Assessing Response of High-Grade Gliomas to Immune Checkpoint Inhibitors

Similar documents
Immunotherapy for the Treatment of Brain Metastases

Bevacizumab in Recurrent Glioma: Patterns of Treatment Failure and Implications

Clinical Trials for Adult Brain Tumors - the Imaging Perspective

NCCN Guidelines for Central Nervous System Cancers V Follow-Up on 02/23/18

Bevacizumab: A Controversial Agent Against High-Grade Gliomas

Assessment of Efficacy and Immune Related RECIST criteria

IMMUNOTHERAPY IN THE TREATMENT OF CERVIX CANCER. Linda Mileshkin, Medical Oncologist Peter MacCallum Cancer Centre, Melbourne Australia

THE EFFECTIVE OF BRAIN CANCER AND XAY BETWEEN THEORY AND IMPLEMENTATION. Mustafa Rashid Issa

J Clin Oncol 28: by American Society of Clinical Oncology INTRODUCTION

Immunotherapy for the Treatment of Melanoma. Marlana Orloff, MD Thomas Jefferson University Hospital

Brain Tumors: Radiologic Perspective

Overview: Immunotherapy in CNS Metastases

Brain mets under I.O.

Glioblastoma and CNS tumors

Immunotherapy for Breast Cancer. Aurelio B. Castrellon Medical Oncology Memorial Healthcare System

Society for Immunotherapy of Cancer (SITC) Immunotherapy for the Treatment of Brain Metastases

THE ROLE OF TARGETED THERAPY AND IMMUNOTHERAPY IN THE TREATMENT OF ADVANCED CERVIX CANCER

Immuno-Oncology Applications

IDH and cancer. Clark O, Yen K, Mellinghoff IK. Clin Cancer Res 2016;22:1837 Copyright 2016 American Association for Cancer Research

Checkpoint Regulators Cancer Immunotherapy takes centre stage. Dr Oliver Klein Department of Medical Oncology 02 May 2015

Pioneering vaccines that transform lives.

Incidence of Early Pseudo-progression in a Cohort of Malignant Glioma Patients Treated With Chemoirradiation With Temozolomide

Current Trends in Melanoma Theresa Medina, MD UCD Cutaneous Oncology

Anderson et al. Journal for ImmunoTherapy of Cancer (2017) 5:76 DOI /s x

Weitere Kombinationspartner der Immunotherapie

Facing pseudoprogression after radiotherapy in low grade gliomas

Special Situation: Brain metastases

J Clin Oncol by American Society of Clinical Oncology INTRODUCTION

CheckMate 012: Safety and Efficacy of First Line Nivolumab and Ipilimumab in Advanced Non-Small Cell Lung Cancer

Survival of High Grade Glioma Patients Treated by Three Radiation Schedules with Chemotherapy: A Retrospective Comparative Study

Melanoma: Immune checkpoints

Glioblastoma and CNS tumors

Goals for this Lecture. Case 1. Key Points MRI TECHNIQUES FOR DIFFERENTIAL DIAGNOSIS OF RECURRENT BRAIN LESIONS

Treatment and management of advanced melanoma: Paul B. Chapman, MD Melanoma Clinical Director, Melanoma and Immunotherapeutics Service MSKCC

Unmasking of intracranial metastatic melanoma during ipilimumab/nivolumab therapy: case report and literature review

Enormous effort has been invested in clinical trials for malignant

CANCER IMMUNOTHERAPY. Pocket Guide

Interferon β and temozolomide combination therapy for temozolomide monotherapy refractory malignant gliomas

Figure 4. Mean Frequency of Parent Cell Population Over Clinical Study Time Points. NKT cells. Ki-67 FMO

Checkpoint regulators a new class of cancer immunotherapeutics. Dr Oliver Klein Medical Oncologist ONJCC Austin Health

Conversations in Oncology. November Kerry Hotel Pudong, Shanghai China

NSCLC: immunotherapy as a first-line treatment. Paolo Bironzo Oncologia Polmonare AOU S. Luigi Gonzaga Orbassano (To)

Immunotherapy for Malignant Brain Cancer: Overcoming Immune Suppression to Improve the Clinical Experience

Radiographic Assessment of Response An Overview of RECIST v1.1

The Efficacy of Combination of Pembrolizumab plus Chemotherapy in Locally Advancedd Squamous Cell Carcinoma of Mandible: Report

Immune Checkpoint Inhibitors: The New Breakout Stars in Cancer Treatment

Magnetic Resonance Imaging for Prediction and Assessment of Treatment Response in Bevacizumab-Treated Recurrent Glioblastoma

Imaging for suspected glioma

Neuro-Oncology Advance Access published April 4, 2012

Immunotherapies in melanoma: regulatory perspective. Jorge Camarero (AEMPS)

Ludwig Presence at 2016 ASCO Annual Meeting

La revolución de la inmunoterapia: dónde la posicionamos? Javier Puente, MD, PhD

How Can We Evaluate Response to New Agents? Recent update of imaging response assessment for cancer immunotherapy

Assessment of treatment response in high grade gliomas: RANO criteria usage for an accurate radiology report.

Management of Brain Metastases Sanjiv S. Agarwala, MD

Immunotherapy for NSCLC: Current State of the Art and Future Directions. H. Jack West, MD Swedish Cancer Institute Seattle, Washington, United States

Durable Response Rate in High Grade Glioma: an Emerging Endpoint for Immunotherapeutics. Timothy Cloughesy, MD University of California, Los Angeles

The Impact of T2/FLAIR Evaluation per RANO Criteria on Response Assessment of Recurrent Glioblastoma Patients Treated with Bevacizumab

Presentation by Dr. Thomas Yau on behalf of his co-authors

The following slides are provided as presented by the author during the live educa7onal ac7vity and are intended for reference purposes only.

Pulmonary changes induced by radiotherapy. HRCT findings

IMMUNOTHERAPY IN THE TREATMENT OF CERVIX CANCER

The Immunotherapy of Oncology

Metastasectomy for Melanoma What s the Evidence and When Do We Stop?

Understanding Checkpoint Inhibitors: Approved Agents, Drugs in Development and Combination Strategies. Michael A. Curran, Ph.D.

Index. Note: Page numbers of article titles are in boldface type.

Hypofractionated radiation therapy for glioblastoma

The MRI Appearance of Tumefactive Demyelinating Lesions

Ipilimumab ASCO Data Review and Discussion Webcast. Monday, June 2, 2008

Treatment With Bevacizumab and Irinotecan for Recurrent High-Grade Glial Tumors

Immunotherapy Concept Turned Reality

Radiologic Pathologic Correlation of Intraosseous Lipomas. Tim Propeck 1, Mary Anne Bullard 1, John Lin 1, Kei Doi 2, William Martel 1

National Horizon Scanning Centre. Bevacizumab (Avastin) for glioblastoma multiforme - relapsed. August 2008

Immunotherapy in non-small cell lung cancer

Mind the Gap : Challenges in First-in- Man Evaluation of Immuno-Oncology Drugs

Melanoma: From Chemotherapy to Targeted Therapy and Immunotherapy. What every patient needs to know. James Larkin

Immunotherapy Treatment Developments in Medical Oncology

Priming the Immune System to Kill Cancer and Reverse Tolerance. Dr. Diwakar Davar Assistant Professor, Melanoma and Phase I Therapeutics

Immunoterapia e farmaci innovativi

Role of functional MRI in evaluating intraaxial brain tumors Advances and pitfalls.

Defining pseudoprogression in glioblastoma multiforme

National Institute for Health and Clinical Excellence. Single Technology Appraisal (STA)

Response Assessment Criteria for Glioblastoma: Practical Adaptation and Implementation in Clinical Trials of Antiangiogenic Therapy

UPDATES ON CHEMOTHERAPY FOR LOW GRADE GLIOMAS

MALIGNANT GLIOMAS: TREATMENT AND CHALLENGES

New Systemic Therapies in Advanced Melanoma

Prostate cancer Management of metastatic castration sensitive cancer

Precision medicine for gliomas

Post-ASCO Immunotherapy Highlights (Part 2): Biomarkers for Immunotherapy

ONCOS-102 in melanoma Dr. Alexander Shoushtari. 4. ONCOS-102 in mesothelioma 5. Summary & closing

