibitors extend life expectancy of patients with BRAFV600 mutant advanced melanoma. Acquired resistance limits duration of benefit, but preclinical and case studies suggest intermittent dosing could overcome this limitation. INTERIM was a phase 2 trial evaluating an intermittent dosing regimen. METHODS Patients with BRAFV600 mutant advanced melanoma due to start dabrafenib+trametinib were randomised to receive either continuous (CONT), or intermittent (INT; dabrafenib d1-21, trametinib d1-14 every 28 days) dosing. A composite primary endpoint included progression-free survival (PFS) and quality of life (QoL). Secondary endpoints included response rate (ORR), overall survival (OS) and adverse events (AEs). Mutant BRAFV600E ctDNA was measured by droplet digital PCR (ddPCR), using mutant allele frequency of > 1 % as the detection threshold. RESULTS 79 patients (39 INT, 40 CONT) were recruited; median age 67 years, 65 % AJCC (7th ed) stage IV M1c, 29 % had brain metastases. With 19 months median follow-up, INT was inferior in all efficacy measures: median PFS 8.5 vs 10.7mo (HR 1.39, 95 %CI 0.79-2.45, p = 0.255); median OS 18.1mo vs not reached (HR 1.69, 95 %CI 0.87-3.28, p = 0.121), ORR 57 % vs 77 %. INT patients experienced fewer treatment-related AEs (76 % vs 88 %), but more grade > 3 AEs (53 % vs 42 %). QoL favoured CONT. Detection of BRAFV600E ctDNA prior to treatment correlated with worse OS (HR 2.55, 95 %CI 1.25-5.21, p = 0.01) in both arms. A change to undetected during treatment did not significantly predict better OS. CONCLUSION INTERIM findings are consistent with other recent clinical trials reporting that intermittent dosing does not improve efficacy of BRAF+MEK inhibitors.
Collapse
Affiliation(s)
- Alimu Dayimu
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK
| | | | - Rubeta N Matin
- Department of Dermatology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Jenny Nobes
- Department of Oncology, Norfolk and Norwich University Hospital NHS Foundation Trust, Norfolk, UK
| | - Ruth Board
- Department of Oncology, Lancashire Teaching Hospitals NHS Trust, Preston, UK
| | - Miranda Payne
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK
| | - Ankit Rao
- Department of Oncology, Nottingham University Hospitals NHS Trust, Nottingham, UK
| | - Alberto Fusi
- Department of Medical Oncology, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Sarah Danson
- Division of Clinical Medicine, The University of Sheffield, Sheffield, UK; Sheffield Teaching Hospital NHS Foundation Trust, Sheffield, UK
| | - Bryony Eccles
- Department of Medical Oncology, University Hospitals Dorset NHS Foundation Trust, Poole, UK
| | - Judith Carser
- Department of Oncology, Belfast Health and Social Care Trust, Belfast, UK
| | | | - Neil Steven
- Department of Oncology, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | | | - Ewan Brown
- Western General Hospital, Lothian NHS Board, Edinburgh, UK
| | - Michael Gonzalez
- Department of Oncology, Imperial College Healthcare NHS Trust, London, UK
| | - Martin Highley
- Oncology Centre, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, UK
| | - Lisa Pickering
- Skin and Renal Units, Royal Marsden NHS Foundation Trust, London, UK
| | - Satish Kumar
- Velindre Cancer Centre, Velindre University NHS Trust, Cardiff, UK
| | | | - George Burghel
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Leigh Demain
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Eleanor Baker
- Manchester Centre for Genomic Medicine, Manchester University NHS Foundation Trust, Manchester, UK
| | - Jerome Wulff
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Wendi Qian
- Clinical Trials Unit, Department of Oncology, University of Cambridge, Cambridge, UK; Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sophie Twelves
- Cambridge Clinical Trials Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Mark Middleton
- Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, UK; Department of Oncology, University of Oxford, Oxford, UK
| | - Pippa Corrie
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| |
Collapse
|
24
|
Zhang L, Ma J, Liu L, Li G, Li H, Hao Y, Zhang X, Ma X, Chen Y, Wu J, Wang X, Yang S, Xu S. Adaptive therapy: a tumor therapy strategy based on Darwinian evolution theory. Crit Rev Oncol Hematol 2023; 192:104192. [PMID: 37898477 DOI: 10.1016/j.critrevonc.2023.104192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 04/07/2023] [Accepted: 10/22/2023] [Indexed: 10/30/2023] Open
Abstract
Cancer progression is a dynamic process of continuous evolution, in which genetic diversity and heterogeneity are generated by clonal and subclonal amplification based on random mutations. Traditional cancer treatment strategies have a great challenge, which often leads to treatment failure due to drug resistance. Integrating evolutionary dynamics into treatment regimens may be an effective way to overcome the problem of drug resistance. In particular, a potential treatment is adaptive therapy, which strategy advocates containment strategies that adjust the treatment cycles according to tumor evolution to control the growth of treatment-resistant cells. In this review, we first summarize the shortcomings of traditional tumor treatment methods in evolution and then introduce the theoretical basis and research status of adaptive therapy. By analyzing the limitations of adaptive therapy and exploring possible solutions, we can broaden people's understanding of adaptive therapy and provide new insights and strategies for tumor treatment.
Collapse
Affiliation(s)
- Lei Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Jianli Ma
- Department of Radiotherapy, Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Lei Liu
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Guozheng Li
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Hui Li
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yi Hao
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Zhang
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xin Ma
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Yihai Chen
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Jiale Wu
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Xinheng Wang
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shuai Yang
- Harbin Medical University Cancer Hospital, Harbin, 150040, China
| | - Shouping Xu
- Harbin Medical University Cancer Hospital, Harbin, 150040, China.
| |
Collapse
|
25
|
Pu Y, Li L, Peng H, Liu L, Heymann D, Robert C, Vallette F, Shen S. Drug-tolerant persister cells in cancer: the cutting edges and future directions. Nat Rev Clin Oncol 2023; 20:799-813. [PMID: 37749382 DOI: 10.1038/s41571-023-00815-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
Drug-tolerant persister (DTP) cell populations were originally discovered in antibiotic-resistant bacterial biofilms. Similar populations with comparable features have since been identified among cancer cells and have been linked with treatment resistance that lacks an underlying genomic alteration. Research over the past decade has improved our understanding of the biological roles of DTP cells in cancer, although clinical knowledge of the role of these cells in treatment resistance remains limited. Nonetheless, targeting this population is anticipated to provide new treatment opportunities. In this Perspective, we aim to provide a clear definition of the DTP phenotype, discuss the underlying characteristics of these cells, their biomarkers and vulnerabilities, and encourage further research on DTP cells that might improve our understanding and enable the development of more effective anticancer therapies.
Collapse
Affiliation(s)
- Yi Pu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Burn Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Lu Li
- Lung Cancer Centre, West China Hospital, Sichuan University, Chengdu, China
| | - Haoning Peng
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Lunxu Liu
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Nantes, France
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France
| | - Caroline Robert
- INSERM U981, Gustave Roussy Cancer Campus, Villejuif, France
- Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - François Vallette
- Institut de Cancérologie de l'Ouest, Saint-Herblain, France.
