251
|
Yu J, Mahipal A, Kim R. Targeted Therapy for Advanced or Metastatic Cholangiocarcinoma: Focus on the Clinical Potential of Infigratinib. Onco Targets Ther 2021; 14:5145-5160. [PMID: 34720591 PMCID: PMC8550543 DOI: 10.2147/ott.s272208] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/06/2021] [Indexed: 12/20/2022] Open
Abstract
Cholangiocarcinoma is one of the most aggressive cancers, with a 5-year survival rate of 11-44% after surgical resection. However, there is no established systemic therapy after failure of the gemcitabine plus cisplatin first-line therapy with exception of FOLFOX. Fibroblast growth factor receptor (FGFR) genomic aberrations have been detected in cholangiocarcinoma, and targeting these genomic aberrations with FGFR inhibitors has shown remarkable clinical benefits in advanced cholangiocarcinoma. In this article, we provide up-to-date information on the clinical development of selective FGFR inhibitors in advanced cholangiocarcinoma, focusing on infigratinib. In a Phase 1 trial, infigratinib showed a safe profile. In a following Phase 2 trial, infigratinib showed remarkable efficacy in advanced cholangiocarcinoma with FGFR2 fusions or rearrangements, and the Food and Drug Administration (FDA) approved infigratinib for cholangiocarcinoma in May 2021 largely based on tumor response and duration of response. Currently infigratinib is on a Phase 3 trial (PROOF301) as a first-line setting compared to the GEMCIS therapy in advanced cholangiocarcinoma. Given that the FGFR genomic aberrations including FGFR2 fusions are rarely accompanied with other targetable mutations, infigratinib and other FGFR inhibitors are continuously expected to be the novel targeted agents in cholangiocarcinoma harboring these aberrations. Acquired resistance to infigratinib was reported in several recent studies which could potentially be a barrier to overcome. Active clinical trials including PROOF301 are expected to elucidate the clinical benefits of infigratinib in this disease. Infigratinib combined with immunotherapy is also a potential future direction of investigation in cholangiocarcinoma.
Collapse
Affiliation(s)
- James Yu
- Department of Internal Medicine, Adventhealth Orlando, Orlando, FL, USA
| | - Amit Mahipal
- Department of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Richard Kim
- Department of Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| |
Collapse
|
252
|
Gil-Rojas Y, Lasalvia P, Hernández F, Castañeda-Cardona C, Castrillón-Correa J, Herrera D, Rosselli D. Cost-Effectiveness of the Dabrafenib Schedule in Combination With Trametinib Compared With Other Targeted Therapies, Immunotherapy, and Dacarbazine for the Treatment of Unresectable or Metastatic Melanoma With BRAFV600 Mutation in Colombia. Value Health Reg Issues 2021; 26:182-190. [PMID: 34673349 DOI: 10.1016/j.vhri.2021.04.008] [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: 11/12/2020] [Revised: 04/06/2021] [Accepted: 04/26/2021] [Indexed: 10/20/2022]
Abstract
OBJECTIVES Advanced melanoma accounts for 4% of malignant skin tumors, and approximately 80% of deaths are attributed to it. The most frequent mutation of the RAF gene is BRAFV600, which has been associated with a worse prognosis. The objective of the research was to evaluate the cost-effectiveness of the combined regimen of dabrafenib plus trametinib (D + T) compared with other targeted therapies, immunotherapy, and dacarbazine for the treatment of unresectable/metastatic melanoma with BRAFV600 mutation from the perspective of the Colombian health system. METHODS A partitioned survival model with 3 states (progression-free survival, progression, and death) was used to evaluate the cost-effectiveness for a time horizon of 20 years. Owing to the perspective of the analysis, only direct medical costs were taken into account. The efficacy of the evaluated treatment and the comparators were measured in terms of overall survival and progression-free survival. All costs were expressed in Colombian pesos as of 2018, and outcomes and costs were discounted at 5% annually. Two analysis scenarios were considered, one in which only monitoring and follow-up costs were included in the progression phase and another in which costs of acquisition of possible treatment sequences were also included. RESULTS In the first scenario (without postprogression medication costs), the combined D + T regimen was a dominant alternative to vemurafenib + cobimetinib but was not a cost-effective option compared with vemurafenib, nivolumab, ipilimumab, nivolumab + ipilimumab, pembrolizumab, and dacarbazine. In the second scenario (with drug costs in postprogression), D + T was dominant compared with vemurafenib + cobimetinib and cost-effective compared with nivolumab and pembrolizumab. Compared with other schemes, the incremental cost-effectiveness ratio was above the threshold of 3 gross domestic product per capita. Probabilistic sensitivity analyses showed that a willingness-to-pay threshold of Col$56 484 300 (US$19 108) per quality-adjusted life-year would not be reached at the current price of schema in Colombia. CONCLUSIONS The combined scheme could be a cost-effective and even a cost-saving alternative to vemurafenib + cobimetinib, nivolumab, and pembrolizumab if the costs associated with the use of other medications are taken into account after progression to the first line of treatment. Compared with the other comparators, it produces a greater number of quality-adjusted life-years, but the incremental cost-effectiveness ratio is above that of the willingness to pay.
Collapse
Affiliation(s)
- Yaneth Gil-Rojas
- Department of Economic Studies, Neuroeconomix, Bogotá, Colombia.
| | | | | | | | | | | | - Diego Rosselli
- Faculty of Medicine, Department of Clinical Epidemiology and Biostatistics, Pontificia Universidad Javeriana, Hospital San Ignacio, Bogotá, Colombia
| |
Collapse
|
253
|
RICTOR Affects Melanoma Tumorigenesis and Its Resistance to Targeted Therapy. Biomedicines 2021; 9:biomedicines9101498. [PMID: 34680615 PMCID: PMC8533235 DOI: 10.3390/biomedicines9101498] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 12/22/2022] Open
Abstract
The network defined by phosphatidylinositol-3-kinase (PI3K), AKT, and mammalian target of rapamycin (mTOR) plays a major role in melanoma oncogenesis and has been implicated in BRAF inhibitor resistance. The central role of RICTOR (rapamycin-insensitive companion of mTOR) in this pathway has only recently begun to be unraveled. In the present study, we assessed the role of mTORC2/RICTOR in BRAF-mutated melanomas and their resistance to BRAF inhibition. We showed that RICTOR was significantly overexpressed in melanoma and associated with bad prognoses. RICTOR overexpression stimulated melanoma-initiating cells (MICs) with ‘stemness’ properties. We also showed that RICTOR contributed to melanoma resistance to BRAF inhibitors and rendered the cells very sensitive to mTORC2 inhibition. We highlighted a connection between mTORC2/RICTOR and STAT3 in resistant cells and revealed an interaction between RAS and RICTOR in resistant melanoma, which, when disrupted, impeded the proliferation of resistant cells. Therefore, as a key signaling node, RICTOR contributes to BRAF-dependent melanoma development and resistance to therapy and, as such, is a valuable therapeutic target in melanoma.
Collapse
|
254
|
Placke JM, Soun C, Bottek J, Herbst R, Terheyden P, Utikal J, Pföhler C, Ulrich J, Kreuter A, Pfeiffer C, Mohr P, Gutzmer R, Meier F, Dippel E, Weichenthal M, Zimmer L, Livingstone E, Becker JC, Lodde G, Sucker A, Griewank K, Horn S, Hadaschik E, Roesch A, Schadendorf D, Engel DR, Ugurel S. Digital Quantification of Tumor PD-L1 Predicts Outcome of PD-1-Based Immune Checkpoint Therapy in Metastatic Melanoma. Front Oncol 2021; 11:741993. [PMID: 34621681 PMCID: PMC8491983 DOI: 10.3389/fonc.2021.741993] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Background PD-1-based immune checkpoint blockade (ICB) is a highly effective therapy in metastatic melanoma. However, 40-60% of patients are primarily resistant, with valid predictive biomarkers currently missing. This study investigated the digitally quantified tumor PD-L1 expression for ICB therapy outcome prediction. Patients and Methods Tumor tissues taken prior to PD-1-based ICB for unresectable metastatic disease were collected within the prospective multicenter Tissue Registry in Melanoma (TRIM). PD-L1 expression (clone 28-8; cut-off=5%) was determined by digital and physician quantification, and correlated with therapy outcome (best overall response, BOR; progression-free survival, PFS; overall survival, OS). Results Tissue samples from 156 patients were analyzed (anti-PD-1, n=115; anti-CTLA-4+anti-PD-1, n=41). Patients with PD-L1-positive tumors showed an improved response compared to patients with PD-L1-negative tumors, by digital (BOR 50.5% versus 32.2%; p=0.026) and physician (BOR 54.2% versus 36.6%; p=0.032) quantification. Tumor PD-L1 positivity was associated with a prolonged PFS and OS by either digital (PFS, 9.9 versus 4.6 months, p=0.021; OS, not reached versus 13.0 months, p=0.001) or physician (PFS, 10.6 versus 5.6 months, p=0.051; OS, not reached versus 15.6 months, p=0.011) quantification. Multivariable Cox regression revealed digital (PFS, HR=0.57, p=0.007; OS, HR=0.44, p=0.001) and physician (OS, HR=0.54, p=0.016) PD-L1 quantification as independent predictors of survival upon PD-1-based ICB. The combination of both methods identified a patient subgroup with particularly favorable therapy outcome (PFS, HR=0.53, p=0.011; OS, HR=0.47, p=0.008). Conclusion Pre-treatment tumor PD-L1 positivity predicted a favorable outcome of PD-1-based ICB in melanoma. Herein, digital quantification was not inferior to physician quantification, and should be further validated for clinical use.
Collapse
Affiliation(s)
- Jan-Malte Placke
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Camille Soun
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Jenny Bottek
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Rudolf Herbst
- Department of Dermatology, Medical Hospital, Erfurt, Germany
| | | | - Jochen Utikal
- Department of Dermatology, Venerology, and Allergology, University Medical Center, Ruprecht-Karls University of Heidelberg, Mannheim, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Claudia Pföhler
- Department of Dermatology, Saarland University Medical School, Homburg, Germany
| | - Jens Ulrich
- Department of Dermatology, Medical Hospital of Quedlinburg, Quedlinburg, Germany
| | - Alexander Kreuter
- Department of Dermatology, Venereology, and Allergology, Helios St. Elisabeth Hospital Oberhausen, University of Witten-Herdecke, Oberhausen, Germany
| | - Christiane Pfeiffer
- Department of Dermatology, Venereology, and Allergology University Ulm, Ulm, Germany
| | - Peter Mohr
- Department of Dermatology, Elbe-Kliniken, Buxtehude, Germany
| | - Ralf Gutzmer
- Skin Cancer Center, Department of Dermatology, Hannover Medical School, Hannover, Germany
| | - Friedegund Meier
- German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Dermatology, University Hospital Dresden, Dresden, Germany
| | - Edgar Dippel
- Hautklinik, Klinikum der Stadt Ludwigshafen am Rhein gGmbH, Ludwigshafen, Germany
| | | | - Lisa Zimmer
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Elisabeth Livingstone
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jürgen C Becker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Translationale Hautkrebsforschung, University Medicine Essen, University of Duisburg-Essen, Essen, Germany
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Antje Sucker
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Klaus Griewank
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Susanne Horn
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Eva Hadaschik
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alexander Roesch
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Consortium of Translational Cancer Research (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Daniel Robert Engel
- Institute of Experimental Immunology and Imaging, Department of Immunodynamics, University Hospital Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
255
|
Jafri S, Yaqub A. Redifferentiation of BRAF V600E-Mutated Radioiodine Refractory Metastatic Papillary Thyroid Cancer After Treatment With Dabrafenib and Trametinib. Cureus 2021; 13:e17488. [PMID: 34595070 PMCID: PMC8465644 DOI: 10.7759/cureus.17488] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2021] [Indexed: 01/01/2023] Open
Abstract
Radioactive iodine-refractory metastatic differentiated thyroid cancer (RAIR) is associated with a poor prognosis. Multikinase inhibitors have demonstrated improvement in progression-free but not overall survival in such patients, but usage is limited by significant adverse effects and the development of resistance. Clinical research has demonstrated improvement in progression-free survival with the combined use of the BRAF/MEK inhibitor in patients with metastatic melanoma and anaplastic thyroid cancer with the BRAFV600E mutation and has shown promise in redifferentiation of BRAF-positive RAIR differentiated thyroid cancer. A 58-year-old woman went to her primary care physician for a growing mass on the left side of her neck. CT imaging noted a 6 x 8 x 6 cm mixed cystic and solid mass and lymphadenopathy. Core biopsy subsequently showed metastatic papillary thyroid cancer (Stage III, PT4a/PN1b), and she underwent a total thyroidectomy with left neck dissection. She then received 204mCi 131I post-total thyroidectomy. Unfortunately, her thyroglobulin continued to increase post-radioactive iodine (RAI) treatment, indicating persistent and/or recurrent thyroid cancer. An RAI-131 whole-body scan on the thyrogen protocol showed no significant RAI uptake. A fluorodeoxyglucose (FDG)-positron emission tomography (PET) CT scan was then performed, which showed recurrent metastatic disease with hypermetabolism noted in the left thyroid bed and FDG-avid bilateral cervical lymph nodes and pulmonary nodules. Given these findings, her cancer was classified as radioactive iodine refractory (RAIR). Molecular testing indicated the BRAFV600E mutation. After a discussion with the patient, it was decided to initiate therapy with a BRAF inhibitor (dabrafenib 150 mg twice a day) and MEK inhibitor (trametinib 2 mg once a day) in an attempt to redifferentiate RAIR. Repeat RAI-131 thyrogen whole body scan one month after initiation of therapy demonstrated left level 2 cervical lymphadenopathy radioiodine uptake. The patient subsequently received 216 mCi 131I treatment given evidence of redifferentiation. Her post-treatment scan indicated additional uptake in a left lower lobe pulmonary nodule as well as a left paratracheal mass indicating successful RAI-131 uptake by metastases. Her thyroglobulin level, six months post-RAI, decreased to 4.0 indicating an encouraging response. Further surveillance, including imaging studies, is planned. This case illustrates the re-differential potential for dabrafenib and trametinib treatment in patients with BRAFV600E-mutated RAIR differentiated thyroid cancer. This therapy has been shown to be successful in small series of patients and could potentially be offered to RAIR patients with the BRAFV600E mutation as an alternative to multikinase treatment given its favorable side-effect profile.
Collapse
Affiliation(s)
- Sabih Jafri
- Internal Medicine, University of Cincinnati, College of Medicine, Cincinnati, USA
| | - Abid Yaqub
- Endocrinology, University of Cincinnati, College of Medicine, Cincinnati, USA
| |
Collapse
|
256
|
Ge Y, Che X, Gao X, Zhao S, Su J. Combination of radiotherapy and targeted therapy for melanoma brain metastases: a systematic review. Melanoma Res 2021; 31:413-420. [PMID: 34406985 DOI: 10.1097/cmr.0000000000000761] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Radiotherapy is a mainstay of efficient treatment of brain metastases from solid tumors. Immunotherapy has improved the survival of metastatic cancer patients across many tumor types. However, targeted therapy is a feasible alternative for patients unable to continue immunotherapy or with poor outcomes of immunotherapy. The combination of radiotherapy and targeted therapy for the treatment of brain metastases has a strong theoretical underpinning, but data on the efficacy and safety of this combination is still limited. A systematic search of PubMed, Embase, Web of Science and the Cochrane library database was conducted. Eleven studies were included for a total of 316 patients. Median OS was about 6.2-17.8 months from radiotherapy. Weighted survival and local control at 1 and 2 years were correlated (50.1 and 17.8%, 90.7 and 14.7% at 1 and 2 year, respectively). Radiotherapy given before or concurrently to targeted therapy provided the best effect on the outcome. For patients with brain metastases from cutaneous melanoma, the addition of concurrent targeted therapy to brain radiotherapy can increase survival and provide long-term control.
Collapse
Affiliation(s)
- Yi Ge
- Department of Dermatology, Xiangya Hospital, Central South University
- Hunan Engineering Research Center of Skin Health and Disease
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
| | - Xuanlin Che
- Department of Dermatology, Xiangya Hospital, Central South University
- Hunan Engineering Research Center of Skin Health and Disease
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
| | - Xin Gao
- Department of Dermatology, Xiangya Hospital, Central South University
- Hunan Engineering Research Center of Skin Health and Disease
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
| | - Shuang Zhao
- Department of Dermatology, Xiangya Hospital, Central South University
- Hunan Engineering Research Center of Skin Health and Disease
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University
- Hunan Engineering Research Center of Skin Health and Disease
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
| |
Collapse
|
257
|
Gong X, Fan L, Wang P. MEK inhibition by trametinib overcomes chemoresistance in preclinical nasopharyngeal carcinoma models. Anticancer Drugs 2021; 32:978-985. [PMID: 34282746 DOI: 10.1097/cad.0000000000001092] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The development of chemoresistance is the major cause of treatment failure in nasopharyngeal carcinoma (NPC). Although 'paradoxical' activation of extracellular signal-regulated kinase (ERK) has been shown to contribute resistance to anticancer treatment, the role of ERK in NPC chemoresistance has not been yet revealed. In this work, we report that trametinib, a clinically available mitogen-activated protein kinase inhibitor for melanoma treatment, overcomes NPC chemoresistance via suppressing ERK activation induced by chemotherapy. We first showed that trametinib at nanomolar concentrations was active against NPC cells and acted synergistically with cisplatin. Trametinib remarkably decreased phosphorylation of ERK and its downstream effector in NPC cells. We next showed that cisplatin treatment stimulates ERK signaling, and furthermore that this can be abolished by trametinib. We finally generated cisplatin-resistant NPC models and demonstrated that trametinib was effective in inhibiting cisplatin-resistant NPC growth, colony formation and survival via suppressing ERK signaling in vitro and in vivo. Our work demonstrates the potential of trametinib in overcoming chemoresistance in preclinical NPC models and provides evidence of initializing clinical trials of using trametinib for NPC treatment.
Collapse
Affiliation(s)
| | - Lei Fan
- Department of Urologic Surgery
| | - Pu Wang
- Department of Neurology, Xiangyang No. 1 People's Hospital, Affiliated Hospital of Hubei University of Medicine, Xiangyang, Hubei, People's Republic of China
| |
Collapse
|
258
|
Nassar KW, Hintzsche JD, Bagby SM, Espinoza V, Langouët-Astrié C, Amato CM, Chimed TS, Fujita M, Robinson W, Tan AC, Schweppe RE. Targeting CDK4/6 Represents a Therapeutic Vulnerability in Acquired BRAF/MEK Inhibitor-Resistant Melanoma. Mol Cancer Ther 2021; 20:2049-2060. [PMID: 34376578 PMCID: PMC9768695 DOI: 10.1158/1535-7163.mct-20-1126] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 06/18/2021] [Accepted: 07/23/2021] [Indexed: 12/24/2022]
Abstract
There is a clear need to identify targetable drivers of resistance and potential biomarkers for salvage therapy for patients with melanoma refractory to the combination of BRAF and MEK inhibition. In this study, we performed whole-exome sequencing on BRAF-V600E-mutant melanoma patient tumors refractory to the combination of BRAF/MEK inhibition and identified acquired oncogenic mutations in NRAS and loss of the tumor suppressor gene CDKN2A We hypothesized the acquired resistance mechanisms to BRAF/MEK inhibition were reactivation of the MAPK pathway and activation of the cell-cycle pathway, which can both be targeted pharmacologically with the combination of a MEK inhibitor (trametinib) and a CDK4/6 inhibitor (palbociclib). In vivo, we found that combination of CDK4/6 and MEK inhibition significantly decreased tumor growth in two BRAF/MEK inhibitor-resistant patient-derived xenograft models. In vitro, we observed that the combination of CDK4/6 and MEK inhibition resulted in synergy and significantly reduced cellular growth, promoted cell-cycle arrest, and effectively inhibited downstream signaling of MAPK and cell-cycle pathways in BRAF inhibitor-resistant cell lines. Knockdown of CDKN2A in BRAF inhibitor-resistant cells increased sensitivity to CDK4/6 inhibition alone and in combination with MEK inhibition. A key implication of our study is that the combination of CDK4/6 and MEK inhibitors overcomes acquired resistance to BRAF/MEK inhibitors, and loss of CDKN2A may represent a biomarker of response to the combination. Inhibition of the cell-cycle and MAPK pathway represents a promising strategy for patients with metastatic melanoma who are refractory to BRAF/MEK inhibitor therapy.
Collapse
Affiliation(s)
- Kelsey W Nassar
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Jennifer D Hintzsche
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Stacey M Bagby
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Veronica Espinoza
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Christophe Langouët-Astrié
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Carol M Amato
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Tugs-Saikhan Chimed
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Mayumi Fujita
- Department of Dermatology, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - William Robinson
- Division of Medical Oncology, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Aik Choon Tan
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center, Tampa, Florida.
| | - Rebecca E Schweppe
- Division of Endocrinology, Metabolism and Diabetes, Department of Medicine, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- University of Colorado Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
259
|
Sinniah RS, Shapses MS, Ahmed MU, Babiker H, Chandana SR. Novel biomarkers for cholangiocarcinoma: how can it enhance diagnosis, prognostication, and investigational drugs? Part-1. Expert Opin Investig Drugs 2021; 30:1047-1056. [PMID: 34579607 DOI: 10.1080/13543784.2021.1985461] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION The development of novel biomarkers for cancer has exploded over the last decade with advances in novel technologies. Cholangiocarcinoma (CCA), a cancer of the bile ducts, has a dearth of strong disease and pathophysiology biomarkers, making early detection and prognostication a difficult task. AREAS COVERED In this comprehensive review, we discuss the spectrum of biomarkers for CCA diagnosis and prognostication. We elaborate on novel biomarker discovery through a comprehensive multi-omics approach. We also cover, how certain biomarkers may also serve as unique and potent targets for therapeutic development. EXPERT OPINION Despite the relatively poor diagnostic and prognostic performance of existing biomarkers for CCA, there is a vast range of novel biomarkers with exquisite diagnostic and prognostic performance for CCA in the pipeline. Moreover, these biomarkers may serve as potential targets for precision medicine. Existing strategies to target unique biomolecular classes are discussed, within the context of an overall 'omics' focused profiling strategy. Omics profiling will simultaneously allow for enhanced biomarker development and identification of unique subtypes of cholangiocarcinoma and how they are influenced by an individual's unique context. In this manner, patient management strategy and clinical trial design can be optimized to the individual.
Collapse
Affiliation(s)
- Ranu S Sinniah
- College of Human Medicine, Michigan State University, Grand Rapids, MI, USA
| | - Mark S Shapses
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Hani Babiker
- Department of Medicine, Division of Hematology-Oncology, Mayo Clinic, Jacksonville, Florida, USA
| | - Sreenivasa R Chandana
- Phase I Program, Start Midwest, Grand Rapids, MI, USA.,Cancer and Hematology Centers of Western Michigan, Grand Rapids, MI, USA.,Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
260
|
Forschner A, Loquai C, Meiss F, Huening S, Pawlowski J, Bradfisch F, Lehr S, Nashan D. Gibt es eine Überbehandlung von Melanompatienten am Ende ihres Lebens? Ergebnisse einer multizentrischen Studie an 193 Melanompatienten. J Dtsch Dermatol Ges 2021; 19:1297-1306. [PMID: 34541790 DOI: 10.1111/ddg.14501_g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/23/2021] [Indexed: 11/30/2022]
Affiliation(s)
| | | | - Frank Meiss
- Abteilung für Dermatologie und Venerologie, Medizinisches Zentrum - Universität Freiburg, Medizinische Fakultät, Universität Freiburg
| | | | - Johannes Pawlowski
- Abteilung für Dermatologie, Universitätsklinikum Tübingen.,Klinik für Dermatologie, Universitätsklinikum Mainz
| | | | - Saskia Lehr
- Abteilung für Dermatologie und Venerologie, Medizinisches Zentrum - Universität Freiburg, Medizinische Fakultät, Universität Freiburg
| | | |
Collapse
|
261
|
Katsandris A, Ziogas DC, Kontouri M, Staikoglou S, Gogas H. Atezolizumab plus vemurafenib and cobimetinib for the treatment of BRAF V600-mutant advanced melanoma: from an hypothetic triplet to an approved regimen. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1976637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Aikaterini Katsandris
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Dimitrios C. Ziogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Maria Kontouri
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Stavroula Staikoglou
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| | - Helen Gogas
- First Department of Medicine, National and Kapodistrian University of Athens, School of Medicine, Laiko General Hospital, Athens, Greece
| |
Collapse
|
262
|
Wolfe AR, Chablani P, Siedow MR, Miller ED, Walston S, Kendra KL, Wuthrick E, Williams TM. BRAF mutation correlates with worse local-regional control following radiation therapy in patients with stage III melanoma. Radiat Oncol 2021; 16:181. [PMID: 34537078 PMCID: PMC8449455 DOI: 10.1186/s13014-021-01903-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 08/30/2021] [Indexed: 11/26/2022] Open
Abstract
Background In patients with stage III melanoma, the use of adjuvant radiation therapy (RT) after lymph node dissection (LND) may be currently considered in selected high-risk patients to improve tumor control. Melanomas harbor BRAF mutations (BRAF+) in 40–50% of cases, the majority of which are on the V600E residue. This study sought to compare the clinical outcomes after RT between patients with BRAF+ and BRAF− melanoma. Methods This was a retrospective review of 105 Stage III melanoma patients treated at our institution with LND followed by adjuvant RT from 2006 to 2019. BRAF mutational status was determined on the primary skin or nodal tissue samples from all patients. We compared characteristics of the BRAF+ and BRAF− groups using Fisher’s exact test and Wilcoxon rank sum test and performed univariate and multivariate analysis using Kaplan–Meier estimates, log-rank tests, and Cox proportional hazards modeling with the clinical outcomes of local–regional lymph node control, distant metastasis-free survival (DMFS), recurrence-free survival (RFS), and overall survival (OS). Results Fifty-three (50%) patients harbored a BRAF mutation (92%, pV600E). BRAF+ patients were younger and had primary tumors more commonly found in the trunk vs head and neck compared to BRAF- patients (p < 0.05). The 5 year local–regional control in the BRAF + patients was 60% compared to 81% in the BRAF- patients (HR 4.5, 95% CI 1.3–15.5, p = 0.02). There were no significant differences in 5-year DMFS, RFS, and OS rates between the two BRAF patient groups. The presence of 4 or more positive LNs remained a significant prognostic factor for local–regional lymph node control, RFS, and OS in multivariate analysis. Conclusions Stage III melanoma patients with BRAF mutation treated with adjuvant RT had > 4 times increased risk of local recurrence or regional lymph node recurrence. These results could be useful for adjuvant RT consideration in lymph node positive melanoma patients and supports other data that BRAF mutation confers radiation resistance.
Collapse
Affiliation(s)
- Adam R Wolfe
- Department of Radiation Oncology, The University of Arkansas for Medical Sciences, The Winthrop P. Rockefeller Cancer Institute, Little Rock, AR, USA
| | - Priyanka Chablani
- Division of Hematology-Oncology, Department of Internal Medicine, University of Chicago, Chicago, IL, USA
| | - Michael R Siedow
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Eric D Miller
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Steve Walston
- Department of Radiation Oncology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Kari L Kendra
- Division of Hematology-Oncology, Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Evan Wuthrick
- Department of Radiation Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Terence M Williams
- Department of Radiation Oncology, City of Hope National Medical Center, 1500 E. Duarte Road, Duarte, CA, 91010, USA.
| |
Collapse
|
263
|
Dalmasso C, Pagès C, Chaltiel L, Sibaud V, Moyal E, Chira C, Sol JC, Latorzeff I, Meyer N, Modesto A. Intracranial Treatment in Melanoma Patients with Brain Metastasis Is Associated with Improved Survival in the Era of Immunotherapy and Anti-BRAF Therapy. Cancers (Basel) 2021; 13:cancers13174493. [PMID: 34503304 PMCID: PMC8430519 DOI: 10.3390/cancers13174493] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 08/19/2021] [Accepted: 09/01/2021] [Indexed: 11/16/2022] Open
Abstract
Metastatic melanoma patients are at high risk of brain metastases (BM). Although intracranial control is a prognostic factor for survival, impact of local (intracranial) treatment (LT), surgery and/or radiotherapy (stereotactic or whole brain) in the era of novel therapies remains unknown. We evaluated BM incidence in melanoma patients receiving immune checkpoint inhibitors (ICI) or anti-BRAF therapy and identified prognostic factors for overall survival (OS). Clinical data and treatment patterns were retrospectively collected from all patients treated for newly diagnosed locally advanced or metastatic melanoma between May 2014 and December 2017 with available BRAF mutation status and receiving systemic therapy. Prognostic factors for OS were analyzed with univariable and multivariable survival analyses. BMs occurred in 106 of 250 eligible patients (42.4%), 64 of whom received LT. Median OS in patients with BM was 7.8 months (95% CI [5.4-10.4]). In multivariable analyses, LT was significantly correlated with improved OS (HR 0.21, p < 0.01). Median OS was 17.3 months (95% CI [8.3-22.3]) versus 3.6 months (95% CI [1.4-4.8]) in patients with or without LT. LT correlates with improved OS in melanoma patients with BM in the era of ICI and anti-BRAF therapy. The use of LT should be addressed at diagnosis of BM while introducing systemic treatment.
Collapse
Affiliation(s)
- Céline Dalmasso
- Radiation Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France; (C.D.); (E.M.); (C.C.)
| | - Cécile Pagès
- Dermato-Oncology Department, Institut Universitaire du Cancer, CEDEX 9, 31059 Toulouse, France; (C.P.); (V.S.); (N.M.)
| | - Léonor Chaltiel
- Biostatistics Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France;
| | - Vincent Sibaud
- Dermato-Oncology Department, Institut Universitaire du Cancer, CEDEX 9, 31059 Toulouse, France; (C.P.); (V.S.); (N.M.)
| | - Elisabeth Moyal
- Radiation Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France; (C.D.); (E.M.); (C.C.)
- Gamma Knife Unit, CHU–Toulouse-Purpan, 31000 Toulouse, France; (J.C.S.); (I.L.)
| | - Ciprian Chira
- Radiation Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France; (C.D.); (E.M.); (C.C.)
| | - Jean Christophe Sol
- Gamma Knife Unit, CHU–Toulouse-Purpan, 31000 Toulouse, France; (J.C.S.); (I.L.)
- Neuro-Surgery Department, CHU de Toulouse–Purpan, 31000 Toulouse, France
| | - Igor Latorzeff
- Gamma Knife Unit, CHU–Toulouse-Purpan, 31000 Toulouse, France; (J.C.S.); (I.L.)
- Radiation Oncology Department, Oncorad, Clinique Pasteur, 31000 Toulouse, France
| | - Nicolas Meyer
- Dermato-Oncology Department, Institut Universitaire du Cancer, CEDEX 9, 31059 Toulouse, France; (C.P.); (V.S.); (N.M.)
- Dermatology Department, CHU de Toulouse, Hôpital Larrey, CEDEX 9, 31059 Toulouse, France
| | - Anouchka Modesto
- Radiation Oncology Department, Institut Claudius Regaud, Institut Universitaire du Cancer de Toulouse, CEDEX 9, 31059 Toulouse, France; (C.D.); (E.M.); (C.C.)
- Correspondence:
| |
Collapse
|
264
|
Teixido C, Castillo P, Martinez-Vila C, Arance A, Alos L. Molecular Markers and Targets in Melanoma. Cells 2021; 10:2320. [PMID: 34571969 PMCID: PMC8469294 DOI: 10.3390/cells10092320] [Citation(s) in RCA: 117] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/28/2021] [Accepted: 09/01/2021] [Indexed: 12/26/2022] Open
Abstract
Melanoma develops as a result of several genetic alterations, with UV radiation often acting as a mutagenic risk factor. Deep knowledge of the molecular signaling pathways of different types of melanoma allows better characterization and provides tools for the development of therapies based on the intervention of signals promoted by these cascades. The latest World Health Organization classification acknowledged the specific genetic drivers leading to melanoma and classifies melanocytic lesions into nine distinct categories according to the associate cumulative sun damage (CSD), which correlates with the molecular alterations of tumors. The largest groups are melanomas associated with low-CSD or superficial spreading melanomas, characterized by frequent presentation of the BRAFV600 mutation. High-CSD melanomas include lentigo maligna type and desmoplastic melanomas, which often have a high mutation burden and can harbor NRAS, BRAFnon-V600E, or NF1 mutations. Non-CSD-associated melanomas encompass acral and mucosal melanomas that usually do not show BRAF, NRAS, or NF1 mutations (triple wild-type), but in a subset may have KIT or SF3B1 mutations. To improve survival, these driver alterations can be treated with targeted therapy achieving significant antitumor activity. In recent years, relevant improvement in the prognosis and survival of patients with melanoma has been achieved, since the introduction of BRAF/MEK tyrosine kinase inhibitors and immune checkpoint inhibitors. In this review, we describe the current knowledge of molecular pathways and discuss current and potential therapeutic targets in melanoma, focusing on their clinical relevance of development.
Collapse
Affiliation(s)
- Cristina Teixido
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (P.C.); (L.A.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain;
| | - Paola Castillo
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (P.C.); (L.A.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain;
| | - Clara Martinez-Vila
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain;
- Department of Medical Oncology, Althaia Xarxa Assistencial Universitària de Manresa, Dr. Joan Soler, 1–3, 08243 Manresa, Spain
| | - Ana Arance
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain;
- Department of Medical Oncology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain;
| | - Llucia Alos
- Department of Pathology, Hospital Clínic of Barcelona, University of Barcelona, Villarroel 170, 08036 Barcelona, Spain; (P.C.); (L.A.)
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Rosselló 149, 08036 Barcelona, Spain;
| |
Collapse
|
265
|
Quaglino P, Fava P, Tonella L, Rubatto M, Ribero S, Fierro MT. Treatment of Advanced Metastatic Melanoma. Dermatol Pract Concept 2021; 11:e2021164S. [PMID: 34447613 DOI: 10.5826/dpc.11s1a164s] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/10/2021] [Indexed: 12/13/2022] Open
Abstract
The introduction in clinical practice of new drug compounds both targeted therapies anti-BRAF and checkpoint inhibitors have largely improved our potential to manage advanced metastatic melanoma patients. This has led to a significant improvement in terms of response rates and particularly in the overall survival (OS). The long-term results of trials with follow-up data of patients treated with targeted or immunotherapies reported median OS rates around 24 months, with 5-year survival rates around 35-40%. As to the drugs currently available and reimbursed by the Italian National Health System, 3 combinations of anti-BRAF/anti-MEK inhibitors are available (dabrafenib/trametinib, vemurafenib/cobimetinib and the most recently introduced encorafenib/binimetinib). As for checkpoint inhibitors, first line immunotherapy is represented by anti-PD1 blockers (nivolumab and pembrolizumab), whilst the anti-CTLA-4 ipilimumab can be used as second line immunotherapy. The decision-making factors that define the best treatment approach in stage IV patients with metastatic melanoma include the mutation pattern, performance status, high/low tumor load, brain metastases, progression pattern (low/fast), and availability of clinical trials. This review will analyze the current therapeutic tools adopted for the treatment of metastatic melanoma patients. It will then focus on the latest results obtained by novel treatments (checkpoint inhibitors and targeted therapies) which can be used in the clinical daily practice.
Collapse
Affiliation(s)
- Pietro Quaglino
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Paolo Fava
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Luca Tonella
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Marco Rubatto
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | - Simone Ribero
- Dermatologic Clinic, University of Turin Medical School, Turin, Italy
| | | |
Collapse
|
266
|
Laxmikeshav K, Kumari P, Shankaraiah N. Expedition of sulfur-containing heterocyclic derivatives as cytotoxic agents in medicinal chemistry: A decade update. Med Res Rev 2021; 42:513-575. [PMID: 34453452 DOI: 10.1002/med.21852] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 04/20/2021] [Accepted: 08/17/2021] [Indexed: 12/13/2022]
Abstract
This review article proposes a comprehensive report of the design strategies engaged in the development of various sulfur-bearing cytotoxic agents. The outcomes of various studies depict that the sulfur heterocyclic framework is a fundamental structure in diverse synthetic analogs representing a myriad scope of therapeutic activities. A number of five-, six- and seven-membered sulfur-containing heterocyclic scaffolds, such as thiazoles, thiadiazoles, thiazolidinediones, thiophenes, thiopyrans, benzothiazoles, benzothiophenes, thienopyrimidines, simple and modified phenothiazines, and thiazepines have been discussed. The subsequent studies of the derivatives unveiled their cytotoxic effects through multiple mechanisms (viz. inhibition of tyrosine kinases, topoisomerase I and II, tubulin, COX, DNA synthesis, and PI3K/Akt and Raf/MEK/ERK signaling pathways), and several others. Thus, our concise illustration explains the design strategy and anticancer potential of these five- and six-membered sulfur-containing heterocyclic molecules along with a brief outline on seven-membered sulfur heterocycles. The thorough assessment of antiproliferative activities with the reference drug allows a proficient assessment of the structure-activity relationships (SARs) of the diversely synthesized molecules of the series.
Collapse
Affiliation(s)
- Kritika Laxmikeshav
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Pooja Kumari
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| | - Nagula Shankaraiah
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, India
| |
Collapse
|
267
|
Phase 2 Study of Dabrafenib Plus Trametinib in Patients With BRAF V600E-Mutant Metastatic NSCLC: Updated 5-Year Survival Rates and Genomic Analysis. J Thorac Oncol 2021; 17:103-115. [PMID: 34455067 DOI: 10.1016/j.jtho.2021.08.011] [Citation(s) in RCA: 133] [Impact Index Per Article: 33.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 08/08/2021] [Accepted: 08/09/2021] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dabrafenib plus trametinib was found to have robust antitumor activity in patients with BRAF V600E-mutant metastatic NSCLC (mNSCLC). We report updated survival analysis of a phase 2 study (NCT01336634) with a minimum of 5-year follow-up and updated genomic data. METHODS Pretreated (cohort B) and treatment-naive (cohort C) patients with BRAF V600E-mutant mNSCLC received dabrafenib 150 mg twice daily and trametinib 2 mg once daily. The primary end point was investigator-assessed overall response rate per Response Evaluation Criteria in Solid Tumors version 1.1. Secondary end points were duration of response, progression-free survival, overall survival, and safety. RESULTS At data cutoff, for cohorts B (57 patients) and C (36 patients), the median follow-up was 16.6 (range: 0.5-78.5) and 16.3 (range: 0.4-80) months, overall response rate (95% confidence interval [CI]) was 68.4% (54.8-80.1) and 63.9% (46.2-79.2), median progression-free survival (95% CI) was 10.2 (6.9-16.7) and 10.8 (7.0-14.5) months, and median overall survival (95% CI) was 18.2 (14.3-28.6) and 17.3 (12.3-40.2) months, respectively. The 4- and 5-year survival rates were 26% and 19% in pretreated patients and 34% and 22% in treatment-naive patients, respectively. A total of 17 patients (18%) were still alive. The most frequent adverse event was pyrexia (56%). Exploratory genomic analysis indicated that the presence of coexisting genomic alterations might influence clinical outcomes in these patients; however, these results require further investigation. CONCLUSIONS Dabrafenib plus trametinib therapy was found to have substantial and durable clinical benefit, with a manageable safety profile, in patients with BRAF V600E-mutant mNSCLC, regardless of previous treatment.
Collapse
|
268
|
Forschner A, Loquai C, Meiss F, Huening S, Pawlowski J, Bradfisch F, Lehr S, Nashan D. Is there an overtreatment of melanoma patients at the end of their life? Results of a multicenter study on 193 melanoma patients. J Dtsch Dermatol Ges 2021; 19:1297-1305. [PMID: 34357677 DOI: 10.1111/ddg.14501] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND OBJECTIVES There is a lack of data regarding the situation of melanoma patients receiving systemic therapies in their last months of life. PATIENTS AND METHODS All melanoma patients who died in 2016 or 2017 and who had been treated by systemic therapies within the last three months of life were retrospectively analyzed. The study was conducted within the Committee "supportive therapy" of the Work Group Dermatological Oncology (ADO). RESULTS 193 patients from four dermato-oncological centers were included. More than 60 % of the patients had ECOG ≥ 2 and most of them received immune checkpoint inhibitors (ICI) or targeted therapies (TT). 41 patients benefited from the last therapy in terms of radiological and laboratory findings or state of health. Although ECOG was worse in the TT cohort compared to the ICI group, the proportion of patients benefiting from the last therapy with TT was significantly higher and TT therapy could be carried out more often on an outpatient basis. CONCLUSIONS This study indicates that there is a tendency towards an overtreatment at the end of life. Nevertheless, TT might be a reasonable therapeutic option for advanced BRAF mutant melanoma, even in highly palliative situations.
Collapse
Affiliation(s)
- Andrea Forschner
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany
| | - Carmen Loquai
- Department of Dermatology, University Hospital Mainz, Mainz, Germany
| | - Frank Meiss
- Department of Dermatology and Venereology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Svea Huening
- Department of Dermatology, Hospital Dortmund, Dortmund, Germany
| | - Johannes Pawlowski
- Department of Dermatology, University Hospital Tübingen, Tübingen, Germany.,Department of Dermatology, University Hospital Mainz, Mainz, Germany
| | | | - Saskia Lehr
- Department of Dermatology and Venereology, Medical Center - University of Freiburg, University of Freiburg, Freiburg, Germany
| | - Dorotheé Nashan
- Department of Dermatology, Hospital Dortmund, Dortmund, Germany
| |
Collapse
|
269
|
Incidence and characteristics of metastatic intracranial lesions in stage III and IV melanoma: a single institute retrospective analysis. J Neurooncol 2021; 154:197-203. [PMID: 34351544 DOI: 10.1007/s11060-021-03813-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 07/22/2021] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The study aimed to describe the brain metastases (BM) incidence, at diagnosis and follow-up, in patients initially presenting with stage III or IV melanoma and characterize their metastatic brain lesions. We also sought to describe the association of common genetic mutations and immunotherapy with BM development in advanced melanoma. METHODS Using our institution's tumor registry, we identified patients with initial diagnoses of stage III and stage IV melanoma. In this cohort, we obtained BM incidence at diagnosis and follow-up, characterized the metastatic brain lesions and primary tumor's genetic profile. RESULTS During the follow-up period, 22.9% of patients with an initial diagnosis of stage III developed BM. In this cohort, the median time for BM occurrence was 20 months; [95% CI (14-29)]. Likewise, 37.7% of patients with Stage IV melanoma presented with BM at the time of diagnosis, and 22.7% of remaining patients developed BM at follow-up over a median duration of 6 months [95% CI (4-11)]. Therefore, suggesting an overall incidence of 51.9% in stage IV melanoma. Next, we observed that the incidence of BM development during the follow-up period significantly decreased from 2012 to 2017 (p < 0.001). Lastly, we found a significantly higher frequency of mutational BRAF in the primary tumor of patients with BM (68.7% vs. 31.2%; p = 0.02). CONCLUSIONS While the overall incidence of BM remains high, the decreasing incidence of BM over the follow-up period is promising. Similar BM incidence in patients with an initial diagnosis of stage III or stage IV warrants appropriate imaging surveillance regimen for stage III patients.
Collapse
|
270
|
Wada F, Hiramoto N, Yamashita D, Hara S, Furukawa Y, Ishii J, Nagata K, Nannya Y, Ogawa S, Ishikawa T. Dramatic response to encorafenib in a patient with Erdheim-Chester disease harboring the BRAF V600E mutation. Am J Hematol 2021; 96:E295-E298. [PMID: 33971044 DOI: 10.1002/ajh.26232] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 11/06/2022]
Affiliation(s)
- Fumiya Wada
- Department of Hematology Kobe City Medical Center General Hospital Kobe Japan
| | - Nobuhiro Hiramoto
- Department of Hematology Kobe City Medical Center General Hospital Kobe Japan
| | - Daisuke Yamashita
- Department of Diagnostic Pathology Kobe City Medical Center General Hospital Kobe Japan
| | - Shigeo Hara
- Department of Diagnostic Pathology Kobe City Medical Center General Hospital Kobe Japan
| | - Yutaka Furukawa
- Department of Cardiovascular Medicine Kobe City Medical Center General Hospital Kobe Japan
| | - Junko Ishii
- Department of Neurology Kobe City Medical Center General Hospital Kobe Japan
| | - Kazuma Nagata
- Department of Respiratory Medicine Kobe City Medical Center General Hospital Kobe Japan
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology Kyoto University Kyoto Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology Kyoto University Kyoto Japan
| | - Takayuki Ishikawa
- Department of Hematology Kobe City Medical Center General Hospital Kobe Japan
| |
Collapse
|
271
|
Kobelt D, Perez-Hernandez D, Fleuter C, Dahlmann M, Zincke F, Smith J, Migotti R, Popp O, Burock S, Walther W, Dittmar G, Mertins P, Stein U. The newly identified MEK1 tyrosine phosphorylation target MACC1 is druggable by approved MEK1 inhibitors to restrict colorectal cancer metastasis. Oncogene 2021; 40:5286-5301. [PMID: 34247190 PMCID: PMC8390371 DOI: 10.1038/s41388-021-01917-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 06/14/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Cancer metastasis causes >90% of cancer deaths and remains a major treatment challenge. Here we deciphered the impact of tyrosine phosphorylation of MACC1, a causative driver for cancer metastasis, for cancer cell signaling and novel interventions to restrict cancer metastasis. We identified MACC1 as new MEK1 substrate. MEK1 directly phosphorylates MACC1, leading to accelerated and increased ERK1 activation. Mutating in silico predicted hierarchical MACC1 tyrosine phosphorylation sites abrogates MACC1-induced migration, invasion, and MET expression, a transcriptional MACC1 target. Targeting MEK1 by RNAi or clinically applicable MEK1 inhibitors AZD6244 and GSK1120212 reduces MACC1 tyrosine phosphorylation and restricts MACC1-induced metastasis formation in mice. Although MEK1 levels, contrary to MACC1, are not of prognostic relevance for CRC patients, MEK1 expression was found indispensable for MACC1-induced metastasis. This study identifies MACC1 as new MEK1 substrate for tyrosine phosphorylation decisively impacting cell motility, tumor growth, and metastasis. Thus, MAP kinase signaling is not linear leading to ERK activation, but branches at the level of MEK1. This fundamental finding opens new therapeutic options for targeting the MEK1/MACC1 axis as novel vulnerability in patients at high risk for metastasis. This might be extended from CRC to further solid tumor entities.
Collapse
Affiliation(s)
- Dennis Kobelt
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Daniel Perez-Hernandez
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Claudia Fleuter
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Mathias Dahlmann
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Fabian Zincke
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Janice Smith
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Rebekka Migotti
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Oliver Popp
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Susen Burock
- Charité Comprehensive Cancer Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Wolfgang Walther
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Gunnar Dittmar
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Proteome and Genome Research Laboratory, Luxembourg Institute of Health, Strassen, Luxembourg
| | - Philipp Mertins
- Mass Spectrometry Core Unit, Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Ulrike Stein
- Translational Oncology of Solid Tumors, Experimental and Clinical Research Center, Charité-Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| |
Collapse
|
272
|
Salama Y, Jaradat N, Hattori K, Heissig B. Aloysia Citrodora Essential Oil Inhibits Melanoma Cell Growth and Migration by Targeting HB-EGF-EGFR Signaling. Int J Mol Sci 2021; 22:ijms22158151. [PMID: 34360915 PMCID: PMC8347434 DOI: 10.3390/ijms22158151] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 07/22/2021] [Accepted: 07/24/2021] [Indexed: 12/25/2022] Open
Abstract
Patients diagnosed with melanoma have a poor prognosis due to regional invasion and metastases. The receptor tyrosine kinase epidermal growth factor receptor (EGFR) is found in a subtype of melanoma with a poor prognosis and contributes to drug resistance. Aloysia citrodora essential oil (ALOC-EO) possesses an antitumor effect. Understanding signaling pathways that contribute to the antitumor of ALOC-EO is important to identify novel tumor types that can be targeted by ALOC-EO. Here, we investigated the effects of ALOC-EO on melanoma growth and tumor cell migration. ALOC-EO blocked melanoma growth in vitro and impaired primary tumor cell growth in vivo. Mechanistically, ALOC-EO blocked heparin-binding-epidermal growth factor (HB-EGF)-induced EGFR signaling and suppressed ERK1/2 phosphorylation. Myelosuppressive drugs upregulated HB-EGF and EGFR expression in melanoma cells. Cotreatment of myelosuppressive drugs with ALOC-EO improved the antitumor activity and inhibited the expression of matrix metalloproteinase-7 and -9 and a disintegrin and metalloproteinase domain-containing protein9. In summary, our study demonstrates that ALOC-EO blocks EGFR and ERK1/2 signaling, with preclinical efficacy as a monotherapy or in combination with myelosuppressive drugs in melanoma.
Collapse
Affiliation(s)
- Yousef Salama
- An-Najah Center for Cancer and Stem Cell Research, Faculty of Medicine and Health Sciences, An-Najah National University, P.O. Box 7, Nablus 99900800, Palestine
- Correspondence: (Y.S.); (B.H.)
| | - Nidal Jaradat
- Department of Pharmacy, Faculty of Medicine and Health Sciences, An-Najah National University, Nablus 00970, Palestine;
| | - Koichi Hattori
- Center for Genomic & Regenerative Medicine, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan;
| | - Beate Heissig
- Department of Immunological Diagnosis, School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-Ku, Tokyo 113-8421, Japan
- Correspondence: (Y.S.); (B.H.)
| |
Collapse
|
273
|
Xu QQ, Li QJ, Huang CL, Cai MY, Zhang MF, Yin SH, Lu LX, Chen L. Prognostic Value of an Immunohistochemical Signature in Patients With Head and Neck Mucosal Melanoma. Front Immunol 2021; 12:708293. [PMID: 34394109 PMCID: PMC8358394 DOI: 10.3389/fimmu.2021.708293] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Purpose We aimed to develop a prognostic immunohistochemistry (IHC) signature for patients with head and neck mucosal melanoma (MMHN). Methods In total, 190 patients with nonmetastatic MMHN with complete clinical and pathological data before treatment were included in our retrospective study. Results We extracted five IHC markers associated with overall survival (OS) and then constructed a signature in the training set (n=116) with the least absolute shrinkage and selection operator (LASSO) regression model. The validation set (n=74) was further built to confirm the prognostic significance of this classifier. We then divided patients into high- and low-risk groups according to the IHC score. In the training set, the 5-year OS rate was 22.0% (95% confidence interval [CI]: 11.2%- 43.2%) for the high-risk group and 54.1% (95% CI: 41.8%-69.9%) for the low-risk group (P<0.001), and in the validation set, the 5-year OS rate was 38.1% (95% CI: 17.9%-81.1%) for the high-risk group and 43.1% (95% CI: 30.0%-61.9%) for the low-risk group (P=0.26). Multivariable analysis revealed that IHC score, T stage, and primary tumor site were independent variables for predicting OS (all P<0.05). We developed a nomogram incorporating clinicopathological risk factors (primary site and T stage) and the IHC score to predict 3-, 5-, and 10-year OS. Conclusions A nomogram was generated and confirmed to be of clinical value. Our IHC classifier integrating five IHC markers could help clinicians make decisions and determine optimal treatments for patients with MMHN.
Collapse
Affiliation(s)
- Qing-Qing Xu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Qing-Jie Li
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Cheng-Long Huang
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Mu-Yan Cai
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Mei-Fang Zhang
- Department of Pathology, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Shao-Han Yin
- Imaging Diagnosis and Interventional Center, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in Southern China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Li-Xia Lu
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| | - Lei Chen
- Department of Radiation Oncology, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Key Laboratory of Nasopharyngeal Carcinoma Diagnosis and Therapy, Guangzhou, China
| |
Collapse
|
274
|
Loss of skeletal muscle area and fat-free mass during dabrafenib/trametinib and vemurafenib/cobimetinib treatments in patients with BRAF-mutant metastatic malignant melanoma. Melanoma Res 2021; 30:477-483. [PMID: 32898388 DOI: 10.1097/cmr.0000000000000678] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This study aimed to assess whether dabrafenib/trametinib and vemurafenib/cobimetinib treatments are associated with a change in skeletal muscle area (SMA) and total fat-free mass (FFM) assessed by computed tomography (CT), and to compare the efficacy and safety profile of these treatments in patients with metastatic melanoma. Thirty-one patients treated with B-Raf proto-oncogene, serine/threonine kinase/MAPK extracellular receptor kinase inhibitors were included between 2016 and 2019. Eighteen patients received dabrafenib/trametinib and remaining patients received vemurafenib/cobimetinib. CT scans were performed at baseline and at 4-6 months of follow-up to measure cross-sectional areas of SMA. FFM and skeletal muscle index (SMI) values were calculated. Of the patients, including 18 treated with dabrafenib/trametinib (58.1%) and 13 with vemurafenib/cobimetinib (41.9%); 58.1% were male, 41.9% were female and median age was 52 years. A significant decrease in SMA was observed after dabrafenib/trametinib and vemurafenib/cobimetinib treatments (P = 0.003 and P = 0.002, respectively). A significant decrease in FFM values was observed after dabrafenib/trametinib and vemurafenib/cobimetinib treatments (P = 0.003 and P = 0.002, respectively). Dose-limiting toxicity (DLT) was observed in 35.9% of the patients with sarcopenia. No significant difference was seen between the dabrafenib/trametinib and vemurafenib/cobimetinib groups in median progression-free survival (PFS) (11.9 vs. 7.3 months, respectively, P = 0.28) and in median overall survival (OS) (25.46 vs. 13.7 months, respectively, P = 0.41). Baseline sarcopenia was not significantly associated with PFS or OS (P = 0.172 and P = 0.326, respectively). We found a significant decrease in SMI values determined at 4-6 months compared to the values before treatment both in dabrafenib/trametinib and vemurafenib/cobimetinib groups. DLT was similar with both treatments. Baseline sarcopenia was not significantly associated with PFS or OS.
Collapse
|
275
|
Lassen UN, Makaroff LE, Stenzinger A, Italiano A, Vassal G, Garcia-Foncillas J, Avouac B. Precision oncology: a clinical and patient perspective. Future Oncol 2021; 17:3995-4009. [PMID: 34278817 DOI: 10.2217/fon-2021-0688] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Molecular characterization of tumors has shifted cancer treatment strategies away from nonspecific cytotoxic treatment of histology-specific tumors toward targeting of actionable mutations that can be found across multiple cancer types. The development of high-throughput technologies such as next-generation sequencing, combined with decision support applications and availability of patient databases, has provided tools that optimize disease management. Precision oncology has proven success in improving outcomes and quality of life, as well as identifying and overcoming mechanisms of drug resistance and relapse. Addressing challenges that impede its use will improve matching of therapies to patients. Here we review the current status of precision oncology medicine, emphasizing its impact on patients - what they understand about precision oncology medicine and their hopes for the future.
Collapse
Affiliation(s)
| | - Lydia E Makaroff
- Fight Bladder Cancer, Oxfordshire, OX39 4DJ, UK.,World Bladder Cancer Patient Coalition, Brussels, Belgium
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, 69120, Germany
| | | | - Gilles Vassal
- Gustave Roussy Comprehensive Cancer Center, & Unversity Paris-Saclay, Villejuif, 94805, France
| | - Jesus Garcia-Foncillas
- University Cancer Institute & The Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, 28033, Spain
| | | |
Collapse
|
276
|
Emile JF, Cohen-Aubart F, Collin M, Fraitag S, Idbaih A, Abdel-Wahab O, Rollins BJ, Donadieu J, Haroche J. Histiocytosis. Lancet 2021; 398:157-170. [PMID: 33901419 PMCID: PMC9364113 DOI: 10.1016/s0140-6736(21)00311-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 01/25/2021] [Accepted: 02/01/2021] [Indexed: 02/06/2023]
Abstract
Histiocytoses constitute a heterogeneous group of rare disorders, characterised by infiltration of almost any organ by myeloid cells with diverse macrophage or dendritic cell phenotypes. Histiocytoses can start at any age. Diagnosis is based on histology in combination with appropriate clinical and radiological findings. The low incidence and broad spectrum of clinical manifestations often leads to diagnostic delay, especially for adults. In most cases, biopsy specimens infiltrated by histiocytes have somatic mutations in genes activating the MAP kinase cell-signalling pathway. These mutations might also be present in blood cells and haematopoietic progenitors of patients with multisystem disease. A comprehensive range of investigations and molecular typing are essential to accurately predict prognosis, which can vary from spontaneous resolution to life-threatening disseminated disease. Targeted therapies with BRAF or MEK inhibitors have revolutionised salvage treatment. However, the type and duration of treatment are still debated, and the prevention of neurological sequelae remains a crucial issue.
Collapse
Affiliation(s)
- Jean-François Emile
- EA4340 BECCOH, Université de Versailles SQY, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France.
| | - Fleur Cohen-Aubart
- Internal Medicine Department 2, French National Referral Center for Rare Systemic Diseases and Histiocytoses, Pitié-Salpêtrière Hospital, AP-HP and Sorbonne Université, Paris, France
| | - Matthew Collin
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Sylvie Fraitag
- Pathology Department, Necker-Enfants Malades Hospital, AP-HP, Paris, France
| | - Ahmed Idbaih
- UMR S 1127, CNRS/Inserm, Institut du Cerveau et de la Moelle Épinière, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, AP-HP and Sorbonne Université, Paris, France
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Barrett J Rollins
- Department of Medical Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jean Donadieu
- EA4340 BECCOH, Université de Versailles SQY, Service de Pathologie, Hôpital Ambroise Paré, AP-HP, Boulogne, France; Service d'Hématologie Oncologie Pédiatrique, Centre de Référence des Histiocytoses, Hôpital Armand-Trousseau, AP-HP, Paris, France
| | - Julien Haroche
- Internal Medicine Department 2, French National Referral Center for Rare Systemic Diseases and Histiocytoses, Pitié-Salpêtrière Hospital, AP-HP and Sorbonne Université, Paris, France
| |
Collapse
|
277
|
Efficacy, safety and factors associated with disease progression in patients with unresectable (stage III) or distant metastatic (stage IV) BRAF V600-mutant melanoma: An open label, non-randomized, phase IIIb study of trametinib in combination with dabrafenib. Eur J Cancer 2021; 154:57-65. [PMID: 34243078 DOI: 10.1016/j.ejca.2021.05.031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/13/2021] [Accepted: 05/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND BRAF and MEK inhibitors combination, including dabrafenib (D) and trametinib (T) have transformed the treatment of BRAF V600-mutant advanced melanoma patients, including patients with brain metastasis (BM). In a large phase IIIb, single-arm, open-label, multicenter French study, we assessed safety, response to treatment, progression-free survival (PFS) and factors associated with progression, and stratified the population into risk groups. METHODS Patients with unresectable, advanced, BRAF V600-mutant melanoma were included, including those with the presence of BM, Eastern Cooperative Oncology Group Performance Status (ECOG PS) ≤2, elevated lactate dehydrogenase (LDH) or previous melanoma treatments. Responses were determined locally, without central review. PFS was estimated using the Kaplan-Meier analysis and modelled with multivariate Cox model. Risk subgroups were identified using a regression tree analysis. RESULTS Between March 2015 and November 2016, 856 patients received at least one D + T dose. Overall, 92% had stage IV melanoma, 38% ECOG PS ≥1, 32% BM and 37.5% elevated LDH. Median PFS was 8.02 months (95% confidence interval [CI] 7.33-8.77). Significant factors associated with lower PFS were ECOG PS ≥1, elevated LDH, ≥3 metastatic sites and presence of BM. Patients with <3 metastatic sites, ECOG = 0 and no BM had the highest probability of PFS at 6 months (83%, 95% CI 76-87) and 12 months (56%, 95% CI 47-64), respectively. CONCLUSIONS This is the largest prospective study in advanced BRAF V600-mutant melanoma patients treated with D + T, conducted in conditions close to 'real-world practice'. We confirm previous findings that LDH, ECOG PS and ≥3 metastatic sites are associated with shorter PFS, but the real-world setting introduces BM as a major prognostic factor.
Collapse
|
278
|
Borch TH, Harbst K, Rana AH, Andersen R, Martinenaite E, Kongsted P, Pedersen M, Nielsen M, Kjeldsen JW, Kverneland AH, Lauss M, Hölmich LR, Hendel H, Met Ö, Jönsson G, Donia M, Marie Svane I. Clinical efficacy of T-cell therapy after short-term BRAF-inhibitor priming in patients with checkpoint inhibitor-resistant metastatic melanoma. J Immunother Cancer 2021; 9:jitc-2021-002703. [PMID: 34210820 PMCID: PMC8252872 DOI: 10.1136/jitc-2021-002703] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/28/2021] [Indexed: 11/04/2022] Open
Abstract
PURPOSE Despite impressive response rates following adoptive transfer of autologous tumor-infiltrating lymphocytes (TILs) in patients with metastatic melanoma, improvement is needed to increase the efficacy and broaden the applicability of this treatment. We evaluated the use of vemurafenib, a small-molecule BRAF inhibitor with immunomodulatory properties, as priming before TIL harvest and adoptive T cell therapy in a phase I/II clinical trial. METHODS 12 patients were treated with vemurafenib for 7 days before tumor excision and during the following weeks until TIL infusion. TILs were grown from tumor fragments, expanded in vitro and reinfused to the patient preceded by a lymphodepleting chemotherapy regimen and followed by interleukin-2 infusion. Extensive immune monitoring, tumor profiling and T cell receptor sequencing were performed. RESULTS No unexpected toxicity was observed, and treatment was well tolerated. Of 12 patients, 1 achieved a complete response, 8 achieved partial response and 3 achieved stable disease. A PR and the CR are ongoing for 23 and 43 months, respectively. In vitro anti-tumor reactivity was found in TILs from 10 patients, including all patients achieving objective response. Serum and tumor biomarker analyses indicate that baseline cytokine levels and the number of T cell clones may predict response to TIL therapy. Further, TCR sequencing suggested skewing of TCR repertoire during in vitro expansion, promoting certain low frequency clonotypes. CONCLUSIONS Priming with vemurafenib before infusion of TILs was safe and feasible, and induced objective clinical responses in this cohort of patients with checkpoint inhibitor-resistant metastatic melanoma. In this trial, vemurafenib treatment seemed to decrease attrition and could be considered to bridge the waiting time while TILs are prepared.
Collapse
Affiliation(s)
- Troels Holz Borch
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Katja Harbst
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Aynal Haque Rana
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Rikke Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Evelina Martinenaite
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Per Kongsted
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Magnus Pedersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Morten Nielsen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Julie Westerlin Kjeldsen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Anders Handrup Kverneland
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark
| | - Martin Lauss
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Lisbet Rosenkrantz Hölmich
- Department of Plastic Surgery, Herlev University Hospital, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Helle Hendel
- Department of Clinical Physiology and Nuclear Medicine, Herlev University Hospital, Herlev, Denmark
| | - Özcan Met
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark.,Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Göran Jönsson
- Department of Oncology, Clinical Sciences, Lund University, Lund, Sweden
| | - Marco Donia
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Inge Marie Svane
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev University Hospital, Herlev, Denmark .,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
279
|
Indini A, Roila F, Grossi F, Massi D, Mandalà M. Impact of Circulating and Tissue Biomarkers in Adjuvant and Neoadjuvant Therapy for High-Risk Melanoma: Ready for Prime Time? Am J Clin Dermatol 2021; 22:511-522. [PMID: 34036489 PMCID: PMC8200339 DOI: 10.1007/s40257-021-00608-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2021] [Indexed: 12/17/2022]
Abstract
The prognosis of patients with metastatic melanoma has substantially improved over the last years with the advent of novel treatment strategies, mainly immune checkpoint inhibitors and BRAF and MEK inhibitors. Given the survival benefit provided in the metastatic setting and the evidence from prospective clinical trials in the early stages, these drugs have been introduced as adjuvant therapies for high-risk resected stage III disease. Several studies have also investigated immune checkpoint inhibitors, as well as BRAF and MEK inhibitors, for neoadjuvant treatment of high-risk stage III melanoma, with preliminary evidence suggesting this could be a very promising approach in this setting. However, even with new strategies, the risk of disease recurrence varies widely among stage III patients, and no available biomarkers for predicting disease recurrence have been established to date. Improved risk stratification is particularly relevant in this setting to avoid unnecessary treatment for patients who have minimum risk of disease recurrence and to reduce toxicities and costs. Research for predictive and prognostic biomarkers in this setting is ongoing to potentially shed light on the complex interplay between the tumor and the host immune system, and to further personalize treatment. This review provides an insight into available data on circulating and tissue biomarkers, including the tumor microenvironment and associated gene signatures, and their predictive and prognostic role during neoadjuvant and adjuvant treatment for cutaneous high-risk melanoma patients.
Collapse
Affiliation(s)
- Alice Indini
- Medical Oncology Unit, Department of Internal Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Fausto Roila
- Unit of Medical Oncology, Department of Surgery and Medicine, University of Perugia, Perugia, Italy
| | - Francesco Grossi
- Unit of Medical Oncology, Ospedale di Circolo e Fondazione Macchi, Università dell'Insubria, Varese, Italy
| | - Daniela Massi
- Section of Pathological Anatomy, Department of Health Sciences, University of Florence, Florence, Italy
| | - Mario Mandalà
- Unit of Medical Oncology, Department of Surgery and Medicine, University of Perugia, Perugia, Italy.
| |
Collapse
|
280
|
Battisti NML, Decoster L, Williams GR, Kanesvaran R, Wildiers H, Ring A. Targeted Therapies in Older Adults With Solid Tumors. J Clin Oncol 2021; 39:2128-2137. [PMID: 34043448 PMCID: PMC8260907 DOI: 10.1200/jco.21.00132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/07/2021] [Accepted: 02/23/2021] [Indexed: 12/23/2022] Open
Affiliation(s)
- Nicolò Matteo Luca Battisti
- Department of Medicine—Breast Unit, The Royal Marsden NHS Foundation Trust, Breast Cancer Research Division, The Institute of Cancer Research, Surrey, United Kingdom
| | - Lore Decoster
- Department of Medical Oncology, Universitair Ziekenhuis (UZ) Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Grant R. Williams
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham School of Medicine, Birmingham, AL
| | | | - Hans Wildiers
- Department of General Medical Oncology and Multidisciplinary Breast Centre, University Hospitals Leuven, Leuven Cancer Institute, Laboratory of Experimental Oncology, Leuven, Belgium
- Department of Oncology, KU Leuven, Leuven, Belgium
| | - Alistair Ring
- Department of Medicine—Breast Unit, The Royal Marsden NHS Foundation Trust, Breast Cancer Research Division, The Institute of Cancer Research, Surrey, United Kingdom
| |
Collapse
|
281
|
Peng C, Jie-Xin L. The incidence and risk of cutaneous toxicities associated with dabrafenib in melanoma patients: a systematic review and meta-analysis. Eur J Hosp Pharm 2021; 28:182-189. [PMID: 32883694 PMCID: PMC8239268 DOI: 10.1136/ejhpharm-2020-002347] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/20/2020] [Accepted: 06/30/2020] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVE Dabrafenib, an inhibitor of mutated BRAF, has significant clinical activity in melanoma patients but is linked to a spectrum of cutaneous toxicities. Thus, our meta-analysis was conducted to evaluate the type, incidence and risks of dermatological toxicities from dabrafenib. METHODS Systematic searches were performed using electronic databases such as Embase and PubMed and conference abstracts published by the American Society of Clinical Oncology. Eligible studies were limited to prospective phase I, II and III clinical trials and expanded-access (ie, outside clinical trials) programmes of melanoma patients receiving dabrafenib monotherapy (150 mg, twice daily) or combination therapy of dabrafenib (150 mg, twice daily) plus trametinib (2 mg, once daily). The outcomes were mainly the incidence rate and risk of all-grade cutaneous toxicities associated with dabrafenib in melanoma patients. RESULTS Twenty trials comprising a total of 3359 patients were included in the meta-analysis. The meta-analysis showed that the overall incidence of all-grade rash for melanoma patients assigned dabrafenib was 30.00% (95% CI 0.07 to 0.71), cutaneous squamous-cell carcinoma (cSCC) 16.00% (95% CI 0.11 to 0.24), alopecia 21% (95% CI 0.11 to 0.37), keratoacanthoma (KA) 20.00% (95% CI 0.12 to 0.31), hyperkeratosis (HK) 14.00% (95% CI 0.09 to 0.22) and pruritus 8.00% (95% CI 0.05 to 0.12). All-grade rash occurred in 19.00% (95% CI 0.15 to 0.25), cSCC in 10.00% (95% CI 0.04 to 0.22), alopecia in 6.00% (95% CI 0.03 to 0.12), KA in 6.00% (95% CI 0.04 to 0.09) and pruritus in 2/1265 patients assigned dabrafenib plus trametinib. The summary risk ratio (RR) showed that the combination of dabrafenib with trametinib versus dabrafenib was associated with a significantly increased risk of all-grade rash (RR 1.35, 95% CI 1.01 to 1.80) and a decreased risk of cSCC (RR 0.40, 95% CI 0.18 to 0.89), alopecia (RR 0.19, 95% CI 0.12 to 0.30) and HK (RR 0.25, 95% CI 0.10 to 0.62). CONCLUSION In summary, the most frequent cutaneous adverse reactions from dabrafenib were rash, cSCC, alopecia, KA, HK and pruritus. There was a significantly decreased risk of cSCC, alopecia and HK with the combination of dabrafenib with trametinib versus dabrafenib alone. Clinicians should be aware of these risks and perform regular clinical monitoring.
Collapse
Affiliation(s)
- Chen Peng
- Department of Pharmacy, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | | |
Collapse
|
282
|
Bauer C, Herwig R, Lienhard M, Prasse P, Scheffer T, Schuchhardt J. Large-scale literature mining to assess the relation between anti-cancer drugs and cancer types. J Transl Med 2021; 19:274. [PMID: 34174885 PMCID: PMC8236166 DOI: 10.1186/s12967-021-02941-z] [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: 03/18/2021] [Accepted: 06/13/2021] [Indexed: 12/09/2022] Open
Abstract
Background There is a huge body of scientific literature describing the relation between tumor types and anti-cancer drugs. The vast amount of scientific literature makes it impossible for researchers and physicians to extract all relevant information manually. Methods In order to cope with the large amount of literature we applied an automated text mining approach to assess the relations between 30 most frequent cancer types and 270 anti-cancer drugs. We applied two different approaches, a classical text mining based on named entity recognition and an AI-based approach employing word embeddings. The consistency of literature mining results was validated with 3 independent methods: first, using data from FDA approvals, second, using experimentally measured IC-50 cell line data and third, using clinical patient survival data. Results We demonstrated that the automated text mining was able to successfully assess the relation between cancer types and anti-cancer drugs. All validation methods showed a good correspondence between the results from literature mining and independent confirmatory approaches. The relation between most frequent cancer types and drugs employed for their treatment were visualized in a large heatmap. All results are accessible in an interactive web-based knowledge base using the following link: https://knowledgebase.microdiscovery.de/heatmap. Conclusions Our approach is able to assess the relations between compounds and cancer types in an automated manner. Both, cancer types and compounds could be grouped into different clusters. Researchers can use the interactive knowledge base to inspect the presented results and follow their own research questions, for example the identification of novel indication areas for known drugs. Supplementary Information The online version contains supplementary material available at 10.1186/s12967-021-02941-z.
Collapse
Affiliation(s)
- Chris Bauer
- MicroDiscovery GmbH, Marienburger Straße 1, 10405, Berlin, Germany.
| | - Ralf Herwig
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195, Berlin, Germany
| | - Matthias Lienhard
- Department of Computational Molecular Biology, Max Planck Institute for Molecular Genetics, Ihnestraße 63, 14195, Berlin, Germany
| | - Paul Prasse
- Department of Informatics, University of Potsdam, August-Bebel-Str. 89, 14482, Potsdam, Germany
| | - Tobias Scheffer
- Department of Informatics, University of Potsdam, August-Bebel-Str. 89, 14482, Potsdam, Germany
| | | |
Collapse
|
283
|
Raynal M, Alvarez JC, Saiag P, Beauchet A, Funck-Brentano C, Funck-Brentano E. Monitoring of plasma concentrations of dabrafenib and trametinib in advanced BRAFV600 mut melanoma patients. Ann Dermatol Venereol 2021; 149:32-38. [PMID: 34183171 DOI: 10.1016/j.annder.2021.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/28/2021] [Accepted: 04/14/2021] [Indexed: 10/21/2022]
Abstract
BACKGROUND Dabrafenib (D) and trametinib (T) improved survival in patients with BRAFV600mut melanoma. High plasma concentration of D (PCD) is weakly associated with adverse events (AE). We investigated the relationship between PCD/T and tumour control or AE. METHODS We analysed PCD/T in patients treated with D+T for metastatic melanoma. We collected data of tumour response (RECIST 1.1) and AE (CTCAE 4.0) blinded to PCD/T results. RESULTS We analysed 71 D and 58T assays from 34 patients. High inter-individual variability of PCD (median: 65.0ng/mL; interquartile range (IQR) [4-945]) and of PCT (median: 8.6ng/mL; IQR [5-39]) was observed. We found a weak relationship between PCD and progression-free survival, taking follow-up time into account (hazard ratio 0.991; 95%CI, 0.981 to 1.000; P=0.06). However, no difference was observed between mean PCD/T of progressing patients (n=21; 125±183ng/mL and 9.3±3.6ng/mL, respectively) and responders (complete, partial or stable response) (n=13; 159±225ng/mL, P=0.58 and 10.6±24.4ng/mL, P=0.29, respectively). No significant relationship was found between PCD/T and most common AEs (fever, lymphopenia, CPK increase, and hepatic cytolysis), body mass index, or age. Mean CPT (n=16) was significantly higher for female subjects (n=18; 11.5±4.8ng/mL) than for male subjects (8.8ng/mL±2.9, P=0.01), but no difference was observed between sex and CPD (P=0.32). CONCLUSION Our study showed a weak relationship between PCD and progression-free survival, but no relationship between PCD/T and AE was found. Monitoring PCD and PCT alone is unlikely to be useful in assessing response to treatment.
Collapse
Affiliation(s)
- M Raynal
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France; Research Unit EA4340 'Biomarkers and clinical trials in oncology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris - Saclay University, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France
| | - J-C Alvarez
- Department of Pharmacology and Toxicology, Versailles Saint-Quentin-en-Yvelines University, Paris-Saclay University, Inserm U-1173, Raymond Poincaré hospital, AP-HP, 104, boulevard Raymond Poincaré, 92380 Garches, France
| | - P Saiag
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France; Research Unit EA4340 'Biomarkers and clinical trials in oncology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris - Saclay University, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France
| | - A Beauchet
- Department of Bioinformatics, Ambroise Paré Hospital, AP-HP, 9 avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France
| | - C Funck-Brentano
- Sorbonne Université, INSERM CIC Paris-Est (CIC-1901), AP-HP, Sorbonne Université, ICAN, Pitié-Salpêtrière Hospital, Department of Pharmacology, 47-83, boulevard de l'Hôpital, 75013 Paris, France
| | - E Funck-Brentano
- Department of General and Oncologic Dermatology, Ambroise-Paré hospital, AP-HP, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France; Research Unit EA4340 'Biomarkers and clinical trials in oncology and onco-hematology', Versailles-Saint-Quentin-en-Yvelines University, Paris - Saclay University, 9, avenue Charles de Gaulle, 92100 Boulogne-Billancourt, France.
| |
Collapse
|
284
|
Moore A, Bar Y, Maurice-Dror C, Finkel I, Goldvaser H, Dudnik E, Goldstein DA, Gordon N, Billan S, Gutfeld O, Wolf I, Popovtzer A. Next-generation sequencing in thyroid cancers: do targetable alterations lead to a therapeutic advantage?: A multicenter experience. Medicine (Baltimore) 2021; 100:e26388. [PMID: 34160418 PMCID: PMC8238320 DOI: 10.1097/md.0000000000026388] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 04/29/2021] [Accepted: 05/25/2021] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Radioiodine-refractory thyroid cancers (IRTCs) are uncommon and have a poor prognosis. Treatment options for radioiodine-refractory and anaplastic tumors (ATCs) are limited. Although the genomic landscape of thyroid cancer has been studied, there is little evidence on whether next-generation sequencing (NGS) findings translate to tumor control.We analyzed all patients with IRTC and ATC who underwent commercially available NGS in 3 cancer centers.Twenty-two patients were identified, 16 patients with IRTCs and 6 patients with ATCs. Eighteen (82%) had targetable findings in NGS, nine patients were treated accordingly. Median progression-free survival for targeted treatment was 50 months [95% confidence interval (CI95%) 9.8-66.6] and2 months (CI95% 0.2-16.5) for IRTC and ATC, respectively. Of 4 patients who achieved durable responses of 7 to 50 months, 2 are ongoing. The estimated median OS of IRTC receiving targeted treatment was not reached (CI95% 89.7-111.4 months) and was 77.8 months (CI95% 52.5-114.6) for patients treated conventionally (P = .3).NGS may detect clinically significant genetic alterations and benefit patients with advanced thyroid cancers.
Collapse
Affiliation(s)
- Assaf Moore
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Yael Bar
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Corinne Maurice-Dror
- Institute of Oncology, Rambam Health Care Campus
- Ruth & Bruce Rappaport, Faculty of Medicine, Technion Israel Institute of Technology, Haifa
| | - Inbar Finkel
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
| | - Hadar Goldvaser
- Oncology Institute, Shaare Zedek Medical Center
- The Hebrew University Faculty of Medicine, Jerusalem, Israel
| | - Elizabeth Dudnik
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Daniel A. Goldstein
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| | - Noa Gordon
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
| | - Salem Billan
- Institute of Oncology, Rambam Health Care Campus
- Ruth & Bruce Rappaport, Faculty of Medicine, Technion Israel Institute of Technology, Haifa
| | - Orit Gutfeld
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Ido Wolf
- Sackler Faculty of Medicine, Tel Aviv University
- Oncology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv
| | - Aron Popovtzer
- Institute of Oncology, Davidoff Center, Rabin Medical Center – Beilinson Hospital, Petach Tikva
- Sackler Faculty of Medicine, Tel Aviv University
| |
Collapse
|
285
|
Becquart O, Oriano B, Dalle S, Mortier L, Leccia MT, Dutriaux C, Dalac S, Montaudié H, De Quatrebarbes J, Brunet-Possenti F, Saiag P, Lesimple T, Beylot-Barry M, Aubin F, Stoebner PE, Arnault JP, Dreno B, Porcher R, Lebbe C, Guillot B. Tolerance and Effectiveness of Targeted Therapies in Aged Patients with Metastatic Melanoma. Cancers (Basel) 2021; 13:cancers13123042. [PMID: 34207200 PMCID: PMC8235702 DOI: 10.3390/cancers13123042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/14/2021] [Indexed: 12/31/2022] Open
Abstract
Simple Summary A majority of melanoma occurs in people over 65 years. BRAF and MEK inhibitors are standard of care for BRAF mutated metastatic melanoma. The aim of the study was to explore tolerability of targeted therapy in a cohort of patients extracted from a biobank. Patients treated by BRAF and/or MEK inhibitors were included in two groups (<65 or >65 years) and analyzed for tolerance and efficacy. The cohort included 353 patients: 231 < 65 years and 122 > 65. A total of 80% had at least one adverse effect mainly skin, general, and gastrointestinal disorders. No statistical difference was observed for severe adverse events, adverse events grades, dose modifications, and interruptions in the two groups. Median overall survival was 20.3 and 16.3 months, respectively. This study shows that tolerance of targeted therapy is as good in older patients as in younger with a similar efficacy. There is no argument against using these treatments in elderly people. Abstract Purpose: Melanoma’s incidence is increasing, and elderly people could be significantly impacted since the majority occurs in people over 65 years of age. Combined BRAF and MEK targeted therapies (TT) are current standard regimen for BRAF mutated metastatic melanoma (MM). Except for subgroups of pivotal trials, little data are available for TT in this population. Materials and Methods: Outcomes were explored in real life patients from MelBase, a French multicentric biobank dedicated to the prospective follow-up of unresectable stage III or IV melanoma. Patients treated by BRAF TT and/or MEK TT combined or not, were included from 2013 to 2017 in 2 groups: group 1 ≤ 65-year-old (yo), group 2 > 65 yo, analyzed for tolerance and efficacy. Results: 353 patients were included: 231 in group 1, 122 in group 2. Median follow-up was 12 months (M). Median time of treatment was 6.9 M. A total of 80% had at least one Adverse Effect (AE). Most frequent AE (all grades) were mainly skin and subcutaneous, general, and gastrointestinal disorders. A total of 31% of AE were grade 3–4: 28% in group 1 and 39% in group 2 (p = 0.05). No differences were observed in all AE grades proportion, dose modifications, interruptions, and discontinuations. For each group, median overall survival was 20.3 M (CI 95%: 15.5–27.9) and 16.3 M (CI: 14.5–26.9), respectively (p = 0.8). Median progression free survival was 7.8 M (6.4–9.9) and 7.7 M (CI: 5.8–11.3) (p = 0.4). Objective response rate was 59% and 50% (p = 0.6). Conclusion: This study on a large multicentric cohort is the first to assess that TT is well tolerated in elderly BRAF-mutated patients such as in patients younger than 65. Efficacy was similar between groups with outcomes reaching those from pivotal studies. There is thus no argument against using TT in elderly people, although an onco-geriatric opinion is welcome for the most vulnerable.
Collapse
Affiliation(s)
- Ondine Becquart
- CHU de Montpellier, Service de Dermatologie, 34295 Montpellier, France;
| | - Bastien Oriano
- Hôpital St Louis, APHP, Service de Dermatologie, 75010 Paris, France; (B.O.); (C.L.)
- Hôpital Hôtel-Dieu, APHP, Centre d’Épidémiologie Clinique, 75010 Paris, France;
| | - Stéphane Dalle
- Service de Dermatologie, Hospices Civils de Lyon, Centre de Recherche en Cancérologie de Lyon, 69002 Lyon, France;
| | | | | | - Caroline Dutriaux
- CHU Bordeaux Saint-André, Service de Dermatologie, 33000 Bordeaux, France; (C.D.); (M.B.-B.)
| | - Sophie Dalac
- CHU Dijon, Service de Dermatologie, 21000 Dijon, France;
| | | | | | | | - Philippe Saiag
- Hôpital Ambroise Pare, APHP, Service de Dermatologie, 92100 Boulogne-Billancourt, France;
| | | | - Marie Beylot-Barry
- CHU Bordeaux Saint-André, Service de Dermatologie, 33000 Bordeaux, France; (C.D.); (M.B.-B.)
| | - Francois Aubin
- CHU Jean Mermoz, Service de Dermatologie, 25000 Besançon, France;
| | | | | | - Brigitte Dreno
- CHU Nantes, Service de Dermatologie, 44000 Nantes, France;
| | - Raphael Porcher
- Hôpital Hôtel-Dieu, APHP, Centre d’Épidémiologie Clinique, 75010 Paris, France;
| | - Celeste Lebbe
- Hôpital St Louis, APHP, Service de Dermatologie, 75010 Paris, France; (B.O.); (C.L.)
| | - Bernard Guillot
- Departement de Dermatologie, University of Montpellier, 34000 Montpellier, France
- Correspondence:
| |
Collapse
|
286
|
Wang X, Haaland B, Hu-Lieskovan S, Colman H, Holmen SL. First line immunotherapy extends brain metastasis free survival, improves overall survival, and reduces the incidence of brain metastasis in patients with advanced melanoma. Cancer Rep (Hoboken) 2021; 4:e1419. [PMID: 34137219 PMCID: PMC8714542 DOI: 10.1002/cnr2.1419] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Recent advances in targeted therapy and immunotherapy have improved the prognosis of melanoma patients but brain metastasis remains a major challenge. Currently, it is unclear how existing therapies can be best used to prevent or treat brain metastasis in melanoma patients. AIMS We aimed to assess brain metastasis free survival (BMFS), overall survival (OS), incidence of brain metastases, and sequencing strategies of immunotherapy and targeted therapy in patients with BRAF-mutated advanced melanoma. METHODS AND RESULTS We retrospectively analyzed 683 patients with BRAF-mutated advanced melanoma treated with first line (1L) immunotherapy (N = 266) or targeted therapy (N = 417). The primary outcome was BMFS. Secondary outcomes included OS of all patients and incidence of brain metastases in patients without documented brain metastases prior to 1L therapy. The median BMFS was 13.7 months [95% confidence interval (CI): 12.4-16.0] among all patients. The median BMFS for patients receiving 1L immunotherapy was 41.9 months [95% CI: 22.8-not reached (NR)] and targeted therapy was 11.0 months (95% CI: 8.8-12.5). Median OS results were qualitatively similar to BMFS results. The cumulative incidence of brain metastases for patients receiving 1L targeted therapy was higher than for patients receiving 1L immunotherapy (P < .001). Patients receiving 1L anti-CTLA4 plus anti-PD1 combination immunotherapy only or followed by second line (2L) targeted therapy had better BMFS (HR 0.40, 95% CI: 0.24-0.67, P = .001), improved OS (HR 0.49, 95% CI: 0.30-0.81, P = .005), and reduced incidence of brain metastases (HR 0.47, 95% CI: 0.24-0.67, P = .047) than patients receiving 1L combination BRAF and MEK targeted therapy followed by 2L immunotherapy. CONCLUSION Patients with advanced BRAF mutant melanoma treated with 1L immunotherapy have significantly longer BMFS and OS, and reduced incidence of brain metastases, compared with those treated with 1L targeted therapy. Further studies evaluating the ability of immunotherapy and targeted therapy to improve OS and prevent brain metastases are warranted.
Collapse
Affiliation(s)
- Xuechen Wang
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Benjamin Haaland
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Population Health Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Siwen Hu-Lieskovan
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Howard Colman
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Neurosurgery, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| | - Sheri L Holmen
- Huntsman Cancer Institute, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Surgery, University of Utah Health Sciences Center, Salt Lake City, Utah, USA.,Department of Oncological Sciences, University of Utah Health Sciences Center, Salt Lake City, Utah, USA
| |
Collapse
|
287
|
Trojaniello C, Luke JJ, Ascierto PA. Therapeutic Advancements Across Clinical Stages in Melanoma, With a Focus on Targeted Immunotherapy. Front Oncol 2021; 11:670726. [PMID: 34178657 PMCID: PMC8222774 DOI: 10.3389/fonc.2021.670726] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/10/2021] [Indexed: 12/31/2022] Open
Abstract
Melanoma is the most fatal skin cancer. In the early stages, it can be safely treated with surgery alone. However, since 2011, there has been an important revolution in the treatment of melanoma with new effective treatments. Targeted therapy and immunotherapy with checkpoint inhibitors have changed the history of this disease. To date, more than half of advanced melanoma patients are alive at 5 years; despite this breakthrough, approximately half of the patients still do not respond to treatment. For these reasons, new therapeutic strategies are required to expand the number of patients who can benefit from immunotherapy or combination with targeted therapy. Current research aims at preventing primary and acquired resistance, which are both responsible for treatment failure in about 50% of patients. This could increase the effectiveness of available drugs and allow for the evaluation of new combinations and new targets. The main pathways and molecules under study are the IDO inhibitor, TLR9 agonist, STING, LAG-3, TIM-3, HDAC inhibitors, pegylated IL-2 (NKTR-214), GITR, and adenosine pathway inhibitors, among others (there are currently about 3000 trials that are evaluating immunotherapeutic combinations in different tumors). Other promising strategies are cancer vaccines and oncolytic viruses. Another approach is to isolate and remove immune cells (DCs, T cells, and NK cells) from the patient's blood or tumor infiltrates, add specific gene fragments, expand them in culture with growth factors, and re-inoculate into the same patient. TILs, TCR gene transfer, and CAR-T therapy follow this approach. In this article, we give an overview over the current status of melanoma therapies, the clinical rationale for choosing treatments, and the new immunotherapy approaches.
Collapse
Affiliation(s)
- Claudia Trojaniello
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| | - Jason J. Luke
- Cancer Immunotherapeutics Center, University of Pittsburgh Medical Center and Hillman Cancer Center, Pittsburgh, PA, United States
| | - Paolo A. Ascierto
- Unit of Melanoma, Cancer Immunotherapy and Development Therapeutics, Istituto Nazionale Tumori IRCCS Fondazione G. Pascale, Napoli, Italy
| |
Collapse
|
288
|
Stachyra-Strawa P, Ciesielka M, Janiszewski M, Grzybowska-Szatkowska L. The role of immunotherapy and molecular‑targeted therapy in the treatment of melanoma (Review). Oncol Rep 2021; 46:158. [PMID: 34109986 DOI: 10.3892/or.2021.8109] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 04/23/2021] [Indexed: 11/05/2022] Open
Abstract
Skin melanomas are malignant neoplasms originating from neuroectodermal melanocytes. Compared to other neoplasms, melanomas have a high rate of growth. Their incidence is highest in Australia and New Zealand, in high‑income European countries (Switzerland, Norway, Sweden) and in the US. In Poland, the standardized incidence rate is approximately 5/100,000. Melanomas are typically highly radioresistant and chemoresistant. Before the era of immunotherapy, inoperable lesions were treated using chemotherapy based mainly on dacarbazine, temozolomide or fotemustine, which did not yield the expected results in terms of extending survival time or improving patient comfort. Therefore, there has emerged a need to seek other solutions. In most cases, the use of immunological treatment or targeted therapy has had a positive impact on survival time and relapse‑free survival. However, these periods are still relatively short, hence the need for further research and improvement of treatment. The most promising strategies appear to be antibodies that block programmed death receptor‑1 (PD‑1) and programmed death receptor ligand‑1 (PD‑L1) molecules, anti‑CTLA4 antibodies (cytotoxic T‑lymphocyte antigen 4) and therapy with BRAF and MEK inhibitors.
Collapse
Affiliation(s)
| | - Marzanna Ciesielka
- Department of Forensic Medicine, Medical University of Lublin, 20‑093 Lublin, Poland
| | - Michał Janiszewski
- Department of Radiotherapy, Medical University of Lublin, 20‑093 Lublin, Poland
| | | |
Collapse
|
289
|
Stein MK, Oluoha O, Patel K, VanderWalde A. Precision Medicine in Oncology: A Review of Multi-Tumor Actionable Molecular Targets with an Emphasis on Non-Small Cell Lung Cancer. J Pers Med 2021; 11:518. [PMID: 34198738 PMCID: PMC8226771 DOI: 10.3390/jpm11060518] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Precision medicine is essential for the modern care of a patient with cancer. Comprehensive molecular profiling of the tumor itself is necessary to determine the presence or absence of certain targetable abnormalities or biomarkers. In particular, lung cancer is a disease for which targetable genomic alterations will soon guide therapy in the majority of cases. In this comprehensive review of solid tumor-based biomarkers, we describe the genomic alterations for which targeted agents have been approved by the United States Food and Drug Administration (FDA). While focusing on alterations leading to approvals in a tumor-agnostic fashion (MSI-h, TMB-h, NTRK) and on those alterations with approvals in multiple malignancies (BRAF, ERBB2, RET, BRCA, PD-L1), we also describe several biomarkers or indications that are likely to lead to an approved drug in the near future (e.g., KRAS G12C, PD-L1 amplification, HER2 overexpression in colon cancer, HER2 mutations in lung cancer). Finally, we detail the current landscape of additional actionable alterations (EGFR, ALK, ROS1, MET) in lung cancer, a biomarker-rich malignancy that has greatly benefitted from the precision oncology revolution.