Bevacizumab and dose-intense temozolomide in recurrent high-grade glioma

Clinical Management Protocol Chemotherapy [Glioblastoma Multiforme (CNS)] Protocol for Planning and Treatment

MRI perfusion in determining pseudoprogression in patients with glioblastoma

University of Colorado Cancer Center Brain Disease Site Schema

Clinical Policy: Electric Tumor Treating Fields (Optune) Reference Number: PA.CP.MP.145

Citation Pediatrics international (2015), 57.

Going Past the Data for Temozolomide. J. Lee Villano, M.D., Ph.D., Nathalie Letarte, B.Pharm, M.Sc, Linda R. Bressler, Pharm. D.

Metastatic NSCLC: Expanding Role of Immunotherapy. Evan W. Alley, MD, PhD Abramson Cancer Center at Penn Presbyterian

Treating Multiple. Brain Metastases (BM)

THE FUTURE OF IMMUNOTHERAPY IN COLORECTAL CANCER. Prof. Dr. Hans Prenen, MD, PhD Oncology Department University Hospital Antwerp, Belgium

Transcription:

linicians may encounter unique challenges when evaluating the imaging response of GMs treated with immunotherapy and checkpoint inhibitors. Irena Orlov. Industrial feel 21. igital on canvas, 40" 60". ssessing Response of High-Grade Gliomas to Immune heckpoint Inhibitors Solmaz Sahebjam, M, exter G. Stallworth, M, Sepideh Mokhtari, M, Nam. Tran, M, Ph, and John. rrington, M ackground: Immunotherapeutic agents, especially checkpoint inhibitors, have emerged as the mainstay of therapy for several solid and hematological malignancies. These therapies are under investigation for the treatment of high-grade gliomas and brain metastases. Methods: This article reviews the unique challenges encountered when evaluating changes on magnetic resonance imaging (MRI) of glioblastomas seen in response to immunotherapy and checkpoint inhibitors and how to effectively incorporate MRI findings into the response assessment of high-grade gliomas to these emerging therapies. Results: n increase in tumor size or the appearance of new lesions on MRI may represent either an immune-mediated inflammatory response or true tumor progression, which may precede the subsequent stabilization or response of high-grade gliomas to immunotherapy. These MRI findings should not result in the mandatory cessation of immunotherapy in patients with high-grade glioma. onclusions: lthough immunotherapy Response ssessment for Neuro-Oncology criteria have been developed to assist with response assessment of high-grade gliomas to immunotherapy and to provide guidance with treatment decisions, these criteria have not been validated in prospective clinical trials. In patients with brain tumors who are receiving immunotherapy, MRI findings suggestive of disease progression should be evaluated with caution to prevent premature discontinuation of potentially beneficial therapies. lose, clinical monitoring with appropriate short-term, follow-up imaging is often necessary, and histopathological analysis may be required in some cases to confirm disease progression before a decision on continuation of these novel therapies can accurately be made. From the epartment of Neuro-Oncology (SS, SM, NT) and iagnostic Imaging and Interventional Radiology Program (GS, J), H. Lee Moffitt ancer enter & Research Institute, and the epartment of Oncologic Sciences (SS, GS, SM, NT, J), University of South Florida ollege of Medicine, Tampa, Florida. Submitted February 21, 2017; accepted March 7, 2017. ddress correspondence to Solmaz Sahebjam, M, epartment of Neuro-Oncology, Moffitt ancer enter, 12902 Magnolia rive, W-NEURO PROG and M-RU, Tampa, FL 33612. E-mail: Solmaz.Sahebjam@Moffitt.org No significant relationships exist between the authors and the companies/organizations whose products or services may be referenced in this article. Introduction Immune checkpoint inhibitors have emerged as a mainstay of treatment for a variety of advanced malignancies. ntibodies against cytotoxic T-lymphocyteassociated protein 4, programmed death 1 (P-1), and its ligand (P-L1) have demonstrated significant clinical benefit in several solid and hematological malignancies, and their list of approved indications is expanding. nti P-1/P-L1 antibodies alone or in combination with ipilimumab, an anti cytotoxic T-lymphocyte-associated protein 4 antibody, is being investigated in patients with high-grade gliomas. 1-3 These 180 ancer ontrol pril 2017, Vol. 24, No. 2

therapies are well tolerated in patients with newly diagnosed or recurrent glioblastoma (GM). 1-3 lthough the clinical benefit of these antibodies in the setting of high-grade glioma is yet to be confirmed, the preliminary data from small cohorts suggest an antitumor effect and clinical benefit when these agents are used as single-agent or combination therapy. 1,3,4 ssessing Radiographical Findings Initial increases in tumor size or the appearance of new lesions can precede the subsequent response of solid tumors to immunotherapeutic agents. 5 Premature discontinuation of immunotherapy based on the initial increase in tumor burden can deprive patients of potentially beneficial treatments. To address this challenge, new guidelines for the evaluation of response to immunotherapeutic agents in solid tumors have been developed. Immune-related Response riteria allows continuation of treatment with appropriate follow-up time points to confirm disease progression. 5 Similar imaging findings have been observed in patients with high-grade glioma who have received immunotherapeutic agents such as immune checkpoint inhibitors. 6 The appearance of a new lesion or the progression of existing lesions following the initiation of immunotherapy can represent either an immune-mediated inflammatory response or true tumor progression. It is often difficult to differentiate tumor growth from treatment changes on imaging within the first 6 months after the initiation of immunotherapy. 6 n effective immune response may take time to develop, and brain tumors may initially progress after the initiation of immunotherapy. Therefore, an increase in tumor burden does not necessarily preclude subsequent clinical benefit, and delayed effective, immune response may result in subsequent stabilization or regression of the tumor. ifferentiating high-grade gliomas that initially progress after immunotherapy but will subsequently respond from those tumors that will not respond is a critical challenge. Furthermore, in some cases, the appearance of new lesions or the increase in size of the enhancing lesions can be the result of immunerelated responses rather than tumor growth. 6,7 Hence, response criteria specific for GMs treated with immunotherapies, such as checkpoint inhibitors, is critical to prevent the premature termination of these therapies. To accurately assess the imaging changes of high-grade glial tumors and GMs to immunotherapy, a historical perspective and an understanding of the evolution of imaging response assessment in neuro-oncology is helpful. Macdonald, Response Evaluation riteria In Solid Tumors, and World Health Organization criteria assess the imaging response of brain tumors to the direct antitumoral effect of cytotoxic therapy with either single or bidimensional measurements of the contrast-enhancing component of the tumor. 8-10 Macdonald criteria were developed for computed tomography prior to the advent of magnetic resonance imaging, which is now standard of care for evaluating the imaging response of high-grade gliomas to therapy. 8 Macdonald criteria incorporate the changes in bidimensional tumor measurements with clinical assessment and steroid dose for the response assessment of high-grade gliomas such as GM to therapy. 8 idimensional measurements of the contrast-enhancing component of a GM is not always an accurate assessment of the overall extent of the tumor, and measurable change in the contrast-enhancing portion of a GM does not always represent an accurate assessment of response to therapy. Rather than being an accurate surrogate of GM response to therapy, change in contrast enhancement reflects changes in the vascular permeability of the brain and tumor to contrast agents and can be seen with nontumoral, treatment-related changes such as ischemia, inflammation, acute and delayed radiation effects, and tumor growth. Now that novel agents with different mechanisms of actions are being investigated, the response assessment of imaging changes is more complex because of treatment-related imaging findings that mimic tumor growth (pseudoprogression) and treatment-related imaging findings that mimic tumor response (pseudoresponse). Limiting the imaging response assessment of GMs to bidimensional measurements of enhancing tumors and not incorporating the assessment of the nonenhancing portion of GMs is a significant limitation of the Macdonald criteria. To adequately address the unique imaging challenges of GM and provide guidance in distinguishing tumor growth from treatment-related responses, changes in response-assessment criteria were necessary. Response ssessment in Neuro-Oncology (RNO) criteria published in 2010 were a collaborative effort to update and standardize response criteria for high-grade gliomas and replace the Macdonald criteria. 9 RNO imaging criteria incorporate fluid-attenuated inversion recovery (FLIR) with the bidimensional measurements of the enhancing portion of the GM obtained on contrast-enhanced, T1-weighted imaging. 9 y incorporating FLIR signal changes into the imaging response assessment of GM, RNO criteria allow the assessment of the nonenhancing component of the GM as well as associated vasogenic edema and help distinguish treatment-related changes from true tumoral changes. RNO criteria give specific clinical and imaging findings required for the diagnosis of disease progression. Imaging findings that meet RNO criteria for tumor progression include 9 : evelopment of new lesions outside the radiation field 25% increase in the sum of perpendicular diameters of enhancing lesions Substantial worsening of T2-weighted FLIR signal changes ritical to the RNO criteria is the guidance in assessing treatment-related imaging changes and distinguishing these changes from tumor growth or tumor pril 2017, Vol. 24, No. 2 ancer ontrol 181

regression. hanges in bidimensional measurements of the enhancing component of a GM can reflect nontumoral changes or treatment-related effects and do not always accurately assess response to therapy. Pseudoprogression represents imaging changes related to treatment effects and not true tumor progression. The imaging changes of pseudoprogression include increasing measurable enhancement of a lesion associated with increasing FLIR signal changes and can be seen in patients with GM in the first 3 months following postoperative radiotherapy and temozolomide (Fig 1). RNO criteria require that progressive disease not be diagnosed within 3 months of the completion of therapy with radiation and temozolomide unless there is new enhancement outside of the radiation field or the patient has clinically declined. lthough RNO criteria require the cessation of therapy when there is either clinical or imaging evidence of progressive disease, they do allow continuation of therapy in patients who demonstrate progressive imaging findings of an unclear etiology. Temozolomide should not be stopped in these patients until progressive changes are confirmed on follow-up imaging and as long as the patient remains clinically stable. Pseudoresponse, seen as decreasing le- Fig 1. Progression vs pseudoprogression. (, ) xial fluid-attenuated inversion recovery and (, ) contrast-enhanced, T1-weighted images at baseline (, ) and initial post-treatment examination obtained 6 weeks after completion of postoperative radiotherapy and concurrent temozolomide therapy (, ). Post-treatment examination demonstrated an interval increase in enhancement (green arrows) and vasogenic edema (red arrows), which could represent either tumor growth or treatment-related changes. Response ssessment in Neuro-Oncology criteria allow for the continuation of temozolomide until findings of progression are confirmed on follow-up magnetic resonance imaging. If the imaging changes stabilize or decrease on surveillance examinations, then the diagnosis of treatment-related changes is confirmed. 182 ancer ontrol pril 2017, Vol. 24, No. 2

sion enhancement associated with increasing FLIR signal changes, represents tumor growth that mimics the response of GM to therapy. This can be seen in patients receiving antiangiogenic therapy, including therapy with bevacizumab. Tumor progression can be diagnosed per RNO criteria in these patients by the substantial worsening of FLIR signal changes, even if there is stable or decreasing enhancement (Fig 2). New and additional challenges are encountered when evaluating the imaging changes of high-grade gliomas in response to immunotherapy. orrectly assessing new or progressive lesions that occur during immunotherapy is critical, because early, progressive imaging findings may not preclude subsequent response to therapy and overall benefit. hanges in contrast enhancement and FLIR signal remain the cornerstone of the imaging response assessment for GM and high-grade glial tumors to immunotherapy. When GMs demonstrate a decrease in measurable enhancement and a decrease in FLIR signal changes after the initiation of immunotherapy, the diagnosis of positive response to therapy can confidently be made. When GMs demonstrate an increase in measurable enhancement confirmed on subsequent follow-up imaging, the diagnosis of disease progression can also be made (Fig 3). lthough advanced imaging Fig 2. Pseudoresponse. (, ) xial FLIR and (, ) contrast-enhanced, T1-weighted images obtained at baseline (, ) and after initiation of antiangiogenic therapy (, ). (, ) Post-treatment imaging demonstrates a significant increase in FLIR signal changes (green arrows), with the areas of enhancement (red arrows) either remaining stable or decreasing. The substantial increase seen in FLIR signal changes allows the diagnosis of disease progression to be made without progression of the enhancing tumor. FLIR = fluid-attenuated inversion recovery. pril 2017, Vol. 24, No. 2 ancer ontrol 183

Fig 3. Response to immunotherapy in combination with bevacizumab followed by progression. () xial contrast-enhanced, T1-weighted images at baseline and surveillance examinations obtained at 6-week intervals following initiation of pembrolizumab and bevacizumab ( ). () The 6-week, post-treatment imaging demonstrates an interval decrease in enhancement of the tumor following therapy. decrease was observed in the enhancing tumor in both the superficial (red arrows) and deep anterior tumor margins (green arrows). () The 12-week, post-treatment imaging demonstrates increasing enhancement along the deep anterior tumor margin (green arrows) that continued to progress on () 18-week, post-treatment imaging, thus documenting disease progression. The superficial portion of the glioblastoma remained stable and did not progress (red arrows). E F Fig 4 F. Response to immunotherapy in combination with bevacizumab. (, ) xial perfusion cerebral blood volume, (, E) contrast-enhanced, T1-weighted, and (, F) fluid-attenuated inversion recovery images are shown at baseline ( ) and at 6 weeks following initiation of immunotherapy and bevacizumab ( F). Post-treatment examination demonstrates an interval response to therapy with a decrease in perfusion (red arrows) and enhancement (green arrows) of the glioblastoma as well as a decrease in associated vasogenic edema (yellow arrows). 184 ancer ontrol pril 2017, Vol. 24, No. 2

techniques, including perfusion imaging, are not part of the formal RNO or immunotherapy Response ssessment in Neuro-Oncology (irno) criteria for the response assessment of GMs to immunotherapy, they can be helpful in distinguishing treatment effects from tumor growth. Perfusion imaging can estimate and evaluate changes in cerebral blood volume (V) and cerebral blood flow in tumors and can be helpful in distinguishing progressive enhancement related to tumor growth from progressive enhancement resulting from treatment effects. Enhancement related to tumor growth typically demonstrates increased perfusion and elevated V, whereas enhancement related to treatment-related changes or necrosis usually demonstrates decreased perfusion and decreased V. GMs treated with immunotherapy in combination with bevacizumab will typically demonstrate an interval decrease in measurable enhancement and FLIR signal changes, as well as decreased V on perfusion imaging (Fig 4). lthough RNO criteria allow for the continuation of therapy in patients with progressive imaging changes of unclear etiology, they do not permit the continuation of treatment after there is imaging evidence of disease progression. To allow patients with initial progression of imaging findings after the initiation of immunotherapy who meet the RNO criteria of progression of disease to continue therapy and potentially receive delayed or long-term clinical benefit, new guidelines and the irno criteria were developed. 6 irno criteria integrate the immune-related Response riteria concept of confirmation of radiographical progression with the RNO guidelines, defining complete response, partial response, stable disease, and progressive disease, and they allow the continuation of immunotherapy, such as immune checkpoint inhibitors, in patients who show early imaging progression within the first 6 months of initiating therapy until progression is confirmed on subsequent imaging and as long as the patient remains clinically stable (Fig 5). 6 The progressive imaging findings are expected to stabilize or improve within 3 months in patients who will E F Fig 5 F. Initial progression of imaging findings following initiation of immunotherapy. (, ) xial fluid-attenuated inversion recovery, (, E) contrast-enhanced, T1-weighted, and (, F) sagittal contrast-enhanced, T1-weighted (, F) images at baseline ( ) and 6 weeks following initiation of immunotherapy ( F). Post-treatment magnetic resonance imaging obtained 6 weeks following the initiation of immunotherapy demonstrated an interval increase in the enhancing lesion and associated vasogenic edema. y Response ssessment in Neuro-Oncology criteria, there is imaging evidence of disease progression, and therapy would have to be terminated. However, because imaging was obtained within 6 months of the initiation of immunotherapy, immunotherapy Response ssessment in Neuro-Oncology criteria allow therapy to continue until progression is confirmed on follow-up examination. pril 2017, Vol. 24, No. 2 ancer ontrol 185

receive long-term clinical benefit. To clarify whether the progressive imaging findings represent tumor progression or treatment-related changes, follow-up imaging is typically obtained 3 months after the initial imaging evidence of progressive disease. It is noteworthy that irno criteria have yet to be validated in prospective clinical trials and clinical cohorts. onclusions Response assessment of patients with glioblastomas treated with checkpoint inhibitors is challenging. lthough immune-related Response riteria and Response ssessment in Neuro-Oncology criteria were developed to assist with response assessment to immunotherapy and provide guidance with treatment decisions, Response ssessment in Neuro-Oncology criteria have not been validated in prospective clinical trials in assessing the response of high-grade gliomas treated with checkpoint inhibitors. lose clinical monitoring, short-term imaging follow up, and, in some cases, histopathological analysis may be required to prevent the premature discontinuation of potentially beneficial therapies. References 1. Reardon, Sampson JH, Sahebjam S, et al. Safety and activity of nivolumab (nivo) monotherapy and nivo in combination with ipilimumab (ipi) in recurrent glioblastoma (GM): updated results from checkmate-143. Presented at: merican Society of linical Oncology nnual Meeting; June 3 7, 2016; hicago, Illinois. bstract 2014. Poster 203. 2. Omuro, Gordana V, aehring J, et al. Nivolumab combined with radiotherapy with or without temozolomide in patients with newly diagnosed glioblastoma: results from phase 1 safety cohorts in heckmate 143. Presented at: 21st nnual Scientific Meeting and Education ay of the Society for Neuro-Oncology; November 17 20, 2016; Scottsdale, rizona. bstract TIM-16. 3. Sahebjam S, Johnstone P, Forsyth P, et al. Safety and antitumor activity of hypofractionated stereotactic irradiation (HFSRT) with pembrolizumab (Pembro) and bevacizumab (ev) in patients (pts) with recurrent high grade gliomas: preliminary results from phase I study. Presented at: merican Society of linical Oncology nnual Meeting; June 3 7, 2016; hicago, Illinois. bstract 2041. Poster 229. 4. Reardon, Kim T-M, Frenel J-S, et al. Results of the phase Ib Keynote-028 multi-cohort trial of pembrolizumab monotherapy in patients with recurrent P-L1-positive glioblastoma multiforme (GM). Presented at: 21st nnual Scientific Meeting and Education ay of the Society for Neuro-Oncology; November 17 20, 2016; Scottsdale, rizona. bstract TIM-35. 5. Wolchok J, Hoos, O ay S, et al. Guidelines for the evaluation of immune therapy activity in solid tumors: immune-related response criteria. lin ancer Res. 2009;15(23):7412-7420. 6. Okada H, Weller M, Huang R, et al. Immunotherapy response assessment in neuro-oncology: a report of the RNO working group. Lancet Oncol. 2015;16(15):e534-e542. 7. Sahebjam S, Omuro, aehring J, et al. Pseudoprogression and immunotherapy in glioblastoma: a case series from cohort 1 of HEK- MTE-143 (NT02017717). Presented at: 20th nnual Scientific Meeting and Education ay of the Society for Neuro-Oncology; November 19 22, 2015; San ntonio, Texas. bstract NIMG-16. 8. Macdonald R, ascino TL, Schold S Jr, et al. Response criteria for phase II studies of supratentorial malignant glioma. J lin Oncol. 1990;8(7):1277-1280. 9. Wen PY, Macdonald R, Reardon, et al. Updated response assessment criteria for high-grade gliomas: response assessment in neurooncology working group. J lin Oncol. 2010;28(11):1963-1972. 10. Miller, Hoogstraten, Staquet M, et al. Reporting results of cancer treatment. ancer. 1981;47(1):207-214. 186 ancer ontrol pril 2017, Vol. 24, No. 2