scispace - formally typeset
Search or ask a question

Showing papers by "Michael D. Prados published in 2016"


Journal ArticleDOI
TL;DR: PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy, and additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.
Abstract: Background The colony stimulating factor 1 receptor (CSF1R) ligands, CSF1 and interleukin-34, and the KIT ligand, stem cell factor, are expressed in glioblastoma (GB). Microglia, macrophages, blood vessels, and tumor cells also express CSF1R, and depletion of the microglia reduces tumor burden and invasive capacity. PLX3397 is an oral, small molecule that selectively inhibits CSF1R and KIT, penetrates the blood-brain barrier in model systems, and represents a novel approach for clinical development. Methods We conducted a phase II study in patients with recurrent GB. The primary endpoint was 6-month progression-free survival (PFS6). Secondary endpoints included overall survival response rate, safety, and plasma/tumor tissue pharmacokinetics. Exploratory endpoints included pharmacodynamic measures of drug effect in blood and tumor tissue. Results A total of 37 patients were enrolled, with 13 treated prior to a planned surgical resection (Cohort 1) and 24 treated without surgery (Cohort 2). PLX3397 was given at an oral dose of 1000 mg daily and was well tolerated. The primary efficacy endpoint of PFS6 was only 8.6%, with no objective responses. Pharmacokinetic endpoints revealed a median maximal concentration (Cmax) of 8090 ng/mL, with a time to attain Cmax of 2 hour in plasma. Tumor tissue obtained after 7 days of drug exposure revealed a median drug level of 5500 ng/g. Pharmacodynamic changes included an increase in colony stimulating factor 1 and reduced CD14(dim)/CD16+ monocytes in plasma compared with pretreatment baseline values. Conclusion PLX3397 was well tolerated and readily crossed the blood-tumor barrier but showed no efficacy. Additional studies are ongoing, testing combination strategies and potential biomarkers to identify patients with greater likelihood of response.