- Nantes Université, INSERM, U1307, CRCI2NA, Nantes, France.
| | - Shensi Shen
- Department of Thoracic Surgery and Institute of Thoracic Oncology, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
26
|
Seth R, Agarwala SS, Messersmith H, Alluri KC, Ascierto PA, Atkins MB, Bollin K, Chacon M, Davis N, Faries MB, Funchain P, Gold JS, Guild S, Gyorki DE, Kaur V, Khushalani NI, Kirkwood JM, McQuade JL, Meyers MO, Provenzano A, Robert C, Santinami M, Sehdev A, Sondak VK, Spurrier G, Swami U, Truong TG, Tsai KK, van Akkooi A, Weber J. Systemic Therapy for Melanoma: ASCO Guideline Update. J Clin Oncol 2023; 41:4794-4820. [PMID: 37579248 DOI: 10.1200/jco.23.01136] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 06/09/2023] [Indexed: 08/16/2023] Open
Abstract
PURPOSE To provide guidance to clinicians regarding the use of systemic therapy for melanoma. METHODS American Society of Clinical Oncology convened an Expert Panel and conducted an updated systematic review of the literature. RESULTS The updated review identified 21 additional randomized trials. UPDATED RECOMMENDATIONS Neoadjuvant pembrolizumab was newly recommended for patients with resectable stage IIIB to IV cutaneous melanoma. For patients with resected cutaneous melanoma, adjuvant nivolumab or pembrolizumab was newly recommended for stage IIB-C disease and adjuvant nivolumab plus ipilimumab was added as a potential option for stage IV disease. For patients with unresectable or metastatic cutaneous melanoma, nivolumab plus relatlimab was added as a potential option regardless of BRAF mutation status and nivolumab plus ipilimumab followed by nivolumab was preferred over BRAF/MEK inhibitor therapy. Talimogene laherparepvec is no longer recommended as an option for patients with BRAF wild-type disease who have progressed on anti-PD-1 therapy. Ipilimumab- and ipilimumab-containing regimens are no longer recommended for patients with BRAF-mutated disease after progression on other therapies.This full update incorporates the new recommendations for uveal melanoma published in the 2022 Rapid Recommendation Update.Additional information is available at www.asco.org/melanoma-guidelines.
Collapse
Affiliation(s)
- Rahul Seth
- SUNY Upstate Medical University, Syracuse, NY
| | - Sanjiv S Agarwala
- Lewis Katz School of Medicine at Temple University, Philadelphia, PA
| | | | | | - Paolo A Ascierto
- Istituto Nazionale Tumori IRCCS Fondazione Pascale, Napoli, Italy
| | | | | | - Matias Chacon
- Instituto Alexander Fleming, Buenos Aires, Argentina
| | - Nancy Davis
- Vanderbilt University Medical Center, Nashville, TN
| | - Mark B Faries
- The Angeles Clinic and Research Institute and Cedars Sinai Medical Center, Los Angeles, CA
| | | | | | | | | | | | | | - John M Kirkwood
- University of Pittsburgh School of Medicine and UPMC Hillman Cancer Institute, Pittsburgh, PA
| | | | - Michael O Meyers
- University of North Carolina School of Medicine, Chapel Hill, NC
| | | | - Caroline Robert
- Gustave Roussy Cancer Centre and Paris-Saclay University, Villejuif, France
| | - Mario Santinami
- Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy
| | | | - Vernon K Sondak
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | - Umang Swami
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT
| | | | - Katy K Tsai
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, CA
| | - Alexander van Akkooi
- Melanoma Institute Australia, University of Sydney and Royal Prince Alfred Hospital, Sydney, Australia
| | - Jeffrey Weber
- Laura and Isaac Perlmutter Cancer Center at NYU Langone Health, New York, NY
| |
Collapse
|
27
|
Strobl MAR, Gallaher J, Robertson-Tessi M, West J, Anderson ARA. Treatment of evolving cancers will require dynamic decision support. Ann Oncol 2023; 34:867-884. [PMID: 37777307 PMCID: PMC10688269 DOI: 10.1016/j.annonc.2023.08.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 08/01/2023] [Accepted: 08/21/2023] [Indexed: 10/02/2023] Open
Abstract
Cancer research has traditionally focused on developing new agents, but an underexplored question is that of the dose and frequency of existing drugs. Based on the modus operandi established in the early days of chemotherapies, most drugs are administered according to predetermined schedules that seek to deliver the maximum tolerated dose and are only adjusted for toxicity. However, we believe that the complex, evolving nature of cancer requires a more dynamic and personalized approach. Chronicling the milestones of the field, we show that the impact of schedule choice crucially depends on processes driving treatment response and failure. As such, cancer heterogeneity and evolution dictate that a one-size-fits-all solution is unlikely-instead, each patient should be mapped to the strategy that best matches their current disease characteristics and treatment objectives (i.e. their 'tumorscape'). To achieve this level of personalization, we need mathematical modeling. In this perspective, we propose a five-step 'Adaptive Dosing Adjusted for Personalized Tumorscapes (ADAPT)' paradigm to integrate data and understanding across scales and derive dynamic and personalized schedules. We conclude with promising examples of model-guided schedule personalization and a call to action to address key outstanding challenges surrounding data collection, model development, and integration.
Collapse
Affiliation(s)
- M A R Strobl
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa; Translational Hematology and Oncology Research, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, USA
| | - J Gallaher
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - M Robertson-Tessi
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - J West
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa
| | - A R A Anderson
- Integrated Mathematical Oncology Department, H. Lee Moffitt Cancer Center & Research Institute, Tampa.
| |
Collapse
|
28
|
Chahoud J, Anderson AR, Zhang J, Brown J, Gatenby RA. Evolutionary Dynamics and Intermittent Therapy for Metastatic Cancers. J Clin Oncol 2023; 41:4469-4471. [PMID: 37418680 PMCID: PMC10553063 DOI: 10.1200/jco.23.00647] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 06/09/2023] [Indexed: 07/09/2023] Open
Abstract
Mathematical models show that the induction period of intermittent cancer therapy drives cancer toward resistance
Collapse
Affiliation(s)
- Jad Chahoud
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | | | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Joel Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Robert A. Gatenby
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center, Tampa, FL
- Department of Radiology, Department of Integrated Mathematical Oncology H. Lee Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
29
|
Hartman ML, Koziej P, Kluszczyńska K, Czyz M. Pro-Apoptotic Activity of MCL-1 Inhibitor in Trametinib-Resistant Melanoma Cells Depends on Their Phenotypes and Is Modulated by Reversible Alterations Induced by Trametinib Withdrawal. Cancers (Basel) 2023; 15:4799. [PMID: 37835493 PMCID: PMC10571954 DOI: 10.3390/cancers15194799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 09/25/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Although BRAFV600/MEK inhibitors improved the treatment of melanoma patients, resistance is acquired almost inevitably. METHODS Trametinib withdrawal/rechallenge and MCL-1 inhibition in trametinib-resistance models displaying distinct p-ERK1/2 levels were investigated. RESULTS Trametinib withdrawal/rechallenge caused reversible changes in ERK1/2 activity impacting the balance between pro-survival and pro-apoptotic proteins. Reversible alterations were found in MCL-1 levels and MCL-1 inhibitors, BIM and NOXA. Taking advantage of melanoma cell dependency on MCL-1 for survival, we used S63845. While it was designed to inhibit MCL-1 activity, we showed that it also significantly reduced NOXA levels. S63845-induced apoptosis was detected as the enhancement of Annexin V-positivity, caspase-3/7 activation and histone H2AX phosphorylation. Percentages of Annexin V-positive cells were increased most efficiently in trametinib-resistant melanoma cells displaying the p-ERK1/2low/MCL-1low/BIMhigh/NOXAlow phenotype with EC50 values at concentrations as low as 0.1 μM. Higher ERK1/2 activity associated with increased MCL-1 level and reduced BIM level limited pro-apoptotic activity of S63845 further influenced by a NOXA level. CONCLUSIONS Our study supports the notion that the efficiency of an agent designed to target a single protein can largely depend on the phenotype of cancer cells. Thus, it is important to define appropriate phenotype determinants to stratify the patients for the novel therapy.
Collapse
Affiliation(s)
| | | | | | - Małgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 92-215 Lodz, Poland; (M.L.H.); (P.K.); (K.K.)
| |
Collapse
|
30
|
Glasheen MQ, Caksa S, Young AG, Wilski NA, Ott CA, Chervoneva I, Flaherty KT, Herlyn M, Xu X, Aplin AE, Capparelli C. Targeting Upregulated cIAP2 in SOX10-Deficient Drug Tolerant Melanoma. Mol Cancer Ther 2023; 22:1087-1099. [PMID: 37343247 PMCID: PMC10527992 DOI: 10.1158/1535-7163.mct-23-0025] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 04/07/2023] [Accepted: 06/16/2023] [Indexed: 06/23/2023]
Abstract
Drug tolerance and minimal residual disease (MRD) are likely to prelude acquired resistance to targeted therapy. Mechanisms that allow persister cells to survive in the presence of targeted therapy are being characterized but selective vulnerabilities for these subpopulations remain uncertain. We identified cellular inhibitor of apoptosis protein 2 (cIAP2) as being highly expressed in SOX10-deficient drug tolerant persister (DTP) melanoma cells. Here, we show that cIAP2 is sufficient to induce tolerance to MEK inhibitors, likely by decreasing the levels of cell death. Mechanistically, cIAP2 is upregulated at the transcript level in SOX10-deficient cells and the AP-1 complex protein, JUND, is required for its expression. Using a patient-derived xenograft model, we demonstrate that treatment with the cIAP1/2 inhibitor, birinapant, during the MRD phase delays the onset of resistance to BRAF inhibitor and MEK inhibitor combination therapy. Together, our data suggest that cIAP2 upregulation in SOX10-deficient subpopulations of melanoma cells induces drug tolerance to MAPK targeting agents and provides a rationale to test a novel therapeutical approach to target MRD.
Collapse
Affiliation(s)
- McKenna Q Glasheen
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Signe Caksa
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Amelia G Young
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Nicole A Wilski
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Connor A Ott
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Inna Chervoneva
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Keith T Flaherty
- Massachusetts General Hospital Cancer Center, Boston, Massachusetts
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program, Philadelphia, Pennsylvania
- The Wistar Institute, Philadelphia, Pennsylvania
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Pharmacology, Physiology and Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Claudia Capparelli
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
31
|
Kim HD, Yoo C, Ryu MH, Kang YK. A randomised phase 2 study of continuous or intermittent dosing schedule of imatinib re-challenge in patients with tyrosine kinase inhibitor-refractory gastrointestinal stromal tumours. Br J Cancer 2023; 129:275-282. [PMID: 37179439 PMCID: PMC10338488 DOI: 10.1038/s41416-023-02269-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/15/2023] Open
Abstract
BACKGROUND Imatinib re-challenge is one of the available therapeutic options for patients with treatment-refractory gastrointestinal stromal tumours (GIST). Intermittent dosing of imatinib was suggested to delay outgrow of the imatinib-resistant clones in a preclinical study, and it could potentially reduce the adverse events. METHODS A randomised phase 2 study was performed to evaluate the efficacy and safety of a continuous or intermittent imatinib schedule in GIST patients whose disease had progressed to at least imatinib and sunitinib. RESULTS Fifty patients were included in the full analysis set. The disease control rate at 12 weeks was 34.8% and 43.5%, and median progression-free survival was 1.68 and 1.57 months in the continuous and intermittent groups, respectively. The frequency of diarrhoea, anorexia, decreased neutrophil, or dysphagia was lower in the intermittent group. The scores for global health status/quality of life was not significantly deteriorated over the 8 weeks in both groups. CONCLUSIONS The intermittent dosage did not improve the efficacy outcomes as compared to the continuous dosage, but showed slightly better safety profiles. Given the limited efficacy of imatinib re-challenge, intermittent dosage may also be considered in clinical circumstances where standard fourth-line agent is unavailable or all other viable treatments failed.
Collapse
Affiliation(s)
- Hyung-Don Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min-Hee Ryu
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yoon-Koo Kang
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
32
|
Goyal Y, Busch GT, Pillai M, Li J, Boe RH, Grody EI, Chelvanambi M, Dardani IP, Emert B, Bodkin N, Braun J, Fingerman D, Kaur A, Jain N, Ravindran PT, Mellis IA, Kiani K, Alicea GM, Fane ME, Ahmed SS, Li H, Chen Y, Chai C, Kaster J, Witt RG, Lazcano R, Ingram DR, Johnson SB, Wani K, Dunagin MC, Lazar AJ, Weeraratna AT, Wargo JA, Herlyn M, Raj A. Diverse clonal fates emerge upon drug treatment of homogeneous cancer cells. Nature 2023; 620:651-659. [PMID: 37468627 PMCID: PMC10628994 DOI: 10.1038/s41586-023-06342-8] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 06/19/2023] [Indexed: 07/21/2023]
Abstract
Even among genetically identical cancer cells, resistance to therapy frequently emerges from a small subset of those cells1-7. Molecular differences in rare individual cells in the initial population enable certain cells to become resistant to therapy7-9; however, comparatively little is known about the variability in the resistance outcomes. Here we develop and apply FateMap, a framework that combines DNA barcoding with single-cell RNA sequencing, to reveal the fates of hundreds of thousands of clones exposed to anti-cancer therapies. We show that resistant clones emerging from single-cell-derived cancer cells adopt molecularly, morphologically and functionally distinct resistant types. These resistant types are largely predetermined by molecular differences between cells before drug addition and not by extrinsic factors. Changes in the dose and type of drug can switch the resistant type of an initial cell, resulting in the generation and elimination of certain resistant types. Samples from patients show evidence for the existence of these resistant types in a clinical context. We observed diversity in resistant types across several single-cell-derived cancer cell lines and cell types treated with a variety of drugs. The diversity of resistant types as a result of the variability in intrinsic cell states may be a generic feature of responses to external cues.
Collapse
Affiliation(s)
- Yogesh Goyal
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
| | - Gianna T Busch
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Maalavika Pillai
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jingxin Li
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ryan H Boe
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Emanuelle I Grody
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Manoj Chelvanambi
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ian P Dardani
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Benjamin Emert
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicholas Bodkin
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jonas Braun
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | | | - Amanpreet Kaur
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Naveen Jain
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pavithran T Ravindran
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ian A Mellis
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karun Kiani
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gretchen M Alicea
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Mitchell E Fane
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Syeda Subia Ahmed
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Haiyin Li
- The Wistar Institute, Philadelphia, PA, USA
| | | | - Cedric Chai
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
- Center for Synthetic Biology, Northwestern University, Chicago, IL, USA
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Reproductive Science, Northwestern University, Chicago, IL, USA
| | | | - Russell G Witt
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rossana Lazcano
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Davis R Ingram
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah B Johnson
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Khalida Wani
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margaret C Dunagin
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander J Lazar
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ashani T Weeraratna
- Department of Biochemistry and Molecular Biology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jennifer A Wargo
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
33
|
Hockings H, Lakatos E, Huang W, Mossner M, Khan MA, Metcalf S, Nicolini F, Smith K, Baker AM, Graham TA, Lockley M. Adaptive therapy achieves long-term control of chemotherapy resistance in high grade ovarian cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.21.549688. [PMID: 37546942 PMCID: PMC10401956 DOI: 10.1101/2023.07.21.549688] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Drug resistance results in poor outcomes for most patients with metastatic cancer. Adaptive Therapy (AT) proposes to address this by exploiting presumed fitness costs incurred by drug-resistant cells when drug is absent, and prescribing dose reductions to allow fitter, sensitive cells to re-grow and re-sensitise the tumour. However, empirical evidence for treatment-induced fitness change is lacking. We show that fitness costs in chemotherapy-resistant ovarian cancer cause selective decline and apoptosis of resistant populations in low-resource conditions. Moreover, carboplatin AT caused fluctuations in sensitive/resistant tumour population size in vitro and significantly extended survival of tumour-bearing mice. In sequential blood-derived cell-free DNA and tumour samples obtained longitudinally from ovarian cancer patients during treatment, we inferred resistant cancer cell population size through therapy and observed it correlated strongly with disease burden. These data have enabled us to launch a multicentre, phase 2 randomised controlled trial (ACTOv) to evaluate AT in ovarian cancer.
Collapse
Affiliation(s)
- Helen Hockings
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Eszter Lakatos
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Weini Huang
- School of Mathematical Sciences, Queen Mary University of London, London, UK
| | - Maximilian Mossner
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Mohammed Ateeb Khan
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Stephen Metcalf
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Francesco Nicolini
- Centre for Cancer Cell and Molecular Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Kane Smith
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Ann-Marie Baker
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Trevor A. Graham
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
- Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK
| | - Michelle Lockley
- Centre for Cancer Genomics and Computational Biology, Barts Cancer Institute, Queen Mary University of London, London, UK
| |
Collapse
|
34
|
Vidal-Cruchez O, Nicolini VJ, Rete T, Jacquet K, Rezzonico R, Lacoux C, Domdom MA, Roméo B, Roux J, Hubstenberger A, Mari B, Mograbi B, Hofman P, Brest P. KRAS and NRAS Translation Is Increased upon MEK Inhibitors-Induced Processing Bodies Dissolution. Cancers (Basel) 2023; 15:3078. [PMID: 37370689 DOI: 10.3390/cancers15123078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/29/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Overactivation of the mitogen-activated protein kinase (MAPK) pathway is a critical driver of many human cancers. However, therapies directly targeting this pathway lead to cancer drug resistance. Resistance has been linked to compensatory RAS overexpression, but the mechanisms underlying this response remain unclear. Here, we find that MEK inhibitors (MEKi) are associated with an increased translation of the KRAS and NRAS oncogenes through a mechanism involving dissolution of processing body (P-body) biocondensates. This effect is seen across different cell types and is extremely dynamic since removal of MEKi and ERK reactivation result in reappearance of P-bodies and reduced RAS-dependent signaling. Moreover, we find that P-body scaffold protein levels negatively impact RAS expression. Overall, we describe a new feedback loop mechanism involving biocondensates such as P-bodies in the translational regulation of RAS proteins and MAPK signaling.
Collapse
Affiliation(s)
- Olivia Vidal-Cruchez
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Victoria J Nicolini
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Tifenn Rete
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Karine Jacquet
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Roger Rezzonico
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
- Université Côte d'Azur, CNRS, INSERM, CNRS UMR7275, IPMC, 06560 Valbonne, France
| | - Caroline Lacoux
- Université Côte d'Azur, CNRS UMR7275, IPMC, 06560 Valbonne, France
| | - Marie-Angela Domdom
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Barnabé Roméo
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Jérémie Roux
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
- Université Côte d'Azur, CNRS UMR7275, IPMC, 06560 Valbonne, France
| | - Arnaud Hubstenberger
- Université Côte d'Azur, Institut Biologie Valrose (IBV), CNRS, Inserm, 06108 Nice, France
| | - Bernard Mari
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
- Université Côte d'Azur, CNRS UMR7275, IPMC, 06560 Valbonne, France
| | - Baharia Mograbi
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| | - Paul Hofman
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
- Université Côte d'Azur, CHU-Nice, Pasteur Hospital, Laboratory of Clinical and Experimental Pathology, Hospital-Integrated Biobank (BB-0033-00025), 06001 Nice, France
| | - Patrick Brest
- Université Côte d'Azur, Institute of Research on Cancer and Aging of Nice (IRCAN), CNRS, INSERM, Centre Antoine Lacassagne, 28, Avenue de Valombrose, 06107 Nice, France
- FHU-OncoAge, IHU-RESPIRera, 06001 Nice, France
| |
Collapse
|
35
|
Haist M, Stege H, Kuske M, Bauer J, Klumpp A, Grabbe S, Bros M. Combination of immune-checkpoint inhibitors and targeted therapies for melanoma therapy: The more, the better? Cancer Metastasis Rev 2023; 42:481-505. [PMID: 37022618 PMCID: PMC10348973 DOI: 10.1007/s10555-023-10097-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/27/2023] [Indexed: 04/07/2023]
Abstract
The approval of immune-checkpoint inhibitors (CPI) and mitogen activated protein kinase inhibitors (MAPKi) in recent years significantly improved the treatment management and survival of patients with advanced malignant melanoma. CPI aim to counter-act receptor-mediated inhibitory effects of tumor cells and immunomodulatory cell types on effector T cells, whereas MAPKi are intended to inhibit tumor cell survival. In agreement with these complementary modes of action preclinical data indicated that the combined application of CPI and MAPKi or their optimal sequencing might provide additional clinical benefit. In this review the rationale and preclinical evidence that support the combined application of MAPKi and CPI either in concurrent or consecutive regimens are presented. Further, we will discuss the results from clinical trials investigating the sequential or combined application of MAPKi and CPI for advanced melanoma patients and their implications for clinical practice. Finally, we outline mechanisms of MAPKi and CPI cross-resistance which limit the efficacy of currently available treatments, as well as combination regimens.
Collapse
Affiliation(s)
- Maximilian Haist
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Henner Stege
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Michael Kuske
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Julia Bauer
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Annika Klumpp
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131, Mainz, Germany
| |
Collapse
|
36
|
Koziej P, Kluszczynska K, Hartman ML, Czyz M. Trametinib-Resistant Melanoma Cells Displaying MITF high/NGFR low/IL-8 low Phenotype Are Highly Responsive to Alternating Periods of Drug Withdrawal and Drug Rechallenge. Int J Mol Sci 2023; 24:ijms24097891. [PMID: 37175614 PMCID: PMC10178474 DOI: 10.3390/ijms24097891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/19/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Despite significant advances in targeted therapies against the hyperactivated BRAFV600/MEK pathway for patients with unresectable metastatic melanoma, acquired resistance remains an unsolved clinical problem. In this study, we focused on melanoma cells resistant to trametinib, an agent broadly used in combination therapies. Molecular and cellular changes were assessed during alternating periods of trametinib withdrawal and rechallenge in trametinib-resistant cell lines displaying either a differentiation phenotype (MITFhigh/NGFRlow) or neural crest stem-like dedifferentiation phenotype (NGFRhigh/MITFlow). Neither drug withdrawal nor drug rechallenge induced cell death, and instead of loss of fitness, trametinib-resistant melanoma cells adapted to altered conditions by phenotype switching. In resistant cells displaying a differentiation phenotype, trametinib withdrawal markedly decreased MITF level and activity, which was associated with reduced cell proliferation capacity, and induced stemness assessed as NGFR-positive cells and senescence features, including IL-8 expression and secretion. All these changes could be reversed by trametinib re-exposure, which emphasizes melanoma cell plasticity. Trametinib-resistant cells displaying a dedifferentiation phenotype were less responsive presumably due to the already low level of MITF, a master regulator of the melanoma phenotype. Considering new directions of the development of anti-melanoma treatment, our study suggests that the phenotype of melanomas resistant to targeted therapy might be a crucial determinant of the selection of second-line therapy for melanoma patients.
Collapse
Affiliation(s)
- Paulina Koziej
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Katarzyna Kluszczynska
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Mariusz L Hartman
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| | - Malgorzata Czyz
- Department of Molecular Biology of Cancer, Medical University of Lodz, 6/8 Mazowiecka Street, 92-215 Lodz, Poland
| |
Collapse
|
37
|
West J, Adler F, Gallaher J, Strobl M, Brady-Nicholls R, Brown J, Roberson-Tessi M, Kim E, Noble R, Viossat Y, Basanta D, Anderson ARA. A survey of open questions in adaptive therapy: Bridging mathematics and clinical translation. eLife 2023; 12:e84263. [PMID: 36952376 PMCID: PMC10036119 DOI: 10.7554/elife.84263] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 02/27/2023] [Indexed: 03/24/2023] Open
Abstract
Adaptive therapy is a dynamic cancer treatment protocol that updates (or 'adapts') treatment decisions in anticipation of evolving tumor dynamics. This broad term encompasses many possible dynamic treatment protocols of patient-specific dose modulation or dose timing. Adaptive therapy maintains high levels of tumor burden to benefit from the competitive suppression of treatment-sensitive subpopulations on treatment-resistant subpopulations. This evolution-based approach to cancer treatment has been integrated into several ongoing or planned clinical trials, including treatment of metastatic castrate resistant prostate cancer, ovarian cancer, and BRAF-mutant melanoma. In the previous few decades, experimental and clinical investigation of adaptive therapy has progressed synergistically with mathematical and computational modeling. In this work, we discuss 11 open questions in cancer adaptive therapy mathematical modeling. The questions are split into three sections: (1) integrating the appropriate components into mathematical models (2) design and validation of dosing protocols, and (3) challenges and opportunities in clinical translation.
Collapse
Affiliation(s)
- Jeffrey West
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Fred Adler
- Department of Mathematics, University of UtahSalt Lake CityUnited States
- School of Biological Sciences, University of UtahSalt Lake CityUnited States
| | - Jill Gallaher
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Maximilian Strobl
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Renee Brady-Nicholls
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Joel Brown
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Mark Roberson-Tessi
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Eunjung Kim
- Natural Product Informatics Research Center, Korea Institute of Science and TechnologyGangneungRepublic of Korea
| | - Robert Noble
- Department of Mathematics, University of LondonLondonUnited Kingdom
| | - Yannick Viossat
- Ceremade, Université Paris-Dauphine, Université Paris Sciences et LettresParisFrance
| | - David Basanta
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| | - Alexander RA Anderson
- Department of Integrated Mathematical Oncology, H. Lee Moffitt Cancer Center & Research InstituteTampaUnited States
| |
Collapse
|
38
|
Jin H, Wang L, Bernards R. Rational combinations of targeted cancer therapies: background, advances and challenges. Nat Rev Drug Discov 2023; 22:213-234. [PMID: 36509911 DOI: 10.1038/s41573-022-00615-z] [Citation(s) in RCA: 165] [Impact Index Per Article: 82.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 12/15/2022]
Abstract
Over the past two decades, elucidation of the genetic defects that underlie cancer has resulted in a plethora of novel targeted cancer drugs. Although these agents can initially be highly effective, resistance to single-agent therapies remains a major challenge. Combining drugs can help avoid resistance, but the number of possible drug combinations vastly exceeds what can be tested clinically, both financially and in terms of patient availability. Rational drug combinations based on a deep understanding of the underlying molecular mechanisms associated with therapy resistance are potentially powerful in the treatment of cancer. Here, we discuss the mechanisms of resistance to targeted therapies and how effective drug combinations can be identified to combat resistance. The challenges in clinically developing these combinations and future perspectives are considered.
Collapse
Affiliation(s)
- Haojie Jin
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Liqin Wang
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| | - René Bernards
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Division of Molecular Carcinogenesis, Oncode Institute, Netherlands Cancer Institute, Amsterdam, the Netherlands.
| |
Collapse
|
39
|
Peng K, Zhang Y, Liu D, Chen J. MMP2 is a immunotherapy related biomarker and correlated with cancer-associated fibroblasts infiltrate in melanoma. Cancer Cell Int 2023; 23:26. [PMID: 36788565 PMCID: PMC9930295 DOI: 10.1186/s12935-023-02862-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Accepted: 01/28/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Mounting evidence supports that matrix metalloproteinase (MMPs) are highly associated with tumor progression and that targeting MMPs may overcome the barrier of immune suppression. Among these, whether MMP2 functions as an immunosuppressive role in melanoma, remains unclear. METHODS The Cancer Genome Atlas (TCGA) and Gene Expression Profiling Interactive Analysis 2 (GEPIA2) databases were used to assess the prognosis of MMP2 in melanoma, after which Tumor immune estimation resource (TIMER) was used to explore the relationship between MMP2 expression and cancer associated fibroblasts (CAFs) infiltration. Finally, we evaluated the efficacy of MMP2 inhibitor on CAFs infiltration and immunotherapy using a mouse melanoma model. RESULTS In general, the expression of MMP2, MMP13, MMP16, MMP17 and MMP25 were significantly associated with skin cutaneous melanoma (SKCM) patients prognosis, among which MMP2 low expression benefited patients the most. Especially, the overall survival (OS) of BRAF mutation patients with high MMP2 expression was significantly lower than the MMP2 low expression group, but there was no significant difference in BRAF wild-type patients. KEGG and GO enrichment analysis indicated that MMP2 related genes were mostly associated with extracellular structure organization, collagen-containing extracellular matrix and extracellular matrix structural constituent. Furthermore, in almost all cancers, MMP2 expression was positively correlated with CAFs infiltration. MMP2 inhibitor works synergistically with PD-1 antibody and induces tumor regression in a mouse melanoma model, which is dependent on decreased CAFs infiltration. CONCLUSIONS This suggests that MMP2 plays a vital role in the regulation of CAFs infiltration, potentially participating in immunotherapy response, and thus representing a valuable target of immunotherapy in melanoma.
Collapse
Affiliation(s)
- Kunwei Peng
- grid.412534.5Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260 Guangdong People’s Republic of China
| | - Yanyan Zhang
- grid.79703.3a0000 0004 1764 3838Department of Infectious Diseases, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong People’s Republic of China
| | - Deyi Liu
- grid.412534.5Department of General Practice, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong People’s Republic of China
| | - Jingqi Chen
- Guangdong Provincial Education Department Key Laboratory of Nano-Immunoregulation Tumour Microenvironment, Department of Medical Oncology, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260, Guangdong, People's Republic of China. .,Translational Medicine Center, The Second Affiliated Hospital of Guangzhou Medical University, No. 250 Changgang East Road, Guangzhou, 510260, Guangdong, People's Republic of China.
| |
Collapse
|
40
|
Chandra S, Choi JS, Sosman JA. Melanoma: Does Sequencing Really Matter? J Clin Oncol 2023; 41:167-169. [PMID: 36126246 DOI: 10.1200/jco.22.01354] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Affiliation(s)
- Sunandana Chandra
- Division of Hematology Oncology, Northwestern University, Chicago, IL
| | - Jacob S Choi
- Division of Hematology Oncology, Northwestern University, Chicago, IL
| | - Jeffrey A Sosman
- Division of Hematology Oncology, Northwestern University, Chicago, IL
| |
Collapse
|
41
|
Garutti M, Bergnach M, Polesel J, Palmero L, Pizzichetta MA, Puglisi F. BRAF and MEK Inhibitors and Their Toxicities: A Meta-Analysis. Cancers (Basel) 2022; 15:cancers15010141. [PMID: 36612138 PMCID: PMC9818023 DOI: 10.3390/cancers15010141] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/15/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
PURPOSE This meta-analysis summarizes the incidence of treatment-related adverse events (AE) of BRAFi and MEKi. METHODS A systematic search of Medline/PubMed was conducted to identify suitable articles published in English up to 31 December 2021. The primary outcomes were profiles for all-grade and grade 3 or higher treatment-related AEs, and the analysis of single side effects belonging to both categories. RESULTS The overall incidence of treatment-related all-grade Aes was 99% for Encorafenib (95% CI: 0.97-1.00) and 97% for Trametinib (95% CI: 0.92-0.99; I2 = 66%) and Binimetinib (95% CI: 0.94-0.99; I2 = 0%). In combined therapies, the rate was 98% for both Vemurafenib + Cobimetinib (95% CI: 0.96-0.99; I2 = 77%) and Encorafenib + Binimetinib (95% CI: 0.96-1.00). Grade 3 or higher adverse events were reported in 69% of cases for Binimetinib (95% CI: 0.50-0.84; I2 = 71%), 68% for Encorafenib (95% CI: 0.61-0.74), and 72% for Vemurafenib + Cobimetinib (95% CI: 0.65-0.79; I2 = 84%). The most common grade 1-2 AEs were pyrexia (43%) and fatigue (28%) for Dabrafenib + Trametinib and diarrhea for both Vemurafenib + Cobimetinib (52%) and Encorafenib + Binimetinib (34%). The most common AEs of grade 3 or higher were pyrexia, rash, and hypertension for Dabrafenib + Trametinib (6%), rash and hypertension for Encorafenib + Binimetinib (6%), and increased AST and ALT for Vemurafenib + Cobimetinib (10%). CONCLUSIONS Our study provides comprehensive data on treatment-related adverse events of BRAFi and MEKi combination therapies, showing related toxicity profiles to offer a helpful tool for clinicians in the choice of therapy.
Collapse
Affiliation(s)
- Mattia Garutti
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Correspondence: ; Tel.: +39-04-3465-9092
| | | | - Jerry Polesel
- Unit of Cancer Epidemiology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, 33081 Aviano, Italy
| | - Lorenza Palmero
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| | - Maria Antonietta Pizzichetta
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Dermatology, University of Trieste, 34123 Trieste, Italy
| | - Fabio Puglisi
- CRO Aviano, National Cancer Institute, IRCCS, 33081 Aviano, Italy
- Department of Medicine, University of Udine, 33100 Udine, Italy
| |
Collapse
|
42
|
Faisal Hamdi AI, How SH, Islam MK, Lim JCW, Stanslas J. Adaptive therapy to circumvent drug resistance to tyrosine kinase inhibitors in cancer: is it clinically relevant? Expert Rev Anticancer Ther 2022; 22:1309-1323. [PMID: 36376248 DOI: 10.1080/14737140.2022.2147671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
INTRODUCTION Cancer is highly adaptable and is constantly evolving against current targeted therapies such as tyrosine kinase inhibitors. Despite advances in recent decades, the emergence of drug resistance to tyrosine kinase inhibitors constantly hampers therapeutic efficacy of cancer treatment. Continuous therapy versus intermittent clinical regimen has been a debate in drug administration of cancer patients. An ecologically-inspired shift in cancer treatment known as 'adaptive therapy' intends to improve the drug administration of drugs to cancer patients that can delay emergence of drug resistance. AREAS COVERED We discuss improved understanding of the concept of drug resistance, the basis of continuous therapy, intermittent clinical regimens, and adaptive therapy will be reviewed. In addition, we discuss how adaptive therapy provides guidance for future cancer treatment. EXPERT OPINION The current understanding of drug resistance in cancer leads to poor prognosis and limited treatment options in patients. Fighting drug resistance mutants is constantly followed by new forms of resistance. In most reported cases, continuous therapy leads to drug resistance and an intermittent clinical regimen vaguely delays it. However, adaptive therapy, conceptually, exploits multiple parameters that can suppress the growth of drug resistance and provides safe treatment for cancer patients in the future.
Collapse
Affiliation(s)
- Amir Imran Faisal Hamdi
- Pharmacotherapeutics Unit, Department of Medicine, Universiti Putra MalaysiaMedicine, 43400, Serdang, Malaysia
| | - Soon Hin How
- Kuliyyah of Medicine, International Islamic University Malaysia, Kuantan Campus, Kuliyyah of Medicine, 25200, Kuantan, Malaysia
| | | | - Jonathan Chee Woei Lim
- Pharmacotherapeutics Unit, Department of Medicine, Universiti Putra MalaysiaMedicine, 43400, Serdang, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Universiti Putra MalaysiaMedicine, 43400, Serdang, Malaysia
| |
Collapse
|
43
|
Farnsworth DA, Inoue Y, Johnson FD, de Rappard-Yuswack G, Lu D, Shi R, Ma LIJ, Mattar MS, Somwar R, Ladanyi M, Unni AM, Lockwood WW. MEK inhibitor resistance in lung adenocarcinoma is associated with addiction to sustained ERK suppression. NPJ Precis Oncol 2022; 6:88. [PMID: 36418460 PMCID: PMC9684561 DOI: 10.1038/s41698-022-00328-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 11/01/2022] [Indexed: 11/25/2022] Open
Abstract
MEK inhibitors (MEKi) have limited efficacy in KRAS mutant lung adenocarcinoma (LUAD) patients, and this is attributed to both intrinsic and adaptive mechanisms of drug resistance. While many studies have focused on the former, there remains a dearth of data regarding acquired resistance to MEKi in LUAD. We established trametinib-resistant KRAS mutant LUAD cells through dose escalation and performed targeted MSK-IMPACT sequencing to identify drivers of MEKi resistance. Comparing resistant cells to their sensitive counterparts revealed alteration of genes associated with trametinib response. We describe a state of "drug addiction" in resistant cases where cells are dependent on continuous culture in trametinib for survival. We show that dependence on ERK2 suppression underlies this phenomenon and that trametinib removal hyperactivates ERK, resulting in ER stress and apoptosis. Amplification of KRASG12C occurs in drug-addicted cells and blocking mutant-specific activity with AMG 510 rescues the lethality associated with trametinib withdrawal. Furthermore, we show that increased KRASG12C expression is lethal to other KRAS mutant LUAD cells, consequential to ERK hyperactivation. Our study determines the drug-addicted phenotype in lung cancer is associated with KRAS amplification and demonstrates that toxic acquired genetic changes can develop de novo in the background of MAPK suppression with MEK inhibitors. We suggest that the presence of mutant KRAS amplification in patients may identify those that may benefit from a "drug holiday" to circumvent drug resistance. These findings demonstrate the toxic potential of hyperactive ERK signaling and highlight potential therapeutic opportunities in patients bearing KRAS mutations.
Collapse
Affiliation(s)
- Dylan A. Farnsworth
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | - Yusuke Inoue
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | - Fraser D. Johnson
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | | | - Daniel Lu
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | - Rocky Shi
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | - Lok In Josephine Ma
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada
| | - Marissa S. Mattar
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Romel Somwar
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA ,grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Marc Ladanyi
- grid.51462.340000 0001 2171 9952Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY USA ,grid.51462.340000 0001 2171 9952Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY USA
| | - Arun M. Unni
- grid.5386.8000000041936877XMeyer Cancer Center, Weill Cornell Medicine, New York, NY USA
| | - William W. Lockwood
- Department of Integrative Oncology, BC Cancer Research Institute, Vancouver, Canada ,grid.17091.3e0000 0001 2288 9830Department of Pathology & Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
44
|
Caksa S, Baqai U, Aplin AE. The future of targeted kinase inhibitors in melanoma. Pharmacol Ther 2022; 239:108200. [PMID: 35513054 PMCID: PMC10187889 DOI: 10.1016/j.pharmthera.2022.108200] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/20/2022] [Accepted: 04/28/2022] [Indexed: 12/13/2022]
Abstract
Melanoma is a cancer of the pigment-producing cells of the body and its incidence is rising. Targeted inhibitors that act against kinases in the MAPK pathway are approved for BRAF-mutant metastatic cutaneous melanoma and increase patients' survival. Response to these therapies is limited by drug resistance and is less durable than with immune checkpoint inhibition. Conversely, rare melanoma subtypes have few therapeutic options for advanced disease and MAPK pathway targeting agents show minimal anti-tumor effects. Nevertheless, there is a future for targeted kinase inhibitors in melanoma: in new applications such as adjuvant or neoadjuvant therapy and in novel combinations with immunotherapies or other targeted therapies. Pre-clinical studies continue to identify tumor dependencies and their corresponding actionable drug targets, paving the way for rational targeted kinase inhibitor combinations as a personalized medicine approach for melanoma.