Collapse
Affiliation(s)
- Matthew K. Stein
- Missouri Baptist Medical Center, Heartland Cancer Research, NCI Community Oncology Research Program, St. Louis, MO 63131, USA;
| | - Oluchukwu Oluoha
- Division of Hematology and Oncology, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (O.O.); (K.P.)
| | - Kruti Patel
- Division of Hematology and Oncology, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (O.O.); (K.P.)
| | - Ari VanderWalde
- West Cancer Center and Research Institute, Germantown, TN 38138, USA
| |
Collapse
|
290
|
Yen I, Shanahan F, Lee J, Hong YS, Shin SJ, Moore AR, Sudhamsu J, Chang MT, Bae I, Dela Cruz D, Hunsaker T, Klijn C, Liau NPD, Lin E, Martin SE, Modrusan Z, Piskol R, Segal E, Venkatanarayan A, Ye X, Yin J, Zhang L, Kim JS, Lim HS, Kim KP, Kim YJ, Han HS, Lee SJ, Kim ST, Jung M, Hong YH, Noh YS, Choi M, Han O, Nowicka M, Srinivasan S, Yan Y, Kim TW, Malek S. ARAF mutations confer resistance to the RAF inhibitor belvarafenib in melanoma. Nature 2021; 594:418-423. [PMID: 33953400 DOI: 10.1038/s41586-021-03515-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Although RAF monomer inhibitors (type I.5, BRAF(V600)) are clinically approved for the treatment of BRAFV600-mutant melanoma, they are ineffective in non-BRAFV600 mutant cells1-3. Belvarafenib is a potent and selective RAF dimer (type II) inhibitor that exhibits clinical activity in patients with BRAFV600E- and NRAS-mutant melanomas. Here we report the first-in-human phase I study investigating the maximum tolerated dose, and assessing the safety and preliminary efficacy of belvarafenib in BRAFV600E- and RAS-mutated advanced solid tumours (NCT02405065, NCT03118817). By generating belvarafenib-resistant NRAS-mutant melanoma cells and analysing circulating tumour DNA from patients treated with belvarafenib, we identified new recurrent mutations in ARAF within the kinase domain. ARAF mutants conferred resistance to belvarafenib in both a dimer- and a kinase activity-dependent manner. Belvarafenib induced ARAF mutant dimers, and dimers containing mutant ARAF were active in the presence of inhibitor. ARAF mutations may serve as a general resistance mechanism for RAF dimer inhibitors as the mutants exhibit reduced sensitivity to a panel of type II RAF inhibitors. The combination of RAF plus MEK inhibition may be used to delay ARAF-driven resistance and suggests a rational combination for clinical use. Together, our findings reveal specific and compensatory functions for the ARAF isoform and implicate ARAF mutations as a driver of resistance to RAF dimer inhibitors.
Collapse
Affiliation(s)
- Ivana Yen
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Frances Shanahan
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Jeeyun Lee
- Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea.,Department of Intelligence Precision Healthcare Convergence, Samsung Medical Center, Sungkyunkwan University School of Medicine, Suwon, South Korea
| | - Yong Sang Hong
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Sang Joon Shin
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Amanda R Moore
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Jawahar Sudhamsu
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.,Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Matthew T Chang
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Inhwan Bae
- Department of New Chemical Entity Discovery, Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Darlene Dela Cruz
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Thomas Hunsaker
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Christiaan Klijn
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Nicholas P D Liau
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Eva Lin
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Scott E Martin
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Zora Modrusan
- Department of Microchemistry, Proteomics and Lipidomics, Genentech Inc., South San Francisco, CA, USA
| | - Robert Piskol
- Department of Bioinformatics, Genentech Inc., South San Francisco, CA, USA
| | - Ehud Segal
- Department of Translational Oncology, Genentech Inc., South San Francisco, CA, USA
| | | | - Xin Ye
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA
| | - Jianping Yin
- Department of Structural Biology, Genentech Inc., South San Francisco, CA, USA
| | - Liangxuan Zhang
- Department of Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Jin-Soo Kim
- Department of Internal Medicine, Seoul National University Boramae Medical Center, Seoul, South Korea
| | - Hyeong-Seok Lim
- Department of Clinical Pharmacology and Therapeutics, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kyu-Pyo Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yu Jung Kim
- Division of Hematology and Medical Oncology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Hye Sook Han
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, South Korea
| | - Soo Jung Lee
- Department of Oncology/Hematology, Kyungpook National University Chilgok Hospital, Kyungpook National University, Daegu, South Korea
| | - Seung Tae Kim
- Division of Hematology-Oncology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Minkyu Jung
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yoon-Hee Hong
- Department of Clinical Research and Development, Hanmi Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Young Su Noh
- Department of Clinical Research and Development, Hanmi Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Munjeong Choi
- Department of Clinical Research and Development, Hanmi Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Oakpil Han
- Department of Clinical Research and Development, Hanmi Pharmaceutical Co., Ltd., Seoul, South Korea
| | - Malgorzata Nowicka
- Department of Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Shrividhya Srinivasan
- Department of Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Yibing Yan
- Department of Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Tae Won Kim
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea.
| | - Shiva Malek
- Department of Discovery Oncology, Genentech Inc., South San Francisco, CA, USA.
| |
Collapse
|
291
|
Wurcel V, Scherrer E, Aguiar-Ibanez R, Altuna JI, Carabajal F, Jain S, Baluni G. Cost-Effectiveness of Pembrolizumab for the Adjuvant Treatment of Melanoma Patients with Lymph Node Involvement Who Have Undergone Complete Resection in Argentina. Oncol Ther 2021; 9:167-185. [PMID: 33624271 PMCID: PMC8140053 DOI: 10.1007/s40487-021-00142-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 01/23/2021] [Indexed: 01/20/2023] Open
Abstract
INTRODUCTION The KEYNOTE-054 trial demonstrated that adjuvant pembrolizumab improves recurrence-free survival in completely resected stage III melanoma versus watchful waiting (hazard ratio [HR] = 0.57; 98.4% confidence interval [CI], 0.43-0.74). We evaluated the cost-effectiveness of pembrolizumab in Argentina, where watchful waiting is still widely used among these patients despite the high risk of recurrence with surgery alone. METHODS A four-health state model was used (recurrence-free, locoregional recurrence [LR], distant metastases [DM], death). Lifetime medical costs to payers (72.08 Argentine pesos [AR$] = 1.00 U.S. dollar [USD]) and outcomes (3% annual discount) were assessed, together with incremental cost-effectiveness ratios (ICERs). First and LR→DM recurrences were modeled using KEYNOTE-054 and real-world data, respectively. No benefits of adjuvant treatment were assumed post-progression. Pre-DM and post-DM mortality was based on KEYNOTE-054 and on a network meta-analysis of advanced treatments expected in each arm, respectively. Utilities were derived from KEYNOTE-054 Euro-QoL data using an Argentinian algorithm, and from the literature. Public ex-factory drug prices were used. RESULTS Patients in the pembrolizumab and the watchful waiting arms accrued 8.78 and 5.83 quality-adjusted life-years (QALYs), 9.91 and 6.98 life-years, and costs of AR$12,698,595 (176,174 USD) and AR$11,967,717 (166,034 USD), respectively. The proportion of life-years accrued that were recurrence-free was 80.8% and 56.9% in the pembrolizumab and the watchful waiting arms, respectively. Pembrolizumab patients gained 2.94 life-years and 2.96 QALYs versus watchful waiting; the ICER per QALY was AR$247,094 (3428 USD). Recurrence rates and advanced melanoma treatments were the key drivers of the ICER. At a threshold of AR$1,445,325 (29,935 USD) per QALY, pembrolizumab had an 83.5% probability of being cost-effective versus watchful waiting. CONCLUSIONS Adjuvant pembrolizumab after complete resection of melanoma with node involvement is highly cost-effective relative to watchful waiting in Argentina, across disease stage subgroups and BRAF mutational status. This strongly supports its coverage and reimbursement across the entire health system.
Collapse
Affiliation(s)
| | | | | | | | | | - Shrishti Jain
- Complete HEOR Solutions, CHEORS, North Wales, PA, USA
| | - Gargi Baluni
- Complete HEOR Solutions, CHEORS, North Wales, PA, USA
| |
Collapse
|
292
|
Bartlett DJ, Erie AJ, Baffour FI, Broski SM, Glazebrook KN. BRAF inhibitor-induced panniculitis in patients treated for stage IV metastatic melanoma: a case series. Skeletal Radiol 2021; 50:1257-1262. [PMID: 33165713 DOI: 10.1007/s00256-020-03665-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 02/02/2023]
Abstract
BRAF and MEK inhibitor combination therapy is the standard treatment for patients with BRAF V600E mutant metastatic melanoma. Neutrophilic panniculitis is a known rare complication of BRAF inhibitor therapy and can act as a potential mimic of melanoma metastases on 18F-FDG PET/CT. In this case series, we present three cases of BRAF inhibitor-induced panniculitis in patients being treated for BRAF-mutant metastatic melanoma and emphasize the use of ultrasound to differentiate between panniculitis lesions, which are typically ill-defined echogenic masses and subcutaneous soft tissue melanoma metastases, which present as hypoechoic vascular masses.
Collapse
Affiliation(s)
- David J Bartlett
- Department of Radiology, Mayo Clinic, 200 1st street SW, Rochester, MN, 55905, USA
| | - Andrew J Erie
- Department of Radiology, Mayo Clinic, 200 1st street SW, Rochester, MN, 55905, USA
| | - Francis I Baffour
- Department of Radiology, Mayo Clinic, 200 1st street SW, Rochester, MN, 55905, USA
| | - Stephen M Broski
- Department of Radiology, Mayo Clinic, 200 1st street SW, Rochester, MN, 55905, USA
| | - Katrina N Glazebrook
- Department of Radiology, Mayo Clinic, 200 1st street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
293
|
Retinal toxicities of systemic anticancer drugs. Surv Ophthalmol 2021; 67:97-148. [PMID: 34048859 DOI: 10.1016/j.survophthal.2021.05.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/07/2023]
Abstract
Newer anticancer drugs have revolutionized cancer treatment in the last decade, but conventional chemotherapy still occupies a central position in many cancers, with combination therapy and newer methods of delivery increasing their efficacy while minimizing toxicities. We discuss the retinal toxicities of anticancer drugs with an emphasis on the mechanism of toxicity. Uveitis is seen with the use of v-raf murine sarcoma viral oncogene homolog B editing anticancer inhibitors as well as immunotherapy. Most of the cases are mild with only anterior uveitis, but severe cases of posterior uveitis, panuveitis, and Vogt-Koyanagi-Harada-like disease may also occur. In the retina, a transient neurosensory detachment is observed in almost all patients on mitogen-activated protein kinase kinase (MEK) inhibitors. Microvasculopathy is often seen with interferon α, but vascular occlusion is a more serious toxicity caused by interferon α and MEK inhibitors. Crystalline retinopathy with or without macular edema may occur with tamoxifen; however, even asymptomatic patients may develop cavitatory spaces seen on optical coherence tomography. A unique macular edema with angiographic silence is characteristic of taxanes. Delayed dark adaptation has been observed with fenretinide. Interestingly, this drug is finding potential application in Stargardt disease and age-related macular degeneration.
Collapse
|
294
|
Outcomes for systemic therapy in older patients with metastatic melanoma: Results from the Dutch Melanoma Treatment Registry. J Geriatr Oncol 2021; 12:1031-1038. [PMID: 34020909 DOI: 10.1016/j.jgo.2021.04.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 03/28/2021] [Accepted: 04/29/2021] [Indexed: 11/21/2022]
Abstract
BACKGROUND The incidence of metastatic melanoma is increasing in all ages. Multiple trials with targeted drugs and immune checkpoint inhibitors showed improved survival in metastatic melanoma. However, patients aged ≥75 years are often under-represented in clinical trials, therefore raising questions on safety and efficacy of treatment. PATIENTS AND METHODS We analyzed a real-world cohort of 3054 patients with metastatic melanoma stratified for age (≤65 years, 66-74 years and ≥ 75 years), and BRAF status, providing data on treatment strategies, toxicity, and survival. Kaplan Meier curves and Cox Proportional Hazard Models were used to present overall survival (OS) and Melanoma Specific Survival (MSS). RESULTS Overall, 52.2% of patients were ≤ 65 years and 18.4% of patients ≥75 years. BRAF mutated tumors were found less often in patients ≥75 years: 34.5% versus 65% in patients ≤65 years. Patients ≥75 years received systemic therapy less frequently compared to their younger counterparts independent of the BRAF status. When receiving treatment, no statistical significant difference in grade 3 or 4 toxicity was observed. Three year Overall Survival rate was 13.7% (9.1-19.3) in patients ≥75 years versus 26.7% (23.1-30.4) in patients ≤65 years, with a Hazard Ratio (HR) of 1.71 (95%CI 1.50-1.95), p < 0.001. Three year Melanoma Specific Survival was 30.4% (22.0-39.2) versus 34.0% (29.7-38.2), HR 1.26 (95% CI 1.07-1.49), p = 0.005 with an adjusted HR of 1.21 (1.00-1.47), p = 0.049. CONCLUSION Patients with metastatic melanoma ≥75 years are less frequently treated, but when treated there is no statistical significant increase in toxicity and only a borderline statistical significant difference in Melanoma Specific Survival was seen, compared to younger patients.
Collapse
|
295
|
Marotta V, Chiofalo MG, Di Gennaro F, Daponte A, Sandomenico F, Vallone P, Costigliola L, Botti G, Ionna F, Pezzullo L. Kinase-inhibitors for iodine-refractory differentiated thyroid cancer: still far from a structured therapeutic algorithm. Crit Rev Oncol Hematol 2021; 162:103353. [PMID: 34000414 DOI: 10.1016/j.critrevonc.2021.103353] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 01/16/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022] Open
Abstract
The kinase-inhibitors (KIs) sorafenib and lenvatinib demonstrated efficacy in iodine-refractory DTC upon phase III studies. However, evidence allowing a punctual balance of benefits and risks is poor. Furthermore, the lack of a direct comparison hampers to establish the proper sequence of administration. However, some insights may provided: a) indirect comparison between phase III trials showed milder toxicity for sorafenib, which should be preferred in case of cardiovascular comorbidities; b) prospective evidence of efficacy in KIs pre-treated patients is available only for lenvatinib, which should be used as second-line. Promising activity was found for the majority of other tested KIs, but no placebo-controlled trials are available. Emerging, but still early, frontiers include the restoration of iodine-sensitivity and the selective activity on pathogenic mutations. In conclusion, the use of KIs in iodine-refractory DTC is far from a structured therapeutic algorithm.
Collapse
Affiliation(s)
- Vincenzo Marotta
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy.
| | - Maria Grazia Chiofalo
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Francesca Di Gennaro
- Struttura Complessa Medicina Nucleare e Terapia Metabolica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Antonio Daponte
- Struttura Complessa Oncologia Clinica Sperimentale Testa-Collo e Muscolo-Scheletrica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Fabio Sandomenico
- Struttura Complessa Radiodiagnostica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Paolo Vallone
- Struttura Complessa Radiodiagnostica, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Luciana Costigliola
- Unità Operativa Compessa Di Chirugia Generale, d'Urgenza e Metabolica, Pineta Grande Hospital, Castel Volturno, Italy
| | - Gerardo Botti
- Struttura Complessa Anatomia Patologica e Citopatologia, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Franco Ionna
- Struttura Complessa Chirurgia Oncologica Maxillo-Facciale Ed ORL, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy
| | - Luciano Pezzullo
- Struttura Complessa Chirurgia Oncologica Della Tiroide, Istituto Nazionale Tumori - Irccs - Fondazione G.Pascale, Napoli, Italy.
| |
Collapse
|
296
|
Abdel-Maksoud MS, El-Gamal MI, Lee BS, Gamal El-Din MM, Jeon HR, Kwon D, Ammar UM, Mersal KI, Ali EMH, Lee KT, Yoo KH, Han DK, Lee JK, Kim G, Choi HS, Kwon YJ, Lee KH, Oh CH. Discovery of New Imidazo[2,1- b]thiazole Derivatives as Potent Pan-RAF Inhibitors with Promising In Vitro and In Vivo Anti-melanoma Activity. J Med Chem 2021; 64:6877-6901. [PMID: 33999621 DOI: 10.1021/acs.jmedchem.1c00230] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BRAF is an important component of MAPK cascade. Mutation of BRAF, in particular V600E, leads to hyperactivation of the MAPK pathway and uncontrolled cellular growth. Resistance to selective inhibitors of mutated BRAF is a major obstacle against treatment of many cancer types. In this work, a series of new (imidazo[2,1-b]thiazol-5-yl)pyrimidine derivatives possessing a terminal sulfonamide moiety were synthesized. Pan-RAF inhibitory effect of the new series was investigated, and structure-activity relationship is discussed. Antiproliferative activity of the target compounds was tested against the NCI-60 cell line panel. The most active compounds were further tested to obtain their IC50 values against cancer cells. Compound 27c with terminal open chain sulfonamide and 38a with a cyclic sulfamide moiety showed the highest activity in enzymatic and cellular assay, and both compounds were able to inhibit phosphorylation of MEK and ERK. Compound 38a was selected for testing its in vivo activity against melanoma. Cellular and animal activities are reported.
Collapse
Affiliation(s)
- Mohammed S Abdel-Maksoud
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Dokki, Giza 12622, Egypt
| | - Mohammed I El-Gamal
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates.,Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, United Arab Emirates.,Department of Medicinal Chemistry, Faculty of Pharmacy, University of Mansoura, Mansoura 35516, Egypt
| | - Bong S Lee
- CTC SCIENCE, 38, Hyundaikia-ro, Paltan-myeon, Hwaseong-si, Gyeonggi-do 18576, Republic of Korea
| | - Mahmoud M Gamal El-Din
- Medicinal & Pharmaceutical Chemistry Department, Pharmaceutical and Drug Industries Research Division, National Research Centre (NRC), Dokki, Giza 12622, Egypt
| | - Hong R Jeon
- CTCBIO Inc., 450-34, Noha-ri, Paltan-myeon, Hwaseong-si, Gyeonggi-do 18576, Republic of Korea
| | - Dow Kwon
- CTC SCIENCE, 38, Hyundaikia-ro, Paltan-myeon, Hwaseong-si, Gyeonggi-do 18576, Republic of Korea
| | - Usama M Ammar
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, 161 Cathedral Street, Glasgow G4 0NR, Scotland, United Kingdom
| | - Karim I Mersal
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seongbuk-gu, Hwarangro 14-gil 5, Seoul 136-791, Seoul, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Yuseong-gu 34113, Republic of Korea
| | - Eslam M H Ali
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seongbuk-gu, Hwarangro 14-gil 5, Seoul 136-791, Seoul, Republic of Korea.,Department of Biomolecular Science, University of Science & Technology (UST), Daejeon, Yuseong-gu 34113, Republic of Korea
| | - Kyung-Tae Lee
- Department of Pharmaceutical Biochemistry, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea.,Department of Life and Nanopharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul 130-701, Republic of Korea
| | - Kyung Ho Yoo
- Chemical Kinomics Research Center, Korea Institute of Science & Technology (KIST), Seoul 136-791, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science, CHA University, Gyeonggi 13488, Republic of Korea
| | - Jae Kyun Lee
- Center for Neuro-Medicine, Korea Institute of Science & Technology (KIST), Seongbuk-gu, Hwarangro 14-gil 5, Seoul 136-791, Seoul, Republic of Korea
| | - Garam Kim
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Hong Seok Choi
- College of Pharmacy, Chosun University, Gwangju 61452, Republic of Korea
| | - Young Jik Kwon
- Department of Chemical Engineering and Materials Science, University of California, Irvine, California 92697, United States.,Department of Molecular Biology and Biochemistry, University of California, Irvine, California 92697, United States
| | - Kwan Hyi Lee
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seongbuk-gu, Hwarangro 14-gil 5, Seoul 136-791, Seoul, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea
| | - Chang Hyun Oh
- Center for Biomaterials, Korea Institute of Science & Technology (KIST), Seongbuk-gu, Hwarangro 14-gil 5, Seoul 136-791, Seoul, Republic of Korea
| |
Collapse
|
297
|
Ullah R, Yin Q, Snell AH, Wan L. RAF-MEK-ERK pathway in cancer evolution and treatment. Semin Cancer Biol 2021; 85:123-154. [PMID: 33992782 DOI: 10.1016/j.semcancer.2021.05.010] [Citation(s) in RCA: 247] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/03/2021] [Accepted: 05/06/2021] [Indexed: 12/13/2022]
Abstract
The RAF-MEK-ERK signaling cascade is a well-characterized MAPK pathway involved in cell proliferation and survival. The three-layered MAPK signaling cascade is initiated upon RTK and RAS activation. Three RAF isoforms ARAF, BRAF and CRAF, and their downstream MEK1/2 and ERK1/2 kinases constitute a coherently orchestrated signaling module that directs a range of physiological functions. Genetic alterations in this pathway are among the most prevalent in human cancers, which consist of numerous hot-spot mutations such as BRAFV600E. Oncogenic mutations in this pathway often override otherwise tightly regulated checkpoints to open the door for uncontrolled cell growth and neoplasia. The crosstalk between the RAF-MEK-ERK axis and other signaling pathways further extends the proliferative potential of this pathway in human cancers. In this review, we summarize the molecular architecture and physiological functions of the RAF-MEK-ERK pathway with emphasis on its dysregulations in human cancers, as well as the efforts made to target the RAF-MEK-ERK module using small molecule inhibitors.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Qing Yin
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Aidan H Snell
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA
| | - Lixin Wan
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA; Department of Cutaneous Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
298
|
Zhou S, Sikorski D, Xu H, Zubarev A, Chergui M, Lagacé F, Miller WH, Redpath M, Ghazal S, Butler MO, Petrella TM, Claveau J, Nessim C, Salopek TG, Gniadecki R, Litvinov IV. Defining the Criteria for Reflex Testing for BRAF Mutations in Cutaneous Melanoma Patients. Cancers (Basel) 2021; 13:2282. [PMID: 34068774 PMCID: PMC8126223 DOI: 10.3390/cancers13092282] [Citation(s) in RCA: 5] [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: 04/01/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/11/2022] Open
Abstract
Targeted therapy has been developed through an in-depth understanding of molecular pathways involved in the pathogenesis of melanoma. Approximately ~50% of patients with melanoma have tumors that harbor a mutation of the BRAF oncogene. Certain clinical features have been identified in BRAF-mutated melanomas (primary lesions located on the trunk, diagnosed in patients <50, visibly pigmented tumors and, at times, with ulceration or specific dermatoscopic features). While BRAF mutation testing is recommended for stage III-IV melanoma, guidelines differ in recommending mutation testing in stage II melanoma patients. To fully benefit from these treatment options and avoid delays in therapy initiation, advanced melanoma patients harboring a BRAF mutation must be identified accurately and quickly. To achieve this, clear definition and implementation of BRAF reflex testing criteria/methods in melanoma should be established so that patients with advanced melanoma can arrive to their first medical oncology appointment with a known biomarker status. Reflex testing has proven effective for a variety of cancers in selecting therapies and driving other medical decisions. We overview the pathophysiology, clinical presentation of BRAF-mutated melanoma, current guidelines, and present recommendations on BRAF mutation testing. We propose that reflex BRAF testing should be performed for every melanoma patient with stages ≥IIB.
Collapse
Affiliation(s)
- Sarah Zhou
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Daniel Sikorski
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Honghao Xu
- Division of Dermatology, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Andrei Zubarev
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - May Chergui
- Department of Pathology, McGill University, Montreal, QC H3A 0G4, Canada
| | - François Lagacé
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Wilson H Miller
- Departments of Medicine and Oncology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Margaret Redpath
- Department of Pathology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Stephanie Ghazal
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| | - Marcus O Butler
- Princess Margaret Cancer Centre, Department of Medical Oncology and Hematology, University of Toronto, Toronto, ON M5G 2C1, Canada
| | - Teresa M Petrella
- Odette Cancer Centre, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, ON M4N 3M5, Canada
| | - Joël Claveau
- Division of Dermatology, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Carolyn Nessim
- Division of General Surgery, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Thomas G Salopek
- Division of Dermatology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Robert Gniadecki
- Division of Dermatology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Ivan V Litvinov
- Division of Dermatology, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
299
|
Han D, van Akkooi ACJ, Straker RJ, Shannon AB, Karakousis GC, Wang L, Kim KB, Reintgen D. Current management of melanoma patients with nodal metastases. Clin Exp Metastasis 2021; 39:181-199. [PMID: 33961168 PMCID: PMC8102663 DOI: 10.1007/s10585-021-10099-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/22/2021] [Indexed: 12/26/2022]
Abstract
The management of melanoma patients with nodal metastases has undergone dramatic changes over the last decade. In the past, the standard of care for patients with a positive sentinel lymph node biopsy (SLNB) was a completion lymph node dissection (CLND), while patients with palpable macroscopic nodal disease underwent a therapeutic lymphadenectomy in cases with no evidence of systemic spread. However, studies have shown that SLN metastases present as a spectrum of disease, with certain SLN-based factors being prognostic of and correlated with outcomes. Furthermore, the results of key clinical trials demonstrate that CLND provides no survival benefit over nodal observation in positive SLN patients, while other clinical trials have shown that adjuvant immune checkpoint inhibitor therapy or targeted therapy after CLND is associated with a recurrence-free survival benefit. Given the efficacy of these systemic therapies in the adjuvant setting, these agents are now being evaluated and utilized as neoadjuvant treatments in patients with regionally-localized or resectable metastatic melanoma. Multiple options now exist to treat melanoma patients with nodal disease, and determining the best treatment course for a particular case requires an in-depth knowledge of current data and an informed discussion with the patient. This review will provide an overview of the various options for treating melanoma patients with nodal metastases and will discuss the data that supported the development of these treatment options.
Collapse
Affiliation(s)
- Dale Han
- Division of Surgical Oncology, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Mail Code: L619, Portland, OR, 97239, USA.
| | - Alexander C J van Akkooi
- Department of Surgical Oncology, Netherlands Cancer Institute - Antoni van Leeuwenhoek, Amsterdam, the Netherlands
| | - Richard J Straker
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Adrienne B Shannon
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Giorgos C Karakousis
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA
| | - Lin Wang
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Kevin B Kim
- California Pacific Medical Center and Research Institute, San Francisco, CA, USA
| | - Douglas Reintgen
- Department of Surgery, Morsani School of Medicine, University of South Florida, Tampa, FL, USA
| |
Collapse
|
300
|
Louveau B, Resche-Rigon M, Lesimple T, Da Meda L, Pracht M, Baroudjian B, Delyon J, Amini-Adle M, Dutriaux C, Reger de Moura C, Sadoux A, Jouenne F, Ghrieb Z, Vilquin P, Bouton D, Tibi A, Huguet S, Rezai K, Battistella M, Mourah S, Lebbe C. Phase I-II Open-Label Multicenter Study of Palbociclib + Vemurafenib in BRAF V600MUT Metastatic Melanoma Patients: Uncovering CHEK2 as a Major Response Mechanism. Clin Cancer Res 2021; 27:3876-3883. [PMID: 33947696 DOI: 10.1158/1078-0432.ccr-20-4050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/16/2021] [Accepted: 04/28/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE In BRAF V600MUT metastatic melanoma, cyclin D-CDK4/6-INK4-Rb pathway alterations are involved in resistance to MAPK inhibitors, suggesting a clinical benefit of cyclin-dependent kinase 4 (CDK4) inhibitors. In this phase I-II study, we aimed to establish the MTD of palbociclib when added to vemurafenib. PATIENTS AND METHODS Patients with BRAF V600E/KMUT metastatic melanoma harboring CDKN2A loss and RB1 expression were included and stratified into two groups according to previous BRAF inhibitor treatment (no:strata 1; yes:strata 2). Treatment comprised palbociclib once daily for 14 days followed by a 7-day break + continuous dosing of vemurafenib. The primary endpoint was the occurrence of dose-limiting toxicity (DLT), and the secondary endpoints included the best response, survival, pharmacokinetics, and tumor molecular profiling. RESULTS Eighteen patients were enrolled, with 15 in strata 2. Characteristics at inclusion were American Joint Committee on Cancer stage IVM1c (N = 16; 88.9%), high lactate dehydrogenase (N = 9; 50.0%), and median number of previous treatments of 2. One and 5 patients experienced DLT in strata 1 and 2, respectively, defining the MTD at palbociclib 25 mg and vemurafenib 960 mg in strata 2. No significant evidence for drug-drug interactions was highlighted. The median progression-free survival was 2.8 months, and 5 (27.8%) patients showed a clinical response. The baseline differential mRNA expression analysis and in vitro data revealed the role of CHEK2 in the response to palbociclib. CONCLUSIONS Although the combination of palbociclib + fixed-dose vemurafenib did not allow an increased palbociclib dosage above 25 mg, a significant clinical benefit was achieved in pretreated patients with melanoma. An association between the transcriptomic data and clinical response was highlighted.
Collapse
Affiliation(s)
- Baptiste Louveau
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
| | - Matthieu Resche-Rigon
- Department of Biostatistics, Saint Louis Hospital APHP, Paris, France
- Université de Paris, INSERM U1153, Saint Louis Hospital APHP, Paris, France
| | - Thierry Lesimple
- Oncodermatology Unit, Eugene Marquis Center, CHU CLCC, Rennes, France
| | - Laetitia Da Meda
- Department of Dermatology, Saint Louis Hospital APHP, Paris, France
- Centre d'Investigation Clinique (CIC 1427), Saint Louis Hospital APHP, Paris, France
| | - Marc Pracht
- Oncodermatology Unit, Eugene Marquis Center, CHU CLCC, Rennes, France
| | - Barouyr Baroudjian
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
- Department of Dermatology, Saint Louis Hospital APHP, Paris, France
| | - Julie Delyon
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
- Department of Dermatology, Saint Louis Hospital APHP, Paris, France
| | | | | | - Coralie Reger de Moura
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
| | - Aurélie Sadoux
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
| | - Fanélie Jouenne
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
| | - Zineb Ghrieb
- Centre d'Investigation Clinique (CIC 1427), Saint Louis Hospital APHP, Paris, France
| | - Paul Vilquin
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
| | | | | | | | | | - Maxime Battistella
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
- Department of Pathology, APHP Saint Louis Hospital, Paris, France
| | - Samia Mourah
- Department of Pharmacology and Solid Tumor Genomics, Saint Louis Hospital APHP, Paris, France
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France
| | - Céleste Lebbe
- Université de Paris, INSERM U976, Team 1, Human Immunology Pathophysiology and Immunotherapy (HIPI), Saint Louis Hospital APHP, Paris, France.
- Department of Dermatology, Saint Louis Hospital APHP, Paris, France
| |
Collapse
|