1
|
Shebrain A, Idris OA, Jawad A, Zhang T, Xing Y. Advancements and Challenges in Personalized Therapy for BRAF-Mutant Melanoma: A Comprehensive Review. J Clin Med 2024; 13:5409. [PMID: 39336897 PMCID: PMC11432393 DOI: 10.3390/jcm13185409] [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: 08/10/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past several decades, advancements in the treatment of BRAF-mutant melanoma have led to the development of BRAF inhibitors, BRAF/MEK inhibitor combinations, anti-PD-1 therapy, and anti-CTLA4 therapy. Although these therapies have shown substantial efficacy in clinical trials, their sustained effectiveness is often challenged by the tumor microenvironment, which is a highly heterogeneous and complex milieu of immunosuppressive cells that affect tumor progression. The era of personalized medicine holds substantial promise for the tailoring of treatments to individual genetic profiles. However, tumor heterogeneity and immune evasion mechanisms contribute to the resistance to immunotherapy. Despite these challenges, tumor-infiltrating lymphocyte (TIL) therapy, as exemplified by lifileucel, has demonstrated notable efficacy against BRAF V600-mutant melanoma. Additionally, early response biomarkers, such as COX-2 and MMP2, along with FDG-PET imaging, offer the potential to improve personalized immunotherapy by predicting patient responses and determining the optimal treatment duration. Future efforts should focus on reducing the T-cell harvesting periods and costs associated with TIL therapy to enhance efficiency and accessibility.
Collapse
Affiliation(s)
- Abdulaziz Shebrain
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Omer A Idris
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
- Malate Institute for Medical Research, Malate Inc., P.O. Box 23, Grandville, MI 49468, USA
| | - Ali Jawad
- Department of Biological Sciences, Western Michigan University, Kalamazoo, MI 49008, USA
| | - Tiantian Zhang
- Toni Stephenson Lymphoma Center, Department of Hematology and Hematopoietic Stem Cell Transplantation, Beckman Research Institute, City of Hope, Duarte, CA 91010, USA
| | - Yan Xing
- Department of Medical Oncology and Therapeutics Research, City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
2
|
Santoro-Fernandes V, Huff DT, Rivetti L, Deatsch A, Schott B, Perlman SB, Jeraj R. An automated methodology for whole-body, multimodality tracking of individual cancer lesions. Phys Med Biol 2024; 69:085012. [PMID: 38457838 DOI: 10.1088/1361-6560/ad31c6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 03/08/2024] [Indexed: 03/10/2024]
Abstract
Objective. Manual analysis of individual cancer lesions to assess disease response is clinically impractical and requires automated lesion tracking methodologies. However, no methodology has been developed for whole-body individual lesion tracking, across an arbitrary number of scans, and acquired with various imaging modalities.Approach. This study introduces a lesion tracking methodology and benchmarked it using 2368Ga-DOTATATE PET/CT and PET/MR images of eight neuroendocrine tumor patients. The methodology consists of six steps: (1) alignment of multiple scans via image registration, (2) body-part labeling, (3) automatic lesion-wise dilation, (4) clustering of lesions based on local lesion shape metrics, (5) assignment of lesion tracks, and (6) output of a lesion graph. Registration performance was evaluated via landmark distance, lesion matching accuracy was evaluated between each image pair, and lesion tracking accuracy was evaluated via identical track ratio. Sensitivity studies were performed to evaluate the impact of lesion dilation (fixed versus automatic dilation), anatomic location, image modalities (inter- versus intra-modality), registration mode (direct versus indirect registration), and track size (number of time-points and lesions) on lesion matching and tracking performance.Main results. Manual contouring yielded 956 lesions, 1570 lesion-matching decisions, and 493 lesion tracks. The median residual registration error was 2.5 mm. The automatic lesion dilation led to 0.90 overall lesion matching accuracy, and an 88% identical track ratio. The methodology is robust regarding anatomic locations, image modalities, and registration modes. The number of scans had a moderate negative impact on the identical track ratio (94% for 2 scans, 91% for 3 scans, and 81% for 4 scans). The number of lesions substantially impacted the identical track ratio (93% for 2 nodes versus 54% for ≥5 nodes).Significance. The developed methodology resulted in high lesion-matching accuracy and enables automated lesion tracking in PET/CT and PET/MR.
Collapse
Affiliation(s)
- Victor Santoro-Fernandes
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Daniel T Huff
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Luciano Rivetti
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| | - Alison Deatsch
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Brayden Schott
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
| | - Scott B Perlman
- School of Medicine and Public Health, Department of Radiology, Section of Nuclear Medicine, University of Wisconsin, Madison, WI, United States of America
| | - Robert Jeraj
- School of Medicine and Public Health, Department of Medical Physics, University of Wisconsin, Madison, WI, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
3
|
van der Hiel B, Aalbersberg EA, van den Eertwegh AJM, de Wit-van der Veen LJ, Stokkel MPM, Lopez-Yurda M, Boellaard R, Kapiteijn EW, Hospers GAP, Aarts MJB, de Vos FYFL, Boers-Sonderen MJ, van der Veldt AAM, de Groot JWB, Haanen JBAG. The Predictive Value of FDG PET/CT for Determining Progression-Free Survival in Advanced Stage III-IV BRAF -Mutated Melanoma Patients Treated With Targeted Therapy-What Can Be Learned From Progression? Clin Nucl Med 2024; 49:138-145. [PMID: 38113329 DOI: 10.1097/rlu.0000000000004988] [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: 12/21/2023]
Abstract
PURPOSE The aims of this study were to investigate whether (early) PERCIST response monitoring with 18 F-FDG PET/CT is predictive for progression-free survival (PFS) in unresectable stage III or IV melanoma patients treated with BRAF/MEK inhibitor (MEKi) and to define dissemination patterns at progression with a lesion-based evaluation in direct comparison to baseline to improve our understanding of 18 F-FDG PET/CT during BRAF/MEKi. PATIENTS AND METHODS This prospective multicenter single-arm study included 70 patients with unresectable stage III/IV BRAF -mutated melanoma who underwent contrast-enhanced CT and 18 F-FDG PET/CT at baseline and 2 and 7 weeks during treatment with vemurafenib plus cobimetinib and at progression if possible. Tumor response assessment was done with RECIST1.1 and PERCIST. Follow-up PET/CT scans were visually compared with baseline to assess dissemination patterns. RESULTS Using RECIST1.1, PFS was not significantly different between the response groups ( P = 0.26). At 2 weeks, PERCIST median PFS was 15.7 months for patients with complete metabolic response (CMR) versus 8.3 months for non-CMR ( P = 0.035). The hazards ratio (HR) for progression/death in non-CMR versus CMR was 1.99 (95% confidence interval [CI], 1.03-3.84; P = 0.040) and 1.77 (95% CI, 0.91-3.43; P = 0.0935) when adjusting for lactate dehydrogenase (LDH). At 7 weeks, median PFS for PERCIST CMR was 16.7 months versus 8.5 months for non-CMR ( P = 0.0003). The HR for progression/death in the non-CMR group was significantly increased (HR, 2.94; 95% CI, 1.60-5.40; P = 0.0005), even when adjusting for LDH (HR, 2.65; 95% CI, 1.43-4.91; P = 0.0020). At week 7, 18 F-FDG PET/CT was false-positive in all 4 (6%) patients with new FDG-avid lesions but CMR of known metastases. When 18 F-FDG PET/CT was performed at progressive disease, 18/22 (82%) patients had progression of known metastases with or without new 18 F-FDG-avid lesions. CONCLUSIONS This study shows that PERCIST response assessment at week 7 is predictive for PFS, regardless of LDH. At 2 weeks, patients with CMR have longer PFS than patients with non-CMR, but different PET parameters should be investigated to further evaluate the added value of early 18 F-FDG PET/CT. Disease progression on PET/CT is predominated by progression of known metastases, and new 18 F-FDG-avid lesions during BRAF/MEKi are not automatically a sign of recurrent disease.