393 citations


Journal ArticleDOI
Vijay Ramaswamy1, Thomas Hielscher2, Stephen C. Mack3, Stephen C. Mack1, Alvaro Lassaletta1, Tong Lin, Kristian W. Pajtler2, David T.W. Jones2, Betty Luu1, Florence M.G. Cavalli1, Kenneth Aldape1, Marc Remke4, Martin Mynarek5, Stefan Rutkowski5, Sridharan Gururangan6, Roger E. McLendon6, Eric S. Lipp6, Christopher Dunham7, Juliette Hukin7, David D. Eisenstat8, Dorcas Fulton8, Frank van Landeghem8, Mariarita Santi9, Marie Lise C. van Veelen10, Erwin G. Van Meir11, Satoru Osuka11, Xing Fan12, Karin M. Muraszko12, Daniela Pretti da Cunha Tirapelli13, Sueli Mieko Oba-Shinjo13, Suely Kazue Nagahashi Marie13, Carlos Gilberto Carlotti13, Ji Yeoun Lee14, Amulya A. Nageswara Rao15, Caterina Giannini15, Claudia C. Faria16, Sofia Nunes17, Jaume Mora18, Ronald L. Hamilton19, Peter Hauser20, Nada Jabado21, Kevin Petrecca21, Shin Jung22, Luca Massimi23, Massimo Zollo24, Giuseppe Cinalli24, László Bognár25, Almos Klekner25, Tibor Hortobágyi25, Sarah Leary26, Sarah Leary27, Sarah Leary28, Ralph P. Ermoian26, Ralph P. Ermoian27, Ralph P. Ermoian28, James M. Olson28, James M. Olson26, James M. Olson27, Jeffrey R. Leonard29, Corrine Gardner29, Wiesława Grajkowska, Lola B. Chambless30, Jason E. Cain31, Charles G. Eberhart32, Sama Ahsan32, Maura Massimino, Felice Giangaspero, Francesca R. Buttarelli33, Roger J. Packer34, Lyndsey Emery9, William H. Yong35, Horacio Soto35, Linda M. Liau35, Richard Everson35, Andrew J. Grossbach36, Tarek Shalaby37, Michael A. Grotzer37, Matthias A. Karajannis38, David Zagzag38, Helen Wheeler39, Katja von Hoff5, Marta M. Alonso40, Teresa Tuñón, Ulrich Schüller41, Karel Zitterbart42, Jaroslav Sterba42, Jennifer A. Chan43, Miguel A. Guzman44, Samer K. Elbabaa44, Howard Colman45, Girish Dhall46, Paul G. Fisher47, Maryam Fouladi48, Amar Gajjar, Stewart Goldman49, Eugene Hwang34, Marcel Kool2, Harshad Ladha50, Elizabeth Vera-Bolanos50, Khalida Wani50, Frank S. Lieberman19, Tom Mikkelsen51, Antonio Omuro52, Ian F. Pollack19, Michael D. Prados53, H. Ian Robins54, Riccardo Soffietti55, Jing Wu56, Phillipe Metellus, Uri Tabori1, Ute Bartels1, Eric Bouffet1, Cynthia Hawkins1, James T. Rutka1, Peter B. Dirks1, Stefan M. Pfister2, Stefan M. Pfister57, Thomas E. Merchant, Mark R. Gilbert50, Mark R. Gilbert58, Terri S. Armstrong59, Andrey Korshunov2, David W. Ellison, Michael D. Taylor1 
University of Toronto1, German Cancer Research Center2, Cleveland Clinic3, University of Düsseldorf4, University of Hamburg5, Duke University6, University of British Columbia7, University of Alberta8, University of Pennsylvania9, Erasmus University Rotterdam10, Emory University11, University of Michigan12, University of São Paulo13, Seoul National University14, Mayo Clinic15, University of Lisbon16, Instituto Português de Oncologia Francisco Gentil17, University of Barcelona18, University of Pittsburgh19, Semmelweis University20, McGill University21, Chonnam National University22, The Catholic University of America23, University of Naples Federico II24, University of Debrecen25, Fred Hutchinson Cancer Research Center26, Seattle Children's27, University of Washington28, Washington University in St. Louis29, Vanderbilt University30, Hudson Institute of Medical Research31, Johns Hopkins University32, Sapienza University of Rome33, Children's National Medical Center34, University of California, Los Angeles35, University of Iowa36, University of Zurich37, New York University38, University of Sydney39, University of Navarra40, Ludwig Maximilian University of Munich41, Masaryk University42, University of Calgary43, Saint Louis University44, University of Utah45, University of Southern California46, Stanford University47, University of Cincinnati48, Children's Memorial Hospital49, University of Texas MD Anderson Cancer Center50, Henry Ford Health System51, Memorial Sloan Kettering Cancer Center52, University of California, San Francisco53, University of Wisconsin-Madison54, University of Turin55, University of North Carolina at Chapel Hill56, Heidelberg University57, National Institutes of Health58, University of Texas Health Science Center at Houston59
TL;DR: The most impactful biomarker for posterior fossa ependymoma is molecular subgroup affiliation, independent of other demographic or treatment variables, with the survival rates being particularly poor for subtotally resected EPN_PFA, even with adjuvant radiation therapy.
Abstract: PurposePosterior fossa ependymoma comprises two distinct molecular variants termed EPN_PFA and EPN_PFB that have a distinct biology and natural history. The therapeutic value of cytoreductive surgery and radiation therapy for posterior fossa ependymoma after accounting for molecular subgroup is not known.MethodsFour independent nonoverlapping retrospective cohorts of posterior fossa ependymomas (n = 820) were profiled using genome-wide methylation arrays. Risk stratification models were designed based on known clinical and newly described molecular biomarkers identified by multivariable Cox proportional hazards analyses.ResultsMolecular subgroup is a powerful independent predictor of outcome even when accounting for age or treatment regimen. Incompletely resected EPN_PFA ependymomas have a dismal prognosis, with a 5-year progression-free survival ranging from 26.1% to 56.8% across all four cohorts. Although first-line (adjuvant) radiation is clearly beneficial for completely resected EPN_PFA, a substantia...

131 citations


Journal ArticleDOI
TL;DR: The results support further investigation and possible clinical translation of palbociclib as an adjuvant to radiation therapy for patients with malignant brain tumors that retain RB expression.
Abstract: BACKGROUND Radiation therapy is the most commonly used postsurgical treatment for primary malignant brain tumors. Consequently, investigating the efficacy of chemotherapeutics combined with radiation for treating malignant brain tumors is of high clinical relevance. In this study, we examined the cyclin-dependent kinase 4/6 inhibitor palbociclib, when used in combination with radiation for treating human atypical teratoid rhabdoid tumor (ATRT) as well as glioblastoma (GBM). METHODS Evaluation of treatment antitumor activity in vitro was based upon results from cell proliferation assays, clonogenicity assays, flow cytometry, and immunocytochemistry for DNA double-strand break repair. Interpretation of treatment antitumor activity in vivo was based upon bioluminescence imaging, animal subject survival analysis, and staining of tumor sections for markers of proliferation and apoptosis. RESULTS For each of the retinoblastoma protein (RB)-proficient tumor models examined (2 ATRTs and 2 GBMs), one or more of the combination therapy regimens significantly (P < .05) outperformed both monotherapies with respect to animal subject survival benefit. Among the combination therapy regimens, concurrent palbociclib and radiation treatment and palbociclib treatment following radiation consistently outperformed the sequence in which radiation followed palbociclib treatment. In vitro investigation revealed that the concurrent use of palbociclib with radiation, as well as palbociclib following radiation, inhibited DNA double-strand break repair and promoted increased tumor cell apoptosis. CONCLUSIONS Our results support further investigation and possible clinical translation of palbociclib as an adjuvant to radiation therapy for patients with malignant brain tumors that retain RB expression.

98 citations


Journal ArticleDOI
TL;DR: While this study failed to meet the primary endpoint for objective radiographic response, patients with high-risk low-grade glioma receiving adjuvant temozolomide demonstrated a high rate of radiographic stability and favorable survival outcomes while meaningfully delaying radiotherapy.
Abstract: Background Optimal adjuvant management of adult low-grade gliomas is controversial. Recently described tumor classification based on molecular subtype has the potential to individualize adjuvant therapy but has not yet been evaluated as part of a prospective trial. Methods Patients aged 18 or older with newly diagnosed World Health Organization grade II low-grade gliomas and gross residual disease after surgical resection were enrolled in the study. Patients received monthly cycles of temozolomide for up to 1 year or until disease progression. For patients with available tissue, molecular subtype was assessed based upon 1p/19q codeletion and isocitrate dehydrogenase-1 R132H mutation status. The primary outcome was radiographic response rate; secondary outcomes included progression-free survival (PFS) and overall survival (OS). Results One hundred twenty patients were enrolled with median follow-up of 7.5 years. Overall response rate was 6%, with median PFS and OS of 4.2 and 9.7 years, respectively. Molecular subtype was associated with rate of disease progression during treatment (P<.001), PFS (P=.007), and OS (P<.001). Patients with 1p/19q codeletion demonstrated a 0% risk of progression during treatment. In an exploratory analysis, pretreatment lesion volume was associated with both PFS (P<.001) and OS (P<.001). Conclusions While our study failed to meet the primary endpoint for objective radiographic response, patients with high-risk low-grade glioma receiving adjuvant temozolomide demonstrated a high rate of radiographic stability and favorable survival outcomes while meaningfully delaying radiotherapy. Patients with 1p/19q codeletion are potential candidates for omission of adjuvant radiotherapy, but further work is needed to directly compare chemotherapy with combined modality therapy.

94 citations


Journal ArticleDOI
TL;DR: For central neurocytomas, MIB-1 labeling index >4 % is predictive of poorer outcome and the data suggest that adjuvant radiotherapy after STR may improve PFS.
Abstract: Central neurocytomas are uncommon intraventricular neoplasms whose optimal management remains controversial due to their rarity. We assessed outcomes for a historical cohort of neurocytoma patients and evaluated effects of tumor atypia, size, resection extent, and adjuvant radiotherapy. Progression-free survival (PFS) was measured by Kaplan-Meier and Cox proportional hazards methods. A total of 28 patients (15 males, 13 females) were treated between 1995 and 2014, with a median age at diagnosis of 26 years (range 5-61). Median follow-up was 62.2 months and 3 patients were lost to follow-up postoperatively. Thirteen patients experienced recurrent/progressive disease and 2-year PFS was 75% (95% CI 53-88%). Two-year PFS was 48% for MIB-1 labeling >4% versus 90% for ≤4% (HR 5.4, CI 2.2-27.8, p = 0.0026). Nine patients (32%) had gross total resections (GTR) and 19 (68%) had subtotal resections (STR). PFS for >80% resection was 83 versus 67% for ≤80% resection (HR 0.67, CI 0.23-2.0, p = 0.47). Three STR patients (16%) received adjuvant radiation which significantly improved overall PFS (p = 0.049). Estimated 5-year PFS was 67% for STR with radiotherapy versus 53% for STR without radiotherapy. Salvage therapy regimens were diverse and resulted in stable disease for 54% of patients and additional progression for 38 %. Two patients with neuropathology-confirmed atypical neurocytomas died at 4.3 and 113.4 months after initial surgery. For central neurocytomas, MIB-1 labeling index >4% is predictive of poorer outcome and our data suggest that adjuvant radiotherapy after STR may improve PFS. Most patients requiring salvage therapy will be stabilized and multiple modalities can be effectively utilized.

36 citations


Journal ArticleDOI
TL;DR: In this paper, a specific BRAF V600E inhibitor, PLX4720, was evaluated in vitro and in vivo to determine whether it enhances the activity of RT in human HGGs.
Abstract: Radiation (RT) is critical to the treatment of high-grade gliomas (HGGs) but cures remain elusive. The BRAF mutation V600E is critical to the pathogenesis of 10-20% of pediatric gliomas, and a small proportion of adult HGGs. Here we aim to determine whether PLX4720, a specific BRAF V600E inhibitor, enhances the activity of RT in human HGGs in vitro and in vivo. Patient-derived HGG lines harboring wild-type BRAF or BRAF V600E were assessed in vitro to determine IC50 values, cell cycle arrest, apoptosis and senescence and elucidate mechanisms of combinatorial activity. A BRAF V600E HGG intracranial xenograft mouse model was used to evaluate in vivo combinatorial efficacy of PLX4720+RT. Tumors were harvested for immunohistochemistry to quantify cell cycle arrest and apoptosis. RT+PLX4720 exhibited greater anti-tumor effects than either monotherapy in BRAF V600E but not in BRAF WT lines. In vitro studies showed increased Annexin V and decreased S phase cells in BRAF V600E gliomas treated with PLX4720+RT, but no significant changes in β-galactosidase levels. In vivo, concurrent and sequential PLX4720+RT each significantly prolonged survival compared to monotherapies, in the BRAF V600E HGG model. Immunohistochemistry of in vivo tumors demonstrated that PLX4720+RT decreased Ki-67 and phospho-MAPK, and increased γH2AX and p21 compared to control mice. BRAF V600E inhibition enhances radiation-induced cytotoxicity in BRAF V600E-mutated HGGs, in vitro and in vivo, effects likely mediated by apoptosis and cell cycle, but not senescence. These studies provide the pre-clinical rationale for clinical trials of concurrent radiotherapy and BRAF V600E inhibitors.

29 citations


Journal ArticleDOI
TL;DR: Integrating metabolic parameters into the assessment of patients with newly diagnosed GBM receiving therapies that include anti-angiogenic agents may be helpful for tracking changes in tumor burden, resolving ambiguities in anatomic images caused by non-specific treatment effects and for predicting outcome.
Abstract: Interpretation of changes in the T1- and T2-weighted MR images from patients with newly diagnosed glioblastoma (GBM) treated with standard of care in conjunction with anti-angiogenic agents is complicated by pseudoprogression and pseudoresponse. The hypothesis being tested in this study was that 3D H-1 magnetic resonance spectroscopic imaging (MRSI) provides estimates of levels of choline, creatine, N-acetylaspartate (NAA), lactate and lipid that change in response to treatment and that metrics describing these characteristics are associated with survival. Thirty-one patients with newly diagnosed GBM and being treated with radiation therapy (RT), temozolomide, erlotinib and bevacizumab were recruited to receive serial MR scans that included 3-D lactate edited MRSI at baseline, mid-RT, post-RT and at specific follow-up time points. The data were processed to provide estimates of metrics representing changes in metabolite levels relative to normal appearing brain. Cox proportional hazards analysis was applied to examine the relationship of these parameters with progression free survival (PFS) and overall survival (OS). There were significant reductions in parameters that describe relative levels of choline to NAA and creatine, indicating that the treatment caused a decrease in tumor cellularity. Changes in the levels of lactate and lipid relative to the NAA from contralateral brain were consistent with vascular normalization. Metabolic parameters from the first serial follow-up scan were associated with PFS and OS, when accounting for age and extent of resection. Integrating metabolic parameters into the assessment of patients with newly diagnosed GBM receiving therapies that include anti-angiogenic agents may be helpful for tracking changes in tumor burden, resolving ambiguities in anatomic images caused by non-specific treatment effects and for predicting outcome.

22 citations


01 Jan 2016
TL;DR: In this paper, the authors evaluated the effects of tumor atypia, size, resection extent, and adjuvant radiotherapy on progression-free survival (PFS) of 28 patients with atypical neurocytomas.
Abstract: Central neurocytomas are uncommon intraventricular neoplasms whose optimal management remains controversial due to their rarity. We assessed outcomes for a historical cohort of neurocytoma patients and evaluated effects of tumor atypia, size, resection extent, and adjuvant radiotherapy. Progression-free survival (PFS) was measured by Kaplan-Meier and Cox proportional hazards methods. A total of 28 patients (15 males, 13 females) were treated between 1995 and 2014, with a median age at diagnosis of 26 years (range 5-61). Median follow-up was 62.2 months and 3 patients were lost to follow-up postoperatively. Thirteen patients experienced recurrent/progressive disease and 2-year PFS was 75% (95% CI 53-88%). Two-year PFS was 48% for MIB-1 labeling >4% versus 90% for ≤4% (HR 5.4, CI 2.2-27.8, p = 0.0026). Nine patients (32%) had gross total resections (GTR) and 19 (68%) had subtotal resections (STR). PFS for >80% resection was 83 versus 67% for ≤80% resection (HR 0.67, CI 0.23-2.0, p = 0.47). Three STR patients (16%) received adjuvant radiation which significantly improved overall PFS (p = 0.049). Estimated 5-year PFS was 67% for STR with radiotherapy versus 53% for STR without radiotherapy. Salvage therapy regimens were diverse and resulted in stable disease for 54% of patients and additional progression for 38 %. Two patients with neuropathology-confirmed atypical neurocytomas died at 4.3 and 113.4 months after initial surgery. For central neurocytomas, MIB-1 labeling index >4% is predictive of poorer outcome and our data suggest that adjuvant radiotherapy after STR may improve PFS. Most patients requiring salvage therapy will be stabilized and multiple modalities can be effectively utilized.

22 citations


Journal ArticleDOI
TL;DR: There is a path forward in terms of the molecular and biological characterization of DIPGs that can be used to further refine therapeutic decisions, and biopsy-directed clinical trials that use tissue to assign patients to rational therapeutic arms are under way.
Abstract: Diffuse intrinsic pontine gliomas (DIPGs) are regionally specific gliomas that arise in a relatively narrow age range in children. As opposed to a pontine location, tumors that arise in the medulla or midbrain are often WHO Grade I or II tumors and have a markedly different prognosis than DIPGs. The characteristic location as well as the presence of extensive infiltration in the ventral pons means that resection is not possible. The overall lack of treatment options, and the resulting dismal prognosis associated with a diagnosis of DIPG has historically led to a sense of nihilism among treating physicians. The biological behavior of these tumors has long been thought to best fit with adult WHO Grade IV astrocytoma, and it was also assumed that the molecular features would be similar to those observed in other locations in the brain. These assumptions led to the use of many experimental agents that are used in the context of adult clinical trials for WHO Grade IV astrocytoma. However, this did not result in any meaningful improvement in survival.7 The recent analysis of tissue obtained from a variety of sources by different groups has shown conclusively that primary brain tumors in children have distinct features when compared to adult gliomas.1,4,12,15 Furthermore, the majority of DIPGs have a particularly unique molecular signature, that of mutations in the histone genes, which leads to changes in gene expression that are believed to account for their oncogenesis.3,9,14 In addition, pharmacological alterations in histone methylation appear to result in inhibition of tumor growth, at least in preclinical studies.5,6 The biological advances as well as the surgical techniques that have been present for the last 10–20 years have allowed biopsies to be performed with acceptable morbidity.2,11 In the series published to date, the majority of complications were transient cranial neuropathies, although more serious ones have been reported. Although the potential for surgical morbidity cannot be ignored or minimized, there is a path forward in terms of the molecular and biological characterization of DIPGs that can be used to further refine therapeutic decisions10: specifically, biopsy-directed clinical trials that use tissue to assign patients to rational therapeutic arms are under way. Recently opened trials using advanced sequencing techniques to identify molecular alterations within specific pathways for a patient’s tumor have allowed therapy to be directed against those targets—an example of personalized medicine (clinicaltrial.gov NCT01182350 and NCT02233049). Of course, it is not clear whether this approach casts too broad a net, or whether other factors such as inadequate drug delivery will lead to failure. However, within the broader scope of precision medicine, and perhaps uniquely for children with rare cancers, continued development of rationally designed clinical trials is needed. In an era of rapid acquisition of RNA and DNA deep sequencing information in individual cases, “rational” means using tumor-derived molecular information wisely. Strategies that use genomic data within individual patients are ongoing in the treatment of most cancers, including adult glioma and now, hopefully, in children within all subsets of CNS tumors. Tumor tissue is a requirement to achieve these goals. Resection and/or biopsy of tumors located in many other eloquent regions of the brain such as the thalamus, insula, perisylvian location, and in the spinal cord are considered the standard of care. Whereas most neurosurgeons would consider biopsy of other areas of the brain acceptable, this perspective is generally not applied to patients

12 citations



Journal ArticleDOI
TL;DR: The goal of this trial was to determine the feasibility to make individualized treatment recommendations within at least 35 days of tumor tissue collection, using whole exome and RNA sequencing based upon those alterations.
Abstract: 2031Background: Pts with recurrent glioblastoma have median survival between 4 to 8 months. "Precision-based" therapies have not been used in this patient population. The goal of this trial was to determine the feasibility to make individualized treatment recommendations within at least 35 days of tumor tissue collection, using whole exome and RNA sequencing based upon those alterations. A sample size of at least 15 patients with sufficient tumor RNA and DNA was used to determine feasibility. Pts. had the option of accepting the recommendation or being treated using other treatment strategies. Adult patients with recurrent glioblastoma who were undergoing tumor resection at the time of relapse as part of their standard of care management were eligible. Methods: Tumor tissue was removed from both enhancing and non-enhancing regions of tumor and characterized by whole exome and RNA sequencing. Molecular profiles were done at Translational Genomics Research Institute (TGen), in a CLIA lab, matching genomic a...

Journal Article
TL;DR: The studies provide the pre-clinical rationale for clinical trials of concurrent radiotherapy and BRAF V600E inhibitors and show that concurrent and sequential PLX4720+RT each significantly prolonged survival compared to monotherapies, in the BRAF v600E HGG model.
Abstract: Radiation (RT) is critical to the treatment of high-grade gliomas (HGGs) but cures remain elusive. The BRAF mutation V600E is critical to the pathogenesis of 10–20 % of pediatric gliomas, and a small proportion of adult HGGs. Here we aim to determine whether PLX4720, a specific BRAF V600E inhibitor, enhances the activity of RT in human HGGs in vitro and in vivo. Patient-derived HGG lines harboring wild-type BRAF or BRAF V600E were assessed in vitro to determine IC50 values, cell cycle arrest, apoptosis and senescence and elucidate mechanisms of combinatorial activity. A BRAF V600E HGG intracranial xenograft mouse model was used to evaluate in vivo combinatorial efficacy of PLX4720+RT. Tumors were harvested for immunohistochemistry to quantify cell cycle arrest and apoptosis. RT+PLX4720 exhibited greater anti-tumor effects than either monotherapy in BRAF V600E but not in BRAF WT lines. In vitro studies showed increased Annexin V and decreased S phase cells in BRAF V600E gliomas treated with PLX4720+RT, but no significant changes in β-galactosidase levels. In vivo, concurrent and sequential PLX4720+RT each significantly prolonged survival compared to monotherapies, in the BRAF V600E HGG model. Immunohistochemistry of in vivo tumors demonstrated that PLX4720+RT decreased Ki-67 and phospho-MAPK, and increased γH2AX and p21 compared to control mice. BRAF V600E inhibition enhances radiation-induced cytotoxicity in BRAF V600E-mutated HGGs, in vitro and in vivo, effects likely mediated by apoptosis and cell cycle, but not senescence. These studies provide the pre-clinical rationale for clinical trials of concurrent radiotherapy and BRAF V600E inhibitors.