scholarly journals Stereotactic radiosurgery combined with immune checkpoint inhibition for the treatment of melanoma brain metastases is associated with high levels of extracranial disease control and survivorship - an abscopal effect?

2021 ◽  
Vol 23 (Supplement_4) ◽  
pp. iv16-iv16
Author(s):  
Philip Webb ◽  
Mark Zorman ◽  
Rhona Watson ◽  
Gemma Austin ◽  
Carol Thurgood ◽  
...  

Abstract Aims Melanoma brain metastases (MBM) are a common presentation to the neuro-oncology MDT. Stereotactic radiosurgery (SRS) is a highly effective treatment for cerebral metastases, with at least 70% control rates of individual metastases,[1] whilst immune checkpoint blockade has revolutionised the management of metastatic melanoma in recent years.[2] Recent studies have demonstrated that immune checkpoint inhibition alone also has activity in the brain, with MBM response rates of 50% or more.[3, 4] When MBM are treated with combination immunotherapy and SRS together, 12-month intracranial progression free survival (PFS) rates of 85% have been achieved.[4, 5] The aim of the current study was to evaluate the local control of MBM treated at our tertiary referral centre, which benefits from specialist neuro-radiology peer review of SRS contour volumes, and further to investigate whether overall survival is also improved, and what the mechanism of this may be. Method A retrospective analysis of all patients treated with SRS for brain metastases at our teriary SRS centre between June 2017 – January 2020 was performed. Inclusion criteria included patients treated for MBM, who received at least 2 doses of any combination of immune checkpoint inhibition concurrently with (defined as at the time of or commenced within 3 months of) SRS. The primary endpoints were the intracranial and extracranial response rates and survival rate at 12 months. Response was defined as complete response, partial response or stable disease. Secondary endpoints included the rate of imaging-defined radionecrosis, median lesional progression free survival (mPFSlesion), non-lesional intracranial PFS (mPFSintracranial), extracranial PFS (mPFSextracranial) and overall survival (mOS), measured from the start date of SRS to the date of event or censored at the start date of data collection. Kaplan-Meier curves and survival statistics were generated using SPSS v26. Results 33 MBM from 18 patients were identified. The median follow up was 25.8 months (minimum 12 months). Of the 18 patients: the median age was 60 (IQR 48 – 72); 17 (94%) patients were ECOG performance status 0-1; the median number of extracranial disease sites was 2 (pre-immunotherapy) and 1 (pre-SRS); the median duration of immunotherapy treatment was 17.6 (12.9 – 28.5) months, and the median number of metastases treated per patient was 2. Of the 33 metastases: 31 (94%) were supratentorial; 6 (18%) underwent prior neurosurgical resection; the median GTV volume (cc) of unresected metastases was 0.5cc (0.1 – 2.7), and 21 (64%) were treated with single fraction SRS. The median OS and PFS for all subtypes were not reached. The rates of OS, PFSlesion, PFSintracranial and PFSextracranial at 12 months were 93.9%, 87.9%, 81.8% & 75.8% respectively. Conclusion Our cohort of MBM patients appear to perform favourably when compared with the current literature. When compared to a recent extensive systematic review of modern management of MBM, our lesional control rate is as good as the weighted average of concurrent SRS + immunotherapy studies (87.9% vs 85.4% 12-month PFS), however we demonstrate a significantly improved 12-month OS rate (93.9% vs 52.8%) compared to the same (mOS of 15.8 – 17.4 months in other studies).[6,7] Our extra-lesional PFS is high and, compared to extracranial PFS rates from 51% at 6-months to 70.4% at 9-months in the literature,[3,4] our 75.8% control at 12 months suggests that extracranial control could drive the OS benefit. This suggests a benefit of SRS beyond the local control of MBM and questions whether patients without brain metastases may benefit from body SABR to extracranial metastases, to elicit a similar, potentially abscopal type effect.