Collapse
Affiliation(s)
- Bernies van der Hiel
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Else A Aalbersberg
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | | | | | - Marcel P M Stokkel
- From the Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Marta Lopez-Yurda
- Department of Biometrics, Netherlands Cancer Institute-Antoni van Leeuwenhoek
| | - Ronald Boellaard
- Department of Nuclear Medicine, Amsterdam UMC location Vrije Universiteit Amsterdam, Amsterdam
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden
| | - Geke A P Hospers
- Department of Medical Oncology, University Medical Center Groningen, Groningen
| | - Maureen J B Aarts
- Department of Medical Oncology, GROW-School for Oncology and Reproduction, Maastricht University Medical Center, Maastricht
| | - Filip Y F L de Vos
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht
| | | | | | | | | |
Collapse
|
4
|
Lim SY, Rizos H. Single-cell RNA sequencing in melanoma: what have we learned so far? EBioMedicine 2024; 100:104969. [PMID: 38241976 PMCID: PMC10831183 DOI: 10.1016/j.ebiom.2024.104969] [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: 10/29/2023] [Revised: 12/18/2023] [Accepted: 01/03/2024] [Indexed: 01/21/2024] Open
Abstract
Over the past decade, there have been remarkable improvements in the treatment and survival rates of melanoma patients. Treatment resistance remains a persistent challenge, however, and is partly attributable to intratumoural heterogeneity. Melanoma cells can transition through a series of phenotypic and transcriptional cell states that vary in invasiveness and treatment responsiveness. The diverse stromal and immune contexture of the tumour microenvironment also contributes to intratumoural heterogeneity and disparities in treatment response in melanoma patients. Recent advances in single-cell sequencing technologies have enabled a more detailed understanding of melanoma heterogeneity and the underlying transcriptional programs that regulate melanoma cell diversity and behaviour. In this review, we examine the concept of intratumoural heterogeneity and the challenges it poses to achieving long-lasting treatment responses. We focus on the significance of next generation single-cell sequencing in advancing our understanding of melanoma diversity and the unique insights gained from single-cell studies.
Collapse
Affiliation(s)
- Su Yin Lim
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia.
| | - Helen Rizos
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Australia; Melanoma Institute Australia, Sydney, Australia
| |
Collapse
|
5
|
Farah C, Mignion L, Jordan BF. Metabolic Profiling to Assess Response to Targeted and Immune Therapy in Melanoma. Int J Mol Sci 2024; 25:1725. [PMID: 38339003 PMCID: PMC10855758 DOI: 10.3390/ijms25031725] [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: 12/21/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
There is currently no consensus to determine which advanced melanoma patients will benefit from targeted therapy, immunotherapy, or a combination of both, highlighting the critical need to identify early-response biomarkers to advanced melanoma therapy. The goal of this review is to provide scientific rationale to highlight the potential role of metabolic imaging to assess response to targeted and/or immune therapy in melanoma cancer. For that purpose, a brief overview of current melanoma treatments is provided. Then, current knowledge with respect to melanoma metabolism is described with an emphasis on major crosstalks between melanoma cell metabolism and signaling pathways involved in BRAF-targeted therapy as well as in immune checkpoint inhibition therapies. Finally, preclinical and clinical studies using metabolic imaging and/or profiling to assess response to melanoma treatment are summarized with a particular focus on PET (Positron Emission Tomography) imaging and 13C-MRS (Magnetic Resonance Spectroscopy) methods.
Collapse
Affiliation(s)
- Chantale Farah
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| | - Lionel Mignion
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| | - Bénédicte F. Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium;
| |
Collapse
|
6
|
Dromain C, Pavel M, Ronot M, Schaefer N, Mandair D, Gueguen D, Cheng C, Dehaene O, Schutte K, Cahané D, Jégou S, Balazard F. Response heterogeneity as a new biomarker of treatment response in patients with neuroendocrine tumors. Future Oncol 2023; 19:2171-2183. [PMID: 37497626 DOI: 10.2217/fon-2022-1137] [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: 07/28/2023] Open
Abstract
Aim: The RAISE project aimed to find a surrogate end point to predict treatment response early in patients with enteropancreatic neuroendocrine tumors (NET). Response heterogeneity, defined as the coexistence of responding and non-responding lesions, has been proposed as a predictive marker for progression-free survival (PFS) in patients with NETs. Patients & methods: Computerized tomography scans were analyzed from patients with multiple lesions in CLARINET (NCT00353496; n = 148/204). Cox regression analyses evaluated association between response heterogeneity, estimated using the standard deviation of the longest diameter ratio of target lesions, and NET progression. Results: Greater response heterogeneity at a given visit was associated with earlier progression thereafter: week 12 hazard ratio (HR; 95% confidence interval): 1.48 (1.20-1.82); p < 0.001; n = 148; week 36: 1.72 (1.32-2.24); p < 0.001; n = 108. HRs controlled for sum of longest diameter ratio: week 12: 1.28 (1.04-1.59); p = 0.020 and week 36: 1.81 (1.20-2.72); p = 0.005. Conclusion: Response heterogeneity independently predicts PFS in patients with enteropancreatic NETs. Further validation is required.
Collapse
Affiliation(s)
| | - Marianne Pavel
- Department of Medicine 1, Friedrich-Alexander-University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Aprile M, Cataldi S, Perfetto C, Federico A, Ciccodicola A, Costa V. Targeting metabolism by B-raf inhibitors and diclofenac restrains the viability of BRAF-mutated thyroid carcinomas with Hif-1α-mediated glycolytic phenotype. Br J Cancer 2023; 129:249-265. [PMID: 37198319 PMCID: PMC10338540 DOI: 10.1038/s41416-023-02282-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/03/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND B-raf inhibitors (BRAFi) are effective for BRAF-mutated papillary (PTC) and anaplastic (ATC) thyroid carcinomas, although acquired resistance impairs tumour cells' sensitivity and/or limits drug efficacy. Targeting metabolic vulnerabilities is emerging as powerful approach in cancer. METHODS In silico analyses identified metabolic gene signatures and Hif-1α as glycolysis regulator in PTC. BRAF-mutated PTC, ATC and control thyroid cell lines were exposed to HIF1A siRNAs or chemical/drug treatments (CoCl2, EGF, HGF, BRAFi, MEKi and diclofenac). Genes/proteins expression, glucose uptake, lactate quantification and viability assays were used to investigate the metabolic vulnerability of BRAF-mutated cells. RESULTS A specific metabolic gene signature was identified as a hallmark of BRAF-mutated tumours, which display a glycolytic phenotype, characterised by enhanced glucose uptake, lactate efflux and increased expression of Hif-1α-modulated glycolytic genes. Indeed, Hif-1α stabilisation counteracts the inhibitory effects of BRAFi on these genes and on cell viability. Interestingly, targeting metabolic routes with BRAFi and diclofenac combination we could restrain the glycolytic phenotype and synergistically reduce tumour cells' viability. CONCLUSION The identification of a metabolic vulnerability of BRAF-mutated carcinomas and the capacity BRAFi and diclofenac combination to target metabolism open new therapeutic perspectives in maximising drug efficacy and reducing the onset of secondary resistance and drug-related toxicity.
Collapse
Affiliation(s)
- Marianna Aprile
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy.
| | - Simona Cataldi
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy
| | - Caterina Perfetto
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy
| | - Antonio Federico
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy
- Tampere Institute for Advanced Study (IAS), Tampere University, Tampere, Finland
- Finnish Hub for Development and Validation of Integrated Approaches (FHAIVE)-Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Alfredo Ciccodicola
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy
- Department of Science and Technology, University of Naples "Parthenope", Naples, Italy
| | - Valerio Costa
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", CNR, Via P. Castellino 111, 80131, Naples, Italy.
| |
Collapse
|
8
|
Farah C, Neveu MA, Bouzin C, Knezevic Z, Gallez B, Leucci E, Baurain JF, Mignion L, Jordan BF. Hyperpolarized 13C-Pyruvate to Assess Response to Anti-PD1 Immune Checkpoint Inhibition in YUMMER 1.7 Melanoma Xenografts. Int J Mol Sci 2023; 24:ijms24032499. [PMID: 36768822 PMCID: PMC9917169 DOI: 10.3390/ijms24032499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/17/2023] [Accepted: 01/18/2023] [Indexed: 01/31/2023] Open
Abstract
There is currently no consensus to determine which advanced melanoma patients will benefit from immunotherapy, highlighting the critical need to identify early-response biomarkers to immune checkpoint inhibitors. The aim of this work was to evaluate in vivo metabolic spectroscopy using hyperpolarized (HP) 13C-pyruvate and 13C-glucose to assess early response to anti-PD1 therapy in the YUMMER1.7 syngeneic melanoma model. The xenografts showed a significant tumor growth delay when treated with two cycles of an anti-PD1 antibody compared to an isotype control antibody. 13C-MRS was performed in vivo after the injection of hyperpolarized 13C-pyruvate, at baseline and after one cycle of immunotherapy, to evaluate early dynamic changes in 13C-pyruvate-13C-lactate exchange. Furthermore, ex vivo 13C-MRS metabolic tracing experiments were performed after U-13C-glucose injection following one cycle of immunotherapy. A significant decrease in the ratio of HP 13C-lactate to 13C-pyruvate was observed in vivo in comparison with the isotype control group, while there was a lack of change in the levels of 13C lactate and 13C alanine issued from 13C glucose infusion, following ex vivo assessment on resected tumors. Thus, these results suggest that hyperpolarized 13C-pyruvate could be used to assess early response to immune checkpoint inhibitors in melanoma patients.
Collapse
Affiliation(s)
- Chantale Farah
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium
| | - Marie-Aline Neveu
- Laboratory of Tumor Inflammation and Angiogenesis, Department of Oncology, K.U. Leuven, B-3001 Leuven, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform, Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvai, (UCLouvain), B-1200 Brussels, Belgium
| | - Zorica Knezevic
- Laboratory for RNA Cancer Biology, Department of Oncology, K.U. Leuven, B-3001 Leuven, Belgium
| | - Bernard Gallez
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (U.C. Louvain), B-1200 Brussels, Belgium
| | - Eleonora Leucci
- Laboratory for RNA Cancer Biology, Department of Oncology, K.U. Leuven, B-3001 Leuven, Belgium
| | - Jean-François Baurain
- Molecular Imaging and Radiation Oncology (MIRO) Group, Institute de Recherche Expérimentale et Clinique (IREC), B-1200 Brussels, Belgium
| | - Lionel Mignion
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (U.C. Louvain), B-1200 Brussels, Belgium
| | - Bénédicte F. Jordan
- Biomedical Magnetic Resonance Research Group, Louvain Drug Research Institute, Université Catholique de Louvain (UCLouvain), B-1200 Brussels, Belgium
- Nuclear and Electron Spin Technologies (NEST) Platform, Louvain Drug Research Institute (LDRI), Université Catholique de Louvain (U.C. Louvain), B-1200 Brussels, Belgium
- Correspondence:
| |
Collapse
|
9
|
Filippi L, Bianconi F, Schillaci O, Spanu A, Palumbo B. The Role and Potential of 18F-FDG PET/CT in Malignant Melanoma: Prognostication, Monitoring Response to Targeted and Immunotherapy, and Radiomics. Diagnostics (Basel) 2022; 12:929. [PMID: 35453977 PMCID: PMC9028862 DOI: 10.3390/diagnostics12040929] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 12/17/2022] Open
Abstract
Novel therapeutic approaches, consisting of immune check-point inhibitors (ICIs) and molecularly targeted therapy, have thoroughly changed the clinical management of malignant melanoma (MM), the most frequent and deadly skin cancer. Since only 30-40% of MM patients respond to ICIs, imaging biomarkers suitable for the pre-therapeutic stratification and response assessment are warmly welcome. In this scenario, positron emission computed tomography (PET/CT) with 18F-fluorodeoxyglucose (18F-FDG) has been successfully utilized for advanced MM staging and therapy response evaluation. Furthermore, several PET-derived parameters (SUVmax, MTV, TLG) were particularly impactful for the prognostic evaluation of patients submitted to targeted and immunotherapy. In this review, we performed a web-based and desktop research on the clinical applications of 18F-FDG PET/CT in MM, with a particular emphasis on the various metabolic criteria developed for interpreting PET/CT scan in patients undergoing immunotherapy or targeted therapy or a combination of both. Furthermore, the emerging role of radiomics, a quantitative approach to medical imaging applying analysis methodology derived by the field of artificial intelligence, was examined in the peculiar context, putting a particular emphasis on the potential of this discipline to support clinicians in the delicate process of building patient-tailored pathways of care.
Collapse
Affiliation(s)
- Luca Filippi
- Nuclear Medicine Unit, “Santa Maria Goretti” Hospital, Via Antonio Canova, 04100 Latina, Italy
| | - Francesco Bianconi
- Department of Engineering, Università Degli Studi di Perugia, Via Goffredo Duranti 93, 06135 Perugia, Italy;
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University Tor Vergata, Viale Oxford 81, 00133 Rome, Italy;
- IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Angela Spanu
- Unit of Nuclear Medicine, Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 8, 07100 Sassari, Italy;
| | - Barbara Palumbo
- Section of Nuclear Medicine and Health Physics, Department of Medicine and Surgery, Università Degli Studi di Perugia, Piazza Lucio Severi 1, 06132 Perugia, Italy;
| |
Collapse
|
10
|
Prochownik EV, Wang H. Normal and Neoplastic Growth Suppression by the Extended Myc Network. Cells 2022; 11:747. [PMID: 35203395 PMCID: PMC8870482 DOI: 10.3390/cells11040747] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/09/2022] [Accepted: 02/15/2022] [Indexed: 12/20/2022] Open
Abstract
Among the first discovered and most prominent cellular oncogenes is MYC, which encodes a bHLH-ZIP transcription factor (Myc) that both activates and suppresses numerous genes involved in proliferation, energy production, metabolism and translation. Myc belongs to a small group of bHLH-ZIP transcriptional regulators (the Myc Network) that includes its obligate heterodimerization partner Max and six "Mxd proteins" (Mxd1-4, Mnt and Mga), each of which heterodimerizes with Max and largely opposes Myc's functions. More recently, a second group of bHLH-ZIP proteins (the Mlx Network) has emerged that bears many parallels with the Myc Network. It is comprised of the Myc-like factors ChREBP and MondoA, which, in association with the Max-like member Mlx, regulate smaller and more functionally restricted repertoires of target genes, some of which are shared with Myc. Opposing ChREBP and MondoA are heterodimers comprised of Mlx and Mxd1, Mxd4 and Mnt, which also structurally and operationally link the two Networks. We discuss here the functions of these "Extended Myc Network" members, with particular emphasis on their roles in suppressing normal and neoplastic growth. These roles are complex due to the temporal- and tissue-restricted expression of Extended Myc Network proteins in normal cells, their regulation of both common and unique target genes and, in some cases, their functional redundancy.
Collapse
Affiliation(s)
- Edward V. Prochownik
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
- The Department of Microbiology and Molecular Genetics, The University of Pittsburgh School of Medicine, Pittsburgh, PA 15224, USA
- The Hillman Cancer Center of UPMC, Pittsburgh, PA 15224, USA
- The Pittsburgh Liver Research Center, Pittsburgh, PA 15224, USA
| | - Huabo Wang
- Division of Hematology/Oncology, The Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA 15224, USA;
| |
Collapse
|
11
|
Alipour R, Iravani A, Hicks RJ. PET Imaging of Melanoma. Nucl Med Mol Imaging 2022. [DOI: 10.1016/b978-0-12-822960-6.00123-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
12
|
Annovazzi A, Ferraresi V, Rea S, Russillo M, Renna D, Carpano S, Sciuto R. Prognostic value of total metabolic tumour volume and therapy-response assessment by [ 18F]FDG PET/CT in patients with metastatic melanoma treated with BRAF/MEK inhibitors. Eur Radiol 2021; 32:3398-3407. [PMID: 34779873 DOI: 10.1007/s00330-021-08355-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/02/2021] [Accepted: 09/24/2021] [Indexed: 12/31/2022]
Abstract
OBJECTIVES Target therapy with BRAF/MEK inhibitors in metastatic melanoma is characterised by a high response rate; however, acquired resistance to treatment develops in many cases. We aimed to investigate if baseline total metabolic tumour volume (TMTV) and therapy-response assessment by [18F]FDG PET/CT have a prognostic role on progression-free survival (PFS) and overall survival (OS) in patients with metastatic melanoma receiving BRAF ± MEK inhibitors. METHODS Fifty-seven patients who performed an [18F]FDG PET/CT at baseline and on treatment were retrospectively evaluated. A Cox proportional-hazard model was used to examine associations between OS and PFS with baseline clinical/PET parameters as well as for PET response. RESULTS According to EORTC criteria, 34 patients were classified as responders (partial/complete metabolic response [PMR/CMR]) and 23 as non-responders (progressive/stable metabolic disease [PMD/SMD]). Baseline characteristics associated with a shorter PFS were more than two metastatic organ sites and TMTV > 56 cm3; the latter was the only independent feature at multivariate analysis. Patients achieving a CMR were associated with a prolonged PFS compared with those with PMR (median PFS 42.9 vs 8.8 months; p = 0.009). Disease progression occurred in new-onset disease sites in 87.5% of CMR, 7.1% of PMR and 34.8% of PMD/SMD (p < 0.001). High baseline TMTV and lack of treatment response were independent prognostic factors for OS, stratifying patients in three different prognostic classes (median OS 6.7, 18.3 and 102.2 months, respectively). CONCLUSIONS Baseline TMTV and metabolic response may be useful prognostic indicators for PFS and OS in patients with advanced melanoma treated with BRAF/MEK inhibitors. KEY POINTS • In a retrospective cohort of 57 metastatic melanoma patients treated with BRAF/MEK inhibitors, a TMTV > 56 cm3 at baseline [18F]FDG PET/CT was significantly correlated with a shorter PFS and OS. • The combined use of baseline TMTV along with PET response during treatment allowed for the identification of three groups of patients with very different median OS.
Collapse
Affiliation(s)
- Alessio Annovazzi
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy.
| | - Virginia Ferraresi
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
- Sarcomas and Rare Tumors Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Sandra Rea
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| | - Michelangelo Russillo
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Davide Renna
- First Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Silvia Carpano
- Second Division of Medical Oncology, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Rosa Sciuto
- Nuclear Medicine Unit, IRCCS - Regina Elena National Cancer Institute, Via Elio Chianesi, 53, 00144, Rome, Italy
| |
Collapse
|
13
|
Humbert O, Chardin D. Dissociated Response in Metastatic Cancer: An Atypical Pattern Brought Into the Spotlight With Immunotherapy. Front Oncol 2020; 10:566297. [PMID: 33072599 PMCID: PMC7531255 DOI: 10.3389/fonc.2020.566297] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 08/12/2020] [Indexed: 12/15/2022] Open
Abstract
When evaluating metastatic tumor response to systemic therapies, dissociated response is defined as the coexistence of responding and non-responding lesions within the same patient. Although commonly observed on interim whole-body imaging, the current response criteria in solid cancer do not consider this evolutive pattern, which is, by default, assimilated to progression. With targeted therapies and chemotherapies, dissociated response is observed with different frequencies, depending on the primary cancer type, treatment, and imaging modality. Because FDG PET/CT can easily assess response on a lesion-by-lesion basis, thus quickly revealing response heterogeneity, a PET/CT dissociated response has been described in up to 48% of women treated for a metastatic breast cancer. Although some studies have underlined a specific prognostic of dissociated response, it has always ended up being described as an unfavorable prognostic pattern and therefore assimilated to the “Progressive Disease” category of RECIST/PERCIST. This dichotomous imaging report (response vs. progression) provides a simple information for clinical decision-support, which probably explains the relatively low consideration for the dissociated response pattern to chemotherapies and targeted therapies until now. With immune checkpoint inhibitors, this paradigm is quickly changing. Dissociated response is observed in around 10% of advanced lung cancer patients and appears to be associated to treatment efficiency. Indeed, for this subset of patients, a clinical benefit of immunotherapy and favorable prognosis are usually observed. This specific pattern should therefore be considered in the future immunotherapy-adapted criteria for response evaluation using CT and PET/CT, and specific clinical managements should be evaluated for this response pattern.
Collapse
Affiliation(s)
- Olivier Humbert
- Department of Nuclear Medicine, Centre Antoine-Lacassagne, Université Côte d'Azur, Nice, France.,TIRO-UMR E 4320, Université Côte d'Azur, Nice, France
| | - David Chardin
- Department of Nuclear Medicine, Centre Antoine-Lacassagne, Université Côte d'Azur, Nice, France.,TIRO-UMR E 4320, Université Côte d'Azur, Nice, France
| |
Collapse
|
14
|
Bisschop C, de Heer E, Brouwers A, Hospers G, Jalving M. Rational use of 18F-FDG PET/CT in patients with advanced cutaneous melanoma: A systematic review. Crit Rev Oncol Hematol 2020; 153:103044. [DOI: 10.1016/j.critrevonc.2020.103044] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 06/13/2020] [Accepted: 06/29/2020] [Indexed: 02/07/2023] Open
|
15
|
Chang GA, Wiggins JM, Corless BC, Syeda MM, Tadepalli JS, Blake S, Fleming N, Darvishian F, Pavlick A, Berman R, Shapiro R, Shao Y, Karlin-Neumann G, Spittle C, Osman I, Polsky D. TERT, BRAF, and NRAS Mutational Heterogeneity between Paired Primary and Metastatic Melanoma Tumors. J Invest Dermatol 2020; 140:1609-1618.e7. [PMID: 32087194 PMCID: PMC7387168 DOI: 10.1016/j.jid.2020.01.027] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 12/06/2019] [Accepted: 01/06/2020] [Indexed: 11/26/2022]
Abstract
Mutational heterogeneity can contribute to therapeutic resistance in solid cancers. In melanoma, the frequencies of intertumoral and intratumoral heterogeneity are controversial. We examined mutational heterogeneity within individual patients with melanoma using multiplatform analysis of commonly mutated driver and nonpassenger genes. We analyzed paired primary and metastatic tumors from 60 patients and multiple metastatic tumors from 39 patients whose primary tumors were unavailable (n = 271 tumors). We used a combination of multiplex SNaPshot assays, Sanger sequencing, mutation-specific PCR, or droplet digital PCR to determine the presence of BRAFV600, NRASQ61, TERT-124C>T, and TERT-146C>T mutations. Mutations were detected in BRAF (39%), NRAS (21%), and/or TERT (78%). Thirteen patients had TERTmutant discordant tumors; seven of these had a single tumor with both TERT-124C>T and TERT-146C>T mutations present at different allele frequencies. Two patients had both BRAF and NRAS mutations; one had different tumors and the other had a single tumor with both mutations. One patient with a BRAFmutant primary lacked mutant BRAF in at least one of their metastases. Overall, we identified mutational heterogeneity in 18 of 99 patients (18%). These results suggest that some primary melanomas may be composed of subclones with differing mutational profiles. Such heterogeneity may be relevant to treatment responses and survival outcomes.
Collapse
Affiliation(s)
- Gregory A Chang
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; St. Georges University School of Medicine, Grenada, West Indies
| | - Jennifer M Wiggins
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Broderick C Corless
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Weill Cornell Medicine Graduate School of Medical Sciences, New York, USA
| | - Mahrukh M Syeda
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Jyothirmayee S Tadepalli
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Shria Blake
- MolecularMD Corporation, Portland, Oregon, USA
| | - Nathaniel Fleming
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Farbod Darvishian
- The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Department of Pathology, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Anna Pavlick
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Division of Medical Oncology, Department of Medicine, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Russell Berman
- The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Department of Surgery, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Richard Shapiro
- The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Department of Surgery, New York University School of Medicine, NYU Langone Health, New York, USA
| | - Yongzhao Shao
- The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Department of Population Health, New York University School of Medicine, NYU Langone Health, New York, USA
| | | | | | - Iman Osman
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA; Division of Medical Oncology, Department of Medicine, New York University School of Medicine, NYU Langone Health, New York, USA
| | - David Polsky
- The Ronald O. Perelman Department of Dermatology, New York University School of Medicine, NYU Langone Health, New York, USA; The Laura and Isaac Perlmutter Cancer Center, New York University School of Medicine, NYU Langone Health, New York, USA.
| |
Collapse
|
16
|
Nuclear Medicine Imaging Techniques in Melanoma. Clin Nucl Med 2020. [DOI: 10.1007/978-3-030-39457-8_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
17
|
Tan AC, Emmett L, Lo S, Liu V, Kapoor R, Carlino MS, Guminski AD, Long GV, Menzies AM. FDG-PET response and outcome from anti-PD-1 therapy in metastatic melanoma. Ann Oncol 2019; 29:2115-2120. [PMID: 30137228 DOI: 10.1093/annonc/mdy330] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Background Immune checkpoint inhibitor therapy has resulted in impressive and durable clinical activity for many cancers including melanoma; however, there remain few reliable predictors for long-term response. This study investigated whether [18F]2-fluoro-2-deoxy-D-glucose (FDG-PET) imaging may better predict long-term outcomes compared with standard computed tomography (CT) response criteria. Patients and methods Retrospective analysis of metastatic melanoma patients treated with anti-PD-1-based immunotherapy with baseline and 1-year FDG-PET and CT imaging at Melanoma Institute Australia. One-year response was determined using RECIST for CT and EORTC criteria for PET, coded as complete response (CR or CMR), partial response (PR or PMR), stable disease (SD or SMD) or progressive disease (PD or PMD). Progression-free survival (PFS) was determined from the 1-year landmark. Results Patients (n = 104) were evaluated with median follow-up 30.1 months and 98% remain alive. Most received anti-PD-1 as monotherapy (67%) or combined with ipilimumab (31%). At 1 year, 28% had CR, 66% had PR and 6% had SD on CT, while 75% had CMR, 16% PMR and 9% SMD/PMD on PET. CMR was observed in 68% of patients with PR on CT. RECIST PFS post 1-year landmark was similar in patients with CR versus PR/SD, but improved in patients with CMR versus non-CMR {median not reached [NR] versus 12.8 month; hazard ratio [HR] 0.06 [95% confidence interval (CI) 0.02-0.23]; P < 0.01}. In patients with PR on CT, PFS was improved in patients with PR + CMR versus PR + non-CMR (median NR versus 12.8 months; HR 0.07 [95% CI 0.02-0.27]; P < 0.01). In the 78 CMR patients, 78% had discontinued treatment and 96% had ongoing response. Conclusions Whilst only a small proportion of patients have a CR at 1 year, most patients with a PR have CMR on PET. Almost all patients with CMR at 1 year have ongoing response to therapy thereafter. PET may have utility in predicting long-term benefit and help guide discontinuation of therapy.
Collapse
Affiliation(s)
- A C Tan
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia
| | - L Emmett
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Nuclear Medicine, St Vincent's Hospital, Sydney, Australia; The University of New South Wales, Sydney, Australia
| | - S Lo
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia
| | - V Liu
- Department of Nuclear Medicine, St Vincent's Hospital, Sydney, Australia
| | - R Kapoor
- Department of Radiology, Royal Prince Alfred Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - M S Carlino
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, Sydney, Australia
| | - A D Guminski
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - G V Long
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia
| | - A M Menzies
- Melanoma Institute Australia and The University of Sydney, Sydney, Australia; Department of Medical Oncology, Northern Sydney Cancer Centre, Royal North Shore Hospital, Sydney, Australia; Mater Hospital, Sydney, Australia.
| |
Collapse
|
18
|
18F-FDG PET/CT longitudinal studies in patients with advanced metastatic melanoma for response evaluation of combination treatment with vemurafenib and ipilimumab. Melanoma Res 2019; 29:178-186. [PMID: 30653029 DOI: 10.1097/cmr.0000000000000541] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Sixteen BRAF-mutation positive, metastatic melanoma patients with highly advanced disease received combination therapy of vemurafenib and ipilimumab as an individual treatment decision. Our aim was to assess the role of fluorine-18-fluorodeoxyglucose (F-FDG) PET/computed tomography (PET/CT) in the evaluation of the clinical benefit (CB) of this combination treatment. After clinical improvement under vemurafenib monotherapy, four cycles of ipilimumab were additionally administered. F-FDG PET/CT was performed before the start, after two cycles and after completion of the combined ipilimumab/vemurafenib treatment. PET-based patient response evaluation to treatment was based on the European Organization for Research and Treatment of Cancer and the PET Response Evaluation Criteria for Immunotherapy criteria. Progression-free survival (PFS) from the end of combination treatment was calculated. According to their best clinical response at the end of combination treatment, eight patients showed CB and eight patients had no-CB. Two patients revealed extraordinary good clinical outcome with PFS of more than 5 years. Overall, 13 out of 16 patients were correctly classified by the European Organization for Research and Treatment of Cancer and 15 out of 16 by the PET Response Evaluation Criteria for Immunotherapy criteria. Median PFS was 8.8 months among PET-responders and 3.6 months among nonresponders. During immunotherapy administration seven patients developed radiologic signs of immune-related adverse events (irAEs), with colitis and arthritis being the most frequent ones; these patients had a significantly longer PFS than those without irAEs (P=0.036). F-FDG PET/CT is a valuable tool for the evaluation of patients receiving a combination of targeted treatment and immunotherapy. The appearance of irAEs on PET/CT might correlate with benefit to immunotherapy.
Collapse
|
19
|
Hoffend J, Sachpekidis C, Dimitrakopoulou-Strauss A. [Response evaluation in nuclear medicine : Criteria, results and pitfalls]. Radiologe 2019; 57:834-839. [PMID: 28875325 DOI: 10.1007/s00117-017-0295-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
CLINICAL/METHODICAL ISSUE Established criteria to categorize metabolic tumor response to cytotoxic chemotherapies may not be suited to capture the effects of therapy with immune checkpoint inhibitors (ICI) or with kinase inhibitors (KI), such as BRAF or MEK inhibitors. NUCLEAR MEDICINE STANDARD METHODS To assess the metabolic response to cytotoxic chemotherapy by positron emission tomography (PET) with 18F-fluorodeoxyglucose (FDG), the criteria of the European Organization for Research and Treatment of Cancer (EORTC) and the positron emission tomography response criteria in solid tumors (PERCIST) were conceived. The salient features of both criteria are detailed in a comparative way. PERFORMANCE AND ACHIEVEMENTS To date only retrospective data exist for the evaluation of therapies with either ICI or KI. They show that response to ICI cannot be reliably determined using the established criteria. Employing the EORTC criteria the responses to KI can be adequately ascertained so that the metabolic tumor response in FDG-PET is regarded as a surrogate marker for the efficacy of these drugs. PRACTICAL RECOMMENDATIONS Tumor response to therapy with ICI cannot at present be assessed with FDG-PET. Responses to BRAF and MEK inhibitors are, however, assessable using the criteria that were originally developed to evaluate responses to cytotoxic chemotherapy.
Collapse
Affiliation(s)
- J Hoffend
- Onkologische Diagnostik/PET-CT, Zentralinstitut für diagnostische und interventionelle Radiologie, Klinikum der Stadt Ludwigshafen am Rhein gGmbH, Bremserstraße 79, 67063, Ludwigshafen, Deutschland.
| | - C Sachpekidis
- Klinische Kooperationseinheit Nuklearmedizin, Forschungsschwerpunkt Bildgebung und Radiologie, Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Deutschland.,Abteilung Radiologie, Forschungsschwerpunkt Bildgebung und Radiologie, Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Deutschland
| | - A Dimitrakopoulou-Strauss
- Klinische Kooperationseinheit Nuklearmedizin, Forschungsschwerpunkt Bildgebung und Radiologie, Deutsches Krebsforschungszentrum Heidelberg, Heidelberg, Deutschland
| |
Collapse
|
20
|
Konieczkowski DJ, Johannessen CM, Garraway LA. A Convergence-Based Framework for Cancer Drug Resistance. Cancer Cell 2018; 33:801-815. [PMID: 29763622 PMCID: PMC5957297 DOI: 10.1016/j.ccell.2018.03.025] [Citation(s) in RCA: 158] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 02/02/2018] [Accepted: 03/26/2018] [Indexed: 02/07/2023]
Abstract
Despite advances in cancer biology and therapeutics, drug resistance remains problematic. Resistance is often multifactorial, heterogeneous, and prone to undersampling. Nonetheless, many individual mechanisms of targeted therapy resistance may coalesce into a smaller number of convergences, including pathway reactivation (downstream re-engagement of original effectors), pathway bypass (recruitment of a parallel pathway converging on the same downstream output), and pathway indifference (development of a cellular state independent of the initial therapeutic target). Similar convergences may also underpin immunotherapy resistance. Such parsimonious, convergence-based frameworks may help explain resistance across tumor types and therapeutic categories and may also suggest strategies to overcome it.
Collapse
|
21
|
Theodosakis N, Micevic G, Langdon CG, Ventura A, Means R, Stern DF, Bosenberg MW. p90RSK Blockade Inhibits Dual BRAF and MEK Inhibitor-Resistant Melanoma by Targeting Protein Synthesis. J Invest Dermatol 2017; 137:2187-2196. [PMID: 28599981 PMCID: PMC6342201 DOI: 10.1016/j.jid.2016.12.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 11/23/2016] [Accepted: 12/02/2016] [Indexed: 01/05/2023]
Abstract
Despite improvements in survival in metastatic melanoma with combined BRAF and mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor treatment, the overwhelming majority of patients eventually acquire resistance to both agents. Consequently, new targets for therapy in resistant tumors are currently being evaluated. Previous studies have identified p90 subfamily of ribosomal S6 kinase (p90RSK) family kinases as key factors for growth and proliferation, as well as protein synthesis via assembly of the 7-methyl-guanosine triphosphate cap-dependent translation complex. We sought to evaluate inhibitors of p90RSK family members: BI-D1870 and BRD7389, for their ability to inhibit both proliferation and protein synthesis in patient-derived melanoma cell lines with acquired resistance to combined treatment with the BRAF inhibitor vemurafenib and the mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor selumetinib. We found that the RSK inhibitors blocked cell proliferation and protein synthesis in multiple dual-resistant melanoma lines. In addition, single agent RSK inhibitor treatment was effective in drug-naïve lines, two of which are innately vemurafenib resistant. We also used Reverse Phase Protein Array screening to identify differential protein expression that correlates with BI-D1870 sensitivity, and identified prognostic biomarkers for survival in human melanoma patients. These findings establish p90RSK inhibition as a therapeutic strategy in treatment-resistant melanoma and provide insight into the mechanism of action.
Collapse
Affiliation(s)
| | - Goran Micevic
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Casey G Langdon
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Alessandra Ventura
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Dermatology, University of Rome Tor Vergata, Rome, Italy
| | - Robert Means
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - David F Stern
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Marcus W Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
22
|
Abstract
Recent advances in genomic profiling and sequencing of melanoma have provided new insights into the development of the basis for molecular biology to more accurately subgroup patients with melanoma. The development of novel mutation-targeted and immunomodulation therapy as a major component of precision oncology has revolutionized the management and outcome of patients with metastatic melanoma. PET imaging plays an important role in noninvasively assessing the tumor biological behavior, to guide individualized treatment and assess response to therapy. This review summarizes the recent genomic discoveries in melanoma in the era of targeted therapy and their implications for functional PET imaging.
Collapse
Affiliation(s)
- Esther Mena
- Molecular Imaging Program, National Cancer Institute, National Institutes of Health, 9000 Rockville Pike, Building 10, Room B3B402, Bethesda, MD 20892-1763, USA.
| | - Yasemin Sanli
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles Marcus
- The Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, 601 North Caroline Street, Baltimore, MD 21231, USA
| | - Rathan M Subramaniam
- Department of Radiology, The University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Clinical Sciences, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Biomedical Engineering, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Advanced Imaging Research Center, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA
| |
Collapse
|
23
|
Stodell M, Thompson JF, Emmett L, Uren RF, Kapoor R, Saw RPM. Melanoma patient imaging in the era of effective systemic therapies. Eur J Surg Oncol 2017. [PMID: 28625798 DOI: 10.1016/j.ejso.2017.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Imaging plays a critical role in the current multi-disciplinary management of patients with melanoma. It is used for primary disease staging, surgical planning, and surveillance in high-risk patients, and for monitoring the effects of systemic or loco-regional therapies. Several different imaging modalities have been utilised in the past. Contemporary imaging practises vary geographically depending on clinical guidelines, physician preferences, availability and cost. Targeted therapies and immunotherapies have revolutionised the treatment of patients with metastatic melanoma over the last few years. With this have come new patterns of disease that were not observed after conventional therapies, and new criteria to assess therapeutic responses. In this article we review the role of imaging for patients with melanoma in the era of effective systemic therapies and discuss likely future developments.
Collapse
Affiliation(s)
- M Stodell
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia
| | - J F Thompson
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Discipline of Surgery, The University of Sydney, Sydney, NSW, Australia; Division of Surgery, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - L Emmett
- Garvan Institute of Medical Research, Discipline of Medicine, The University of New South Wales, Sydney, NSW, Australia
| | - R F Uren
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Alfred Nuclear Medicine and Ultrasound, Newtown, NSW, Australia
| | - R Kapoor
- Mater Imaging, The Mater Hospital Sydney, North Sydney, NSW, Australia
| | - R P M Saw
- Melanoma Institute Australia, The University of Sydney, Sydney, NSW, Australia; Sydney Medical School, The University of Sydney, Sydney, NSW, Australia; Discipline of Surgery, The University of Sydney, Sydney, NSW, Australia; Division of Surgery, Royal Prince Alfred Hospital, Camperdown, NSW, Australia.
| |
Collapse
|
24
|
The Advantages and Challenges of Using FDG PET/CT for Response Assessment in Melanoma in the Era of Targeted Agents and Immunotherapy. Eur J Nucl Med Mol Imaging 2017; 44:67-77. [DOI: 10.1007/s00259-017-3691-7] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 03/23/2017] [Indexed: 12/15/2022]
|
25
|
Wolsztynski E, O'Sullivan F, O'Sullivan J, Eary JF. Statistical assessment of treatment response in a cancer patient based on pre-therapy and post-therapy FDG-PET scans. Stat Med 2016; 36:1172-1200. [PMID: 27990685 DOI: 10.1002/sim.7198] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 07/28/2016] [Accepted: 11/20/2016] [Indexed: 12/12/2022]
Abstract
This work arises from consideration of sarcoma patients in which fluorodeoxyglucose positron emission tomography (FDG-PET) imaging pre-therapy and post-chemotherapy is used to assess treatment response. Our focus is on methods for evaluation of the statistical uncertainty in the measured response for an individual patient. The gamma distribution is often used to describe data with constant coefficient of variation, but it can be adapted to describe the pseudo-Poisson character of PET measurements. We propose co-registering the pre-therapy and post- therapy images and modeling the approximately paired voxel-level data using the gamma statistics. Expressions for the estimation of the treatment effect and its variability are provided. Simulation studies explore the performance in the context of testing for a treatment effect. The impact of misregistration errors and how test power is affected by estimation of variability using simplified sampling assumptions, as might be produced by direct bootstrapping, is also clarified. The results illustrate a marked benefit in using a properly constructed paired approach. Remarkably, the power of the paired analysis is maintained even if the pre-image and post- image data are poorly registered. A theoretical explanation for this is indicated. The methodology is further illustrated in the context of a series of fluorodeoxyglucose-PET sarcoma patient studies. These data demonstrate the additional prognostic value of the proposed treatment effect test statistic. Copyright © 2016 John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- E Wolsztynski
- School of Mathematical Sciences, University College Cork, Cork, Ireland
| | - F O'Sullivan
- School of Mathematical Sciences, University College Cork, Cork, Ireland
| | - J O'Sullivan
- School of Mathematical Sciences, University College Cork, Cork, Ireland
| | - J F Eary
- Department of Radiology, University of Alabama, Birmingham, U.S.A
| |
Collapse
|
26
|
(18)F-FDG PET/CT and Melanoma: Staging, Immune Modulation and Mutation-Targeted Therapy Assessment, and Prognosis. AJR Am J Roentgenol 2015. [PMID: 26204273 DOI: 10.2214/ajr.14.13575] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Monoclonal antibodies that target the programmed cell death 1 (PD-1) immune checkpoint protein and its associated ligands, PD-L1 and PD-L2, and targeted inhibitors of mutated signal transduction molecules such as BRAF inhibitors show immense promise in treating patients with melanoma. We discuss the use of (18)F-FDG PET/CT for assessing therapy effectiveness, staging advanced disease, and determining prognosis of patients with melanoma. CONCLUSION FDG PET/CT is useful in staging disease, assessing therapy, and determining prognosis in patients with melanoma.
Collapse
|
27
|
Theodosakis N, Held MA, Marzuka-Alcala A, Meeth KM, Micevic G, Long GV, Scolyer RA, Stern DF, Bosenberg MW. BRAF Inhibition Decreases Cellular Glucose Uptake in Melanoma in Association with Reduction in Cell Volume. Mol Cancer Ther 2015; 14:1680-92. [PMID: 25948295 DOI: 10.1158/1535-7163.mct-15-0080] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 04/30/2015] [Indexed: 12/30/2022]
Abstract
BRAF kinase inhibitors have dramatically affected treatment of BRAF(V600E) (/) (K)-driven metastatic melanoma. Early responses assessed using [(18)F]fluorodeoxyglucose uptake-positron emission tomography (FDG-PET) have shown dramatic reduction of radiotracer signal within 2 weeks of treatment. Despite high response rates, relapse occurs in nearly all cases, frequently at sites of treated metastatic disease. It remains unclear whether initial loss of (18)FDG uptake is due to tumor cell death or other reasons. Here, we provide evidence of melanoma cell volume reduction in a patient cohort treated with BRAF inhibitors. We present data demonstrating that BRAF inhibition reduces melanoma glucose uptake per cell, but that this change is no longer significant following normalization for cell volume changes. We also demonstrate that volume normalization greatly reduces differences in transmembrane glucose transport and hexokinase-mediated phosphorylation. Mechanistic studies suggest that this loss of cell volume is due in large part to decreases in new protein translation as a consequence of vemurafenib treatment. Ultimately, our findings suggest that cell volume regulation constitutes an important physiologic parameter that may significantly contribute to radiographic changes observed in clinic.
Collapse
Affiliation(s)
| | - Matthew A Held
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | | | - Katrina M Meeth
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Goran Micevic
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Georgina V Long
- Melanoma Institute of Australia, Sydney, New South Wales, Australia. Discipline of Pathology, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard A Scolyer
- Melanoma Institute of Australia, Sydney, New South Wales, Australia. Discipline of Medicine, The University of Sydney, Sydney, New South Wales, Australia
| | - David F Stern
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut
| | - Marcus W Bosenberg
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut. Department of Dermatology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
28
|
Verlinden I, van den Hurk K, Clarijs R, Willig AP, Stallinga CMHA, Roemen GMJM, van den Oord JJ, Zur Hausen A, Speel EJM, Winnepenninckx VJL. BRAFV600E immunopositive melanomas show low frequency of heterogeneity and association with epithelioid tumor cells: a STROBE-compliant article. Medicine (Baltimore) 2014; 93:e285. [PMID: 25526463 PMCID: PMC4603111 DOI: 10.1097/md.0000000000000285] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 10/10/2014] [Accepted: 10/28/2014] [Indexed: 12/26/2022] Open
Abstract
Treatment of BRAFV600E-mutant melanoma by small molecule inhibitors that target BRAFV600E or MEK kinases is increasingly used in clinical practice and significantly improve patient outcome. However, patients eventually become resistant and therapeutic improvement is required. Molecular diversity within individual tumors (intratumor heterogeneity) and between tumors within a single patient (intrapatient heterogeneity) poses a significant challenge to precision medicine. Using immunohistochemistry, we determined the extent of BRAFV600E intratumor and intrapatient heterogeneity and the influence of morphological heterogeneity in a large series of 171 melanomas of 81 patients. The BRAFV600E mutation rate found in our melanoma series is 44%, with none of 22 (0%) melanoma in situ, 23 of 56 (41%) primary tumors, 28 of 59 (48%) regional metastases, and 24 of 34 (71%) distant metastases harboring the mutation. In general, a diffuse homogeneous immunostaining was seen, even in tumors consisting of more than one cell type, that is, epithelioid, spindle, and/or small cell types. Nevertheless, BRAFV600E-mutant melanomas more often had a purely epithelioid cell population (P=0.063), that is more evident among distant metastases (P=0.014). Only two of 75 (3%) mutated specimens (one primary and one metastasis) displayed heterogeneous BRAFV600E expression. The primary tumor was also morphologically heterogeneous and exclusively displayed BRAFV600E in the epithelioid component, confirming an association between BRAFV600E and epithelioid cells. Twenty-eight of 30 patients (93%) had concordant BRAFV600E mutation status between their tumors. Taken together, BRAFV600E intratumor and intrapatient heterogeneity in melanoma is diminutive, nevertheless, the identified exceptions will have important implications for the clinical management of this disease.
Collapse
Affiliation(s)
- Ivana Verlinden
- From the Department of Pathology, Maastricht University Medical Centre, Maastricht, The Netherlands (IV, KvdH, CMHAS, GMJMR, AzH, E-JMS, VJLW); GROW-School for Oncology and Developmental Biology, Maastricht University Medical Centre, Maastricht, The Netherlands (KvdH, AzH, E-JMS, VJLW); Department of Clinical Pathology, Atrium Medical Centre Parkstad, Heerlen, The Netherlands (RC); Department of Pathology, St. Laurentius ziekenhuis, Roermond, The Netherlands (APW); and Laboratory of Translational Cell & Tissue Research and Department of Pathology, University Hospital, KULeuven, Leuven, Belgium (JJvdO)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Carlino MS, Fung C, Shahheydari H, Todd JR, Boyd SC, Irvine M, Nagrial AM, Scolyer RA, Kefford RF, Long GV, Rizos H. Preexisting MEK1P124 mutations diminish response to BRAF inhibitors in metastatic melanoma patients. Clin Cancer Res 2014; 21:98-105. [PMID: 25370473 DOI: 10.1158/1078-0432.ccr-14-0759] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND MEK1 mutations in melanoma can confer resistance to BRAF inhibitors, although preexisting MEK1(P124) mutations do not preclude clinical responses. We sought to determine whether recurrent, preexisting MEK1(P124) mutations affected clinical outcome in BRAF inhibitor-treated patients with melanoma. METHODS Data from four published datasets were analyzed to determine whether preexisting MEK1(P124) mutations affect radiologic response or progression-free survival (PFS) in patients with BRAF(V600)-mutant metastatic melanoma treated with vemurafenib or dabrafenib. The effects of MEK1(P124) mutations on MAPK pathway activity and response to BRAF inhibition were also investigated in a series of cell models. RESULTS In a pooled analysis of 123 patients, the presence of a pretreatment MEK1(P124) mutation (N = 12, 10%) was associated with a poorer RECIST response (33% vs. 72% in MEK1(P124Q/S) vs. MEK1(P124) wild-type, P = 0.018), and a shorter PFS (median 3.1 vs. 4.8 months, P = 0.004). Furthermore, MEK1(P124Q/S) mutations were shown to have independent kinase activity and introduction of these mutations into a BRAF-mutant melanoma cell line diminished inhibition of ERK phosphorylation by dabrafenib and enhanced clonogenic survival in the presence of dabrafenib compared with cells ectopically expressing wild-type MEK1. Consistent with these data, two BRAF-mutant cell lines with endogenous MEK1(P124) mutations showed intermediate sensitivity to dabrafenib, but were highly sensitive to downstream inhibition of MEK or ERK. CONCLUSION Taken together, our data indicate that preexisting MEK1(P124) mutations are associated with a reduced response to BRAF inhibitor therapy and identify a subset of patients with BRAF-mutant melanoma likely to benefit from combination therapies involving MEK or ERK inhibitors.
Collapse
Affiliation(s)
- Matteo S Carlino
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia. Department of Medical Oncology, Crown Princess Mary Cancer Centre, Westmead and Blacktown Hospitals, New South Wales, Australia. Melanoma Institute Australia, Sydney, New South Wales, Australia
| | - Carina Fung
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia. Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia
| | - Hamideh Shahheydari
- Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia
| | - Jason R Todd
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia
| | - Suzanah C Boyd
- Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia
| | - Mal Irvine
- Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia
| | - Adnan M Nagrial
- The Kinghorn Cancer Centre, Cancer Research Program, Garvan Institute of Medical Research, Sydney, Australia
| | - Richard A Scolyer
- Melanoma Institute Australia, Sydney, New South Wales, Australia. Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia. Department of Tissue Pathology and Diagnostic Oncology Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Richard F Kefford
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia. Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia. Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, New South Wales, Australia. Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Helen Rizos
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead, New South Wales, Australia. Australian School of Advanced Medicine, Macquarie University, New South Wales, Australia. Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
30
|
Menzies AM, Long GV. Systemic treatment for BRAF-mutant melanoma: where do we go next? Lancet Oncol 2014; 15:e371-81. [DOI: 10.1016/s1470-2045(14)70072-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
31
|
The pharmacological impact of ATP-binding cassette drug transporters on vemurafenib-based therapy. Acta Pharm Sin B 2014; 4:105-11. [PMID: 26579371 PMCID: PMC4590304 DOI: 10.1016/j.apsb.2013.12.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2013] [Revised: 12/02/2013] [Accepted: 12/10/2013] [Indexed: 01/06/2023] Open
Abstract
Melanoma is the most serious type of skin cancer and one of the most common cancers in the world. Advanced melanoma is often resistant to conventional therapies and has high potential for metastasis and low survival rates. Vemurafenib is a small molecule inhibitor of the BRAF serine-threonine kinase recently approved by the United States Food and Drug Administration to treat patients with metastatic and unresectable melanomas that carry an activating BRAF (V600E) mutation. Many clinical trials evaluating other therapeutic uses of vemurafenib are still ongoing. The ATP-binding cassette (ABC) transporters are membrane proteins with important physiological and pharmacological roles. Collectively, they transport and regulate levels of physiological substrates such as lipids, porphyrins and sterols. Some of them also remove xenobiotics and limit the oral bioavailability and distribution of many chemotherapeutics. The overexpression of three major ABC drug transporters is the most common mechanism for acquired resistance to anticancer drugs. In this review, we highlight some of the recent findings related to the effect of ABC drug transporters such as ABCB1 and ABCG2 on the oral bioavailability of vemurafenib, problems associated with treating melanoma brain metastases and the development of acquired resistance to vemurafenib in cancers harboring the BRAF (V600E) mutation.
Collapse
Key Words
- ABC transporter
- ABC, ATP-binding cassette
- AML, acute myeloid leukemia
- BBB, blood–brain barrier
- CNS, central nervous system
- CSCs, cancer stem cells
- Drug resistance
- GI, gastrointestinal
- MAPK, mitogen-activated protein kinase
- MDR, multidrug resistance
- Melanoma
- NBDs, nucleotide-binding domains
- P-glycoprotein
- PFS, longer progression-free survival
- PKIs, protein kinase inhibitors
- TKIs, tyrosine kinase inhibitors
- TMDs, transmembrane domains
- Vemurafenib
Collapse
|
32
|
Parmenter TJ, Kleinschmidt M, Kinross KM, Bond ST, Li J, Kaadige MR, Rao A, Sheppard KE, Hugo W, Pupo GM, Pearson RB, McGee SL, Long GV, Scolyer RA, Rizos H, Lo RS, Cullinane C, Ayer DE, Ribas A, Johnstone RW, Hicks RJ, McArthur GA. Response of BRAF-mutant melanoma to BRAF inhibition is mediated by a network of transcriptional regulators of glycolysis. Cancer Discov 2014; 4:423-33. [PMID: 24469106 DOI: 10.1158/2159-8290.cd-13-0440] [Citation(s) in RCA: 215] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UNLABELLED Deregulated glucose metabolism fulfills the energetic and biosynthetic requirements for tumor growth driven by oncogenes. Because inhibition of oncogenic BRAF causes profound reductions in glucose uptake and a strong clinical benefit in BRAF-mutant melanoma, we examined the role of energy metabolism in responses to BRAF inhibition. We observed pronounced and consistent decreases in glycolytic activity in BRAF-mutant melanoma cells. Moreover, we identified a network of BRAF-regulated transcription factors that control glycolysis in melanoma cells. Remarkably, this network of transcription factors, including hypoxia-inducible factor-1α, MYC, and MONDOA (MLXIP), drives glycolysis downstream of BRAF(V600), is critical for responses to BRAF inhibition, and is modulated by BRAF inhibition in clinical melanoma specimens. Furthermore, we show that concurrent inhibition of BRAF and glycolysis induces cell death in BRAF inhibitor (BRAFi)-resistant melanoma cells. Thus, we provide a proof-of-principle for treatment of melanoma with combinations of BRAFis and glycolysis inhibitors. SIGNIFICANCE BRAF is suppress glycolysis and provide strong clinical benefi t in BRAF V600 melanoma. We show that BRAF inhibition suppresses glycolysis via a network of transcription factors that are critical for complete BRAFi responses. Furthermore, we provide evidence for the clinical potential of therapies that combine BRAFis with glycolysis inhibitors.
Collapse
Affiliation(s)
- Tiffany J Parmenter
- 1Molecular Oncology Laboratory, Oncogenic Signaling and Growth Control Program, 2Translational Research Laboratory, Cancer Therapeutics Program, 3Bioinformatics Core Facility, 4The Cancer Signalling Laboratory, Oncogenic Signaling and Growth Control Program, 5Gene Regulation Laboratory, Cancer Therapeutics Program, 6Molecular Imaging and Targeted Therapeutics Laboratory, Cancer Therapeutics Program, 7Department of Cancer Imaging, Peter MacCallum Cancer Centre, East Melbourne; 8Sir Peter MacCallum Department of Oncology, Departments of 9Biochemistry and Molecular Biology, and 10Pathology, University of Melbourne, Parkville; 11Metabolic Remodelling Laboratory, Metabolic Research Unit, School of Medicine, Deakin University, Waurn Ponds; 12Department of Medicine, St Vincent's Hospital, University of Melbourne, Fitzroy, Victoria; 13Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead; 14Department of Tissue Pathology & Diagnostic Oncology, Royal Prince Alfred Hospital; 15Discipline of Pathology, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia; 16Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah; and 17Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, California
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Menzies AM, Haydu LE, Carlino MS, Azer MWF, Carr PJA, Kefford RF, Long GV. Inter- and intra-patient heterogeneity of response and progression to targeted therapy in metastatic melanoma. PLoS One 2014; 9:e85004. [PMID: 24400126 PMCID: PMC3882277 DOI: 10.1371/journal.pone.0085004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 11/27/2013] [Indexed: 12/22/2022] Open
Abstract
Background MAPK inhibitors (MAPKi) are active in BRAF-mutant metastatic melanoma patients, but the extent of response and progression-free survival (PFS) is variable, and complete responses are rare. We sought to examine the patterns of response and progression in patients treated with targeted therapy. Methods MAPKi-naïve patients treated with combined dabrafenib and trametinib had all metastases ≥5 mm (lymph nodes ≥15 mm in short axis) visible on computed tomography measured at baseline and throughout treatment. Results 24 patients had 135 measured metastases (median 4.5/patient, median diameter 16 mm). Time to best response (median 5.5 mo, range 1.7–20.1 mo), and the degree of best response (median −70%, range +9 to −100%) varied amongst patients. 17% of patients achieved complete response (CR), whereas 53% of metastases underwent CR, including 42% ≥10 mm. Metastases that underwent CR were smaller than non-CR metastases (median 11 vs 20 mm, P<0.001). PFS was variable among patients (median 8.2 mo, range 2.6–18.3 mo), and 50% of patients had disease progression in new metastases only. Only 1% (1/71) of CR-metastases subsequently progressed. Twelve-month overall survival was poorer in those with a more heterogeneous initial response to therapy than less heterogeneous (67% vs 93%, P = 0.009). Conclusion Melanoma response and progression with MAPKi displays marked inter- and intra-patient heterogeneity. Most metastases undergo complete response, yet only a small proportion of patients achieve an overall complete response. Similarly, disease progression often occurs only in a subset of the tumor burden, and often in new metastases alone. Clinical heterogeneity, likely reflecting molecular heterogeneity, remains a barrier to the effective treatment of melanoma patients.
Collapse
Affiliation(s)
- Alexander M. Menzies
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- * E-mail:
| | - Lauren E. Haydu
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
| | - Matteo S. Carlino
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
- Westmead Institute for Cancer Research, Westmead, Australia
| | - Mary W. F. Azer
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
| | - Peter J. A. Carr
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Department of Radiology, Sydney, Australia
| | - Richard F. Kefford
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
- Westmead Hospital, Crown Princess Mary Cancer Centre, Sydney, Australia
- Westmead Institute for Cancer Research, Westmead, Australia
| | - Georgina V. Long
- Melanoma Institute Australia, Sydney, Australia
- The University of Sydney, Sydney, Australia
| |
Collapse
|
34
|
Wilmott JS, Menzies AM, Haydu LE, Capper D, Preusser M, Zhang YE, Thompson JF, Kefford RF, von Deimling A, Scolyer RA, Long GV. BRAF(V600E) protein expression and outcome from BRAF inhibitor treatment in BRAF(V600E) metastatic melanoma. Br J Cancer 2013; 108:924-31. [PMID: 23403819 PMCID: PMC3590666 DOI: 10.1038/bjc.2013.29] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND To examine the association between level and patterns of baseline intra-tumoural BRAF(V600E) protein expression and clinical outcome of BRAF(V600E) melanoma patients treated with selective BRAF inhibitors. METHODS Fifty-eight BRAF(V600E) metastatic melanoma patients treated with dabrafenib or vemurafenib on clinical trials had pre-treatment tumour BRAF(V600E) protein expression immunohistochemically (IHC) assessed using the BRAF V600E mutant-specific antibody VE1. Sections were examined for staining intensity (score 1-3) and percentage of immunoreactive tumour cells, and from this an immunoreactive score (IRS) was derived (intensity × per cent positive/10). The presence of intra-tumoural heterogeneity for BRAF(V600E) protein expression was also assessed. BRAF(V600E) expression was correlated with RECIST response, time to best response (TTBR), progression-free survival (PFS) and overall survival (OS). RESULTS Expression was generally high (median IRS 28 (range 5-30)) and homogeneous (78%). Expression of mutated protein BRAF(V600E) as measured by intensity, per cent immunoreactive cells, or IRS did not correlate with RECIST response, TTBR, PFS or OS, including on multivariate analysis. Heterogeneity of staining was seen in 22% of cases and did not correlate with outcome. CONCLUSION In the current study population, IHC-measured pre-treatment BRAF(V600E) protein expression does not predict response or outcome to BRAF inhibitor therapy in BRAF(V600E) metastatic melanoma patients.
Collapse
Affiliation(s)
- J S Wilmott
- Melanoma Institute Australia, Sydney, New South Wales, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Menzies AM, Long GV, Murali R. Dabrafenib and its potential for the treatment of metastatic melanoma. Drug Des Devel Ther 2012; 6:391-405. [PMID: 23251089 PMCID: PMC3523565 DOI: 10.2147/dddt.s38998] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The purpose of this study is to review the development of BRAF inhibitors, with emphasis on the trials conducted with dabrafenib (GSK2118436) and the evolving role of dabrafenib in treatment for melanoma patients. Fifty percent of cutaneous melanomas have mutations in BRAF, resulting in elevated activity of the mitogen-activated protein kinase signaling pathway. Dabrafenib inhibits the mutant BRAF (BRAF(mut)) protein in melanomas with BRAF(V600E) and BRAF(V600K) genotypes. BRAF(V600E) metastatic melanoma patients who receive dabrafenib treatment exhibit high clinical response rates and compared with dacarbazine chemotherapy, progression-free survival. Efficacy has also been demonstrated in BRAF(V600K) patients and in those with brain metastases. Dabrafenib has a generally mild and manageable toxicity profile. Cutaneous squamous cell carcinomas and pyrexia are the most significant adverse effects. Dabrafenib appears similar to vemurafenib with regard to efficacy but it is associated with less toxicity. It is expected that new combinations of targeted drugs, such as the combination of dabrafenib and trametinib (GSK1120212, a MEK inhibitor), will provide higher response rates and more durable clinical benefit than dabrafenib monotherapy.
Collapse
Affiliation(s)
| | - Georgina V Long
- Melanoma Institute Australia, Sydney, New South Wales, Australia
- Westmead Institute for Cancer Research and Crown Princess Mary Cancer Centre Westmead, Sydney, New South Wales, Australia
| | - Rajmohan Murali
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogensis Program, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| |
Collapse
|