Collapse
Affiliation(s)
- Signe Caksa
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Usman Baqai
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
45
|
Corrie P, Meyer N, Berardi R, Guidoboni M, Schlueter M, Kolovos S, Macabeo B, Trouiller JB, Laramée P. Comparative efficacy and safety of targeted therapies for BRAF-mutant unresectable or metastatic melanoma: Results from a systematic literature review and a network meta-analysis. Cancer Treat Rev 2022; 110:102463. [PMID: 36099854 DOI: 10.1016/j.ctrv.2022.102463] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/30/2022] [Accepted: 08/30/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND The objective of this study was to estimate the relative efficacy and safety of targeted therapies for the treatment of metastatic melanoma using a network meta-analysis (NMA). METHODS A systematic literature review (SLR) identified studies in Medline, Embase and Cochrane published until November 2020. Screening used prespecified eligibility criteria. Following a transitivity assessment across included studies, Bayesian NMA was conducted. RESULTS A total of 43 publications reporting 15 targeted therapy trials and 42 reporting 18 immunotherapy trials were retained from the SLR and considered for the NMA. Due to substantial between-study heterogeneity with immunotherapy trials, the analysis considered a network restricted to targeted therapies. Among combination therapies, encorafenib + binimetinib was superior to dabrafenib + trametinib for overall response rate (OR = 1.86; 95 % credible interval [CrI] 1.10, 3.17), superior to vemurafenib + cobimetinib with fewer serious adverse events (SAEs) (OR = 0.51; 95 % CrI 0.29, 0.91) and fewer discontinuations due to AEs (OR = 0.45; 95 % CrI 0.21, 0.96), and superior to atezolizumab + vemurafenib + cobimetinib with fewer SAEs (OR = 0.41; 95 % CrI 0.21, 0.82). Atezolizumab + vemurafenib + cobimetinib and encorafenib + binimetinib were generally comparable for efficacy endpoints. Among double combination therapies, encorafenib + binimetinib showed high probabilities of being better for all efficacy and safety endpoints. CONCLUSIONS This NMA confirms that combination therapies are more efficacious than monotherapies. Encorafenib + binimetinib has a favourable efficacy profile compared to other double combination therapies and a favourable safety profile compared to both double and triple combination therapies.
Collapse
Affiliation(s)
- Pippa Corrie
- Oncology Department, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Nicolas Meyer
- Institut Universitaire du Cancer et CHU de Toulouse, Toulouse, France; Inserm UMR 1037 - CRCT, Toulouse, France
| | - Rossana Berardi
- Clinica Oncologica, AOU Ospedali Riuniti, Ancona, Università Politecnica delle Marche, Ancona, Italy
| | - Massimo Guidoboni
- Experimental and Clinical Oncology of Immunotherapy and Rare Tumors, IRCCS IRST "Dino Amadori", Meldola, FC, Italy
| | | | | | - Bérengère Macabeo
- Aix-Marseille Université, Marseille, France; Pierre Fabre Laboratories, Paris, France
| | | | - Philippe Laramée
- Aix-Marseille Université, Marseille, France; Pierre Fabre Laboratories, Paris, France.
| |
Collapse
|
46
|
Poulikakos PI, Sullivan RJ, Yaeger R. Molecular Pathways and Mechanisms of BRAF in Cancer Therapy. Clin Cancer Res 2022; 28:4618-4628. [PMID: 35486097 PMCID: PMC9616966 DOI: 10.1158/1078-0432.ccr-21-2138] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 02/08/2022] [Accepted: 03/30/2022] [Indexed: 01/24/2023]
Abstract
With the identification of activating mutations in BRAF across a wide variety of malignancies, substantial effort was placed in designing safe and effective therapeutic strategies to target BRAF. These efforts have led to the development and regulatory approval of three BRAF inhibitors as well as five combinations of a BRAF inhibitor plus an additional agent(s) to manage cancer such as melanoma, non-small cell lung cancer, anaplastic thyroid cancer, and colorectal cancer. To date, each regimen is effective only in patients with tumors harboring BRAFV600 mutations and the duration of benefit is often short-lived. Further limitations preventing optimal management of BRAF-mutant malignancies are that treatments of non-V600 BRAF mutations have been less profound and combination therapy is likely necessary to overcome resistance mechanisms, but multi-drug regimens are often too toxic. With the emergence of a deeper understanding of how BRAF mutations signal through the RAS/MAPK pathway, newer RAF inhibitors are being developed that may be more effective and potentially safer and more rational combination therapies are being tested in the clinic. In this review, we identify the mechanics of RAF signaling through the RAS/MAPK pathway, present existing data on single-agent and combination RAF targeting efforts, describe emerging combinations, summarize the toxicity of the various agents in clinical testing, and speculate as to where the field may be headed.
Collapse
Affiliation(s)
- Poulikos I. Poulikakos
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ryan J. Sullivan
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065
| |
Collapse
|
47
|
Khan S, Patel SP, Shoushtari AN, Ambrosini G, Cremers S, Lee S, Franks L, Singh-Kandah S, Hernandez S, Sender N, Vuolo K, Nesson A, Mundi P, Izar B, Schwartz GK, Carvajal RD. Intermittent MEK inhibition for the treatment of metastatic uveal melanoma. Front Oncol 2022; 12:975643. [PMID: 36249046 PMCID: PMC9557946 DOI: 10.3389/fonc.2022.975643] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/30/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Uveal melanoma (UM) is associated with poor outcomes in the metastatic setting and harbors activating mutations resulting in upregulation of MAPK signaling in almost all cases. The efficacy of selumetinib, an oral allosteric inhibitor of MEK1/2, was limited when administered at a continual dosing schedule of 75 mg BID. Preclinical studies demonstrate that intermittent MEK inhibition reduces compensatory pathway activation and promotes T cell activation. We hypothesized that intermittent dosing of selumetinib would reduce toxicity, allow for the administration of increased doses, and achieve more complete pathway inhibition, thus resulting in improved antitumor activity. Methods We conducted a phase Ib trial of selumetinib using an intermittent dosing schedule in patients with metastatic UM. The primary objective was to estimate the maximum tolerated dose (MTD) and assess safety and tolerability. Secondary objectives included assessment of the overall response rate (RR), progression-free survival (PFS) and overall survival (OS). Tumor biopsies were collected at baseline, on day 3 (on treatment), and between days 11-14 (off treatment) from 9 patients for pharmacodynamic (PD) assessments. Results 29 patients were enrolled and received at least one dose of selumetinib across 4 dose levels (DL; DL1: 100 mg BID; DL2: 125 mg BID; DL3: 150 mg BID; DL4: 175 mg BID). All patients experienced a treatment-related adverse event (TRAE), with 5/29 (17%) developing a grade 3 or higher TRAE. Five dose limiting toxicities (DLT) were observed: 2/20 in DL2, 2/5 in DL3, 1/1 in DL4. The estimated MTD was 150 mg BID (DL3), with an estimated probability of toxicity of 29% (90% probability interval 16%-44%). No responses were observed; 11/29 patients achieved a best response of stable disease (SD). The median PFS and OS were 1.8 months (95% CI 1.7, 4.5) and 7.1 months (95% CI 5.3, 11.5). PD analysis demonstrated at least partial pathway inhibition in all samples at day 3, with reactivation between days 11-14 in 7 of those cases. Conclusions We identified 150 mg BID as the MTD of intermittent selumetinib, representing a 100% increase over the continuous dose MTD (75 mg BID). However, no significant clinical efficacy was observed using this dosing schedule.
Collapse
Affiliation(s)
- Shaheer Khan
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Sapna P Patel
- Department of Melanoma Medical Oncology University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Alexander N Shoushtari
- Department of Medicine Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Grazia Ambrosini
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Serge Cremers
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Shing Lee
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Lauren Franks
- Mailman School of Public Health, Columbia University, New York, NY, United States
| | - Shahnaz Singh-Kandah
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Susana Hernandez
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Naomi Sender
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Kristina Vuolo
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Alexandra Nesson
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Prabhjot Mundi
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Benjamin Izar
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Gary K Schwartz
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| | - Richard D Carvajal
- Department of Medicine Columbia University Irving Medical Center, New York, NY, United States
| |
Collapse
|
48
|
Kulkarni P, Mohanty A, Bhattacharya S, Singhal S, Guo L, Ramisetty S, Mirzapoiazova T, Mambetsariev B, Mittan S, Malhotra J, Gupta N, Kim P, Babikian R, Rajurkar S, Subbiah S, Tan T, Nguyen D, Merla A, Kollimuttathuillam SV, Phillips T, Baik P, Tan B, Vashi P, Shrestha S, Leach B, Garg R, Rich PL, Stewart FM, Pisick E, Salgia R. Addressing Drug Resistance in Cancer: A Team Medicine Approach. J Clin Med 2022; 11:5701. [PMID: 36233569 PMCID: PMC9572909 DOI: 10.3390/jcm11195701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/16/2022] [Accepted: 09/23/2022] [Indexed: 12/04/2022] Open
Abstract
Drug resistance remains one of the major impediments to treating cancer. Although many patients respond well initially, resistance to therapy typically ensues. Several confounding factors appear to contribute to this challenge. Here, we first discuss some of the challenges associated with drug resistance. We then discuss how a 'Team Medicine' approach, involving an interdisciplinary team of basic scientists working together with clinicians, has uncovered new therapeutic strategies. These strategies, referred to as intermittent or 'adaptive' therapy, which are based on eco-evolutionary principles, have met with remarkable success in potentially precluding or delaying the emergence of drug resistance in several cancers. Incorporating such treatment strategies into clinical protocols could potentially enhance the precision of delivering personalized medicine to patients. Furthermore, reaching out to patients in the network of hospitals affiliated with leading academic centers could help them benefit from such innovative treatment options. Finally, lowering the dose of the drug and its frequency (because of intermittent rather than continuous therapy) can also have a significant impact on lowering the toxicity and undesirable side effects of the drugs while lowering the financial burden carried by the patient and insurance providers.
Collapse
Affiliation(s)
- Prakash Kulkarni
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
- Department of Systems Biology, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Atish Mohanty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Supriyo Bhattacharya
- Integrative Genomics Core, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sharad Singhal
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Linlin Guo
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sravani Ramisetty
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Tamara Mirzapoiazova
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Bolot Mambetsariev
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Sandeep Mittan
- Montefiore Medical Center, The University Hospital for Albert Einstein College of Medicine, Bronx, NY 10467, USA
| | - Jyoti Malhotra
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1000 FivePoint, Irvine, CA 92618, USA
| | - Naveen Gupta
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1100 San Bernardino Road, Suite 1100, Upland, CA 91786, USA
| | - Pauline Kim
- Department of Pharmacy, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Razmig Babikian
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Swapnil Rajurkar
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1100 San Bernardino Road, Suite 1100, Upland, CA 91786, USA
| | - Shanmuga Subbiah
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1250 S. Sunset Ave., Suite 303, West Covina, CA 91790, USA
| | - Tingting Tan
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 1601 Avocado Ave., Newport Beach, CA 92660, USA
| | - Danny Nguyen
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 19671 Beach Blvd. #315, Huntington Beach, CA 92648, USA
| | - Amartej Merla
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 38660 Medical Center Dr, Suite A380, Palmdale, CA 93551, USA
| | - Sudarsan V. Kollimuttathuillam
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 16300 Sand Canyon Ave., Suite 207, Irvine, CA 92618, USA
| | - Tanyanika Phillips
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 44151 15th St. West, Lancaster, CA 93534, USA
| | - Peter Baik
- Cancer Treatment Centers of America, CTCA Chicago, 2520 Elisha Avenue, Zion, IL 60099, USA
| | - Bradford Tan
- Cancer Treatment Centers of America, CTCA Chicago, 2520 Elisha Avenue, Zion, IL 60099, USA
| | - Pankaj Vashi
- Cancer Treatment Centers of America, CTCA Chicago, 2520 Elisha Avenue, Zion, IL 60099, USA
| | - Sagun Shrestha
- Cancer Treatment Centers of America, CTCA Phoenix, 14200 West Celebrate Life Way, Goodyear, AZ 85338, USA
| | - Benjamin Leach
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, 15031 Rinaldi St., Suite 150, Mission Hills, CA 91345, USA
| | - Ruchi Garg
- Cancer Treatment Centers of America, CTCA Atlanta, 600 Celebrate Life Parkway, Newnan, GA 30265, USA
| | - Patricia L. Rich
- Cancer Treatment Centers of America, CTCA Atlanta, 600 Celebrate Life Parkway, Newnan, GA 30265, USA
| | - F. Marc Stewart
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| | - Evan Pisick
- Cancer Treatment Centers of America, CTCA Chicago, 2520 Elisha Avenue, Zion, IL 60099, USA
| | - Ravi Salgia
- Department of Medical Oncology and Therapeutics Research, City of Hope National Medical Center, Duarte, CA 91010, USA
| |
Collapse
|
49
|
Homan M, Warrier G, Lao CD, Yentz S, Kraft S, Fecher LA. Treatment related toxicities with combination BRAF and MEK inhibitor therapy in resected stage III melanoma. Front Oncol 2022; 12:855794. [PMID: 36212431 PMCID: PMC9538392 DOI: 10.3389/fonc.2022.855794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 08/16/2022] [Indexed: 11/23/2022] Open
Abstract
Combination dabrafenib (D) and trametinib (T) is an FDA approved adjuvant therapy for patients with resected stage III BRAF-mutant melanoma. We describe treatment-related toxicities with adjuvant D+T in a real-world population through a retrospective case series. The primary endpoint was development of toxicities.
Collapse
Affiliation(s)
- Morgan Homan
- Department of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Govind Warrier
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Christopher D. Lao
- Department of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI, United States
| | - Sarah Yentz
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, United States
| | - Shawna Kraft
- Department of Pharmacy, University of Michigan, Ann Arbor, MI, United States
| | - Leslie A. Fecher
- Department of Internal Medicine and Dermatology, University of Michigan, Ann Arbor, MI, United States
- *Correspondence: Leslie A. Fecher,
| |
Collapse
|
50
|
Xie C, Li Z, Hua Y, Sun S, Zhong L, Chen Q, Feng H, Ji N, Li T, Zhou X, Zeng X, Tang Z, Sun C, Li J, Chen Q. Identification of a BRAF/PA28γ/MEK1 signaling axis and its role in epithelial-mesenchymal transition in oral submucous fibrosis. Cell Death Dis 2022; 13:701. [PMID: 35961969 PMCID: PMC9374740 DOI: 10.1038/s41419-022-05152-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 01/21/2023]
Abstract
Oral submucous fibrosis (OSF) is a chronic and insidious oral potentially malignant disorder associated with a 4-17% risk of oral squamous cell carcinoma (OSCC). Our previous study found that proteasomal activator 28 gamma (PA28γ) is frequently overexpressed in oral squamous cell carcinoma and negatively correlated with poor patient prognosis. However, the role of PA28γ in the occurrence and development of OSF remains unclear. Here, we screened PA28γ-related genes and investigated their function in OSF. We demonstrated that the expression of PA28γ was positively associated with MEK1 and gradually elevated from normal to progressive stages of OSF tissue. Arecoline, a pathogenic component of OSF, could upregulate the protein levels of PA28γ and phosphorylated MEK1 and contribute to epithelial to mesenchymal transition (EMT) in epithelial cells. Notably, PA28γ could interact with MEK1 and upregulate its phosphorylation level. Furthermore, arecoline upregulated BRAF, which can interact with PA28γ and upregulate its protein level. Additionally, BRAF, PA28γ, and MEK1 could form protein complexes and then enhance the MEK1/ERK signaling pathways. The concrete mechanism of the protein stability of PA28γ is that BRAF mediates its degradation by inhibiting its ubiquitination. These findings underscore the instrumental role of PA28γ in the BRAF/MEK1 pathway and enhanced EMT through MEK1/ERK activation in OSF.
Collapse
Affiliation(s)
- Changqing Xie
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China ,grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Zaiye Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Yufei Hua
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Silu Sun
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Liang Zhong
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qian Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Hui Feng
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China ,grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Ning Ji
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Taiwen Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xikun Zhou
- grid.13291.380000 0001 0807 1581State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041 People’s Republic of China
| | - Xin Zeng
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Zhangui Tang
- grid.216417.70000 0001 0379 7164Department of Oral and Maxillofacial Surgery, Xiangya Stomatological Hospital & School of Stomatology, Postdoctoral Research Workstation, Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, Hunan 410078 People’s Republic of China
| | - Chongkui Sun
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Jing Li
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| | - Qianming Chen
- grid.13291.380000 0001 0807 1581State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stom atology, Sichuan University, Chengdu, Sichuan 610041 People’s Republic of China
| |
Collapse
|