2019 ◽  
Vol 29 (5) ◽  
pp. 516-521 ◽  
Author(s):  
Victoria Grätz ◽  
Ewan A. Langan ◽  
Alexander Neumann ◽  
Detlef Zillikens ◽  
Patrick Terheyden

2021 ◽  
Vol 39 (15_suppl) ◽  
pp. e20557-e20557
Author(s):  
Julian Taugner ◽  
Monika Karin ◽  
Lukas Käsmann ◽  
Chukwuka Eze ◽  
Julian Guggenberger ◽  
...  

e20557 Background: The present study evaluates outcome after chemoradiotherapy (CRT) with concurrent and/or sequential Programmed Cell Death 1 (PD-1) or Ligand 1 (PD-L1) immune checkpoint inhibition (CPI) for inoperable stage III NSCLC patients depending on planning target volume (PTV). Methods: Prospective data of thirty-nine consecutive patients with inoperable stage III NSCLC who completed CRT with sequential durvalumab (72%, 28 patients) or concurrent and sequential nivolumab (28%, 11 patients) were analyzed. Different cut offs for PTV as well as PTV as a continuous variable were evaluated for association with progression-free survival (PFS) and extracranial metastasis-free survival (eMFS). Results: All patients were treated with conventionally fractionated TRT to a total dose of at least 60 Gy (range: 60-63.6Gy), 97% (27 patients) received two cycles of concurrent platinum-based chemotherapy. Median follow-up for the entire cohort was 23.2 (range: 6.0-42.6) months; median overall survival (OS) and eMFS were not reached. Median Progression-free survival (PFS) was 22.8 (95% CI: 10.3-35.2) months. Age (65 years), gender and UICC stage had no significant impact on PFS. There was no significant difference between durvalumab and nivolumab patients. Patients with PTV ≥ 900ccm had a significantly shorter PFS (11.77 vs 26.3 months, p = 0.049) and eMFS (11.7 months vs not reached, p = 0.019). Furthermore, patients with PTV ≥ 900ccm and stage IIIC disease (TNM 8th Ed.) achieved a dismal median PFS of only 3.6 months (vs. 26.3 months p < 0.001). PTV as a continuous variable showed a trend for association with PFS (p = 0.064) and was a significant negative prognosticator for eMFS (p = 0.030; HR: 4.065; 95%CI: 1.148-14.397). Conclusions: PTV has a significant impact on the PFS and eMFS after CRT combined with concurrent and/or sequential CPI in inoperable stage III NSCLC. Patients with PTV ≥ 900ccm had a significantly shorter PFS and eMFS.


2018 ◽  
Vol 36 (15_suppl) ◽  
pp. e14009-e14009
Author(s):  
Eric J Lehrer ◽  
Jennifer Layton Peterson ◽  
Paul D. Brown ◽  
Jason Sheehan ◽  
Alfredo Quinones-Hinojosa ◽  
...  

2020 ◽  
Vol 38 (4_suppl) ◽  
pp. 117-117
Author(s):  
M. Cecilia Monge B. ◽  
Changqing Xie ◽  
Seth M. Steinberg ◽  
Suzanne Fioraventi ◽  
Melissa Walker ◽  
...  

117 Background: The benefit of immune checkpoint inhibition is limited to the small percentage of advanced colorectal cancer (CRC) patients whose tumors present mismatch repair (MMR) gene abnormalities; immunotherapy has not shown benefit in patients with MMR proficient CRC. Oncolytic immunotherapy represents a unique therapeutic platform. This phase I trial tests the safety of the combination of pexastimogene devacirepvec (Pexa-Vec) plus durvalumab (durva) in patients with locally advanced or metastatic CRC. Methods: Eligible patients with advanced proficient mixed match repair (pMMR) CRC received intravenous infusion of Pexa-Vec at dose level 3 x 108 plaque forming units (pfu) (DL1) or at 109 pfu (DL2) every 2 weeks for 4 doses and durva 1500 mg every 28 days. Response was assessed with CT every 8 weeks. Adverse events were recorded and managed. The primary endpoint included safety, tolerability and feasibility of this combination therapy. Samples of tumor and peripheral blood were collected for assessment of immune parameters. Results: Sixteen patients (6 males and 10 females) enrolled with a median age of 52.1 years (range 39-69) from Dec, 2017 to Oct, 2018. Four patients were treated with Pexa-Vec at DL1 and durva;twelve patients were treated with Pexa-Vec at DL2 and durva.The most common treatment related adverse events (TRAE) included fever 15/16 (94 %), hypotension 12/16 (75 %), chills 12/16 (75%), fatigue 8/16 (50%), sinus tachycardia 7/16 (44%) and rash 6/16 (38%). Grade 3/4 TRAEs were reported in 8/16 (50%)patients; the most common were fever 7/16 (44 %), lymphopenia 2/16 (13%), neutropenia 1/16 (6%) and anemia 1/16 (6%). 14 patients were evaluable for response analysis; one patient 1/14 (7 %) achieved a confirmed partial response (lasting 7.1 months) and continues to receive treatment, while 13 patients had progressive disease. The median progression free survival (PFS) was 2.2 months (95% CI: 2.2-2.3 months) and the median overall survival (OS) was 7.5 months (CI: 4.9-10.1 months). Conclusions: Combination therapy of Pexa-Vec with durva ICI issafe, well tolerated and demonstrates possible activity in patients with advanced pMMR CRC. Clinical trial information: NCT03206073.


2020 ◽  
Vol 38 (15_suppl) ◽  
pp. 9577-9577
Author(s):  
Joao Victor Machado Alessi ◽  
Carolyn Glass ◽  
Biagio Ricciuti ◽  
Liam F. Spurr ◽  
Lynette M. Sholl ◽  
...  

9577 Background: The catalytic unit of the SWI/SNF chromatin remodeling complex is encoded by the SMARCA4 gene, which is mutated in ~10% of NSCLCs. We sought to characterize the clinicopathologic characteristics and outcomes to immune checkpoint inhibition in SMARCA4-mutant NSCLC. Methods: We collected clinicopathologic and genomic data from patients with NSCLC that had undergone targeted next generation sequencing (NGS) by OncoPanel at the Dana-Farber Cancer Institute. SMARCA4 frameshift, nonsense, and splice-site mutations were considered pathogenic, as were missense mutations if predicted to be pathogenic by Mutation Assessor and Polyphen-2. Clinical outcomes to immune checkpoint inhibition among SMARCA4-mutant NSCLCs were retrospectively assessed. Results: Of 2690 patients with NSCLC, 8% (N = 211) harbored SMARCA4 mutations. Clinicopathological characteristics were balanced between SMARCA4 mut and SMARCA4 wild-type (wt) in terms of age, histology, and PD-L1 expression. We observed a male predominance (P = 0.03), greater use of tobacco (P < 0.001), a higher tumor mutational burden (TMB) (P < 0.001), a higher prevalence of advanced disease (P < 0.001), and a lower prevalence of concurrent targetable driver mutations (P < 0.001) in SMARCA4mut vs SMARCA4wt NSCLCs. Among 513 patients with nonsquamous NSCLC who received immune checkpoint inhibitors, 11% (N = 57) harbored SMARCA4 mutations. From the start of immunotherapy, we observed no difference in overall response rate (ORR 21.5% vs 19.3%; P = 0.3), median progression free survival (mPFS 3.2 months vs 2.1 months; P = 0.4), or median overall survival (mOS 12.0 months vs 8.2 months; P = 0.09) in SMARCA4wt vs SMARCA4mut NSCLC, respectively. However, among KRASmut NSCLC, a concurrent SMARCA4 mut conferred a significantly lower ORR (23.1% vs 0.0%; P = 0.02), a significantly shorter mPFS (4.8 months vs 1.7 month; HR: 0.31 [95% CI: 0.15-0.61]; P < 0.001), and a significantly shorter mOS (15.6 months vs 2.7 months; HR: 0.25 [95%CI: 0.12-0.49]; P < 0.001). The deleterious effect of SMARCA4 mut on immunotherapy outcomes in KRAS mut NSCLC was maintained when controlling for concurrent STK11 mut. Conclusions: SMARCA4 mutations define a genomic subset of NSCLC with unique clinicopathologic characteristics, and confer worse outcomes to immunotherapy in KRAS mut NSCLC.


2021 ◽  
Vol 3 (Supplement_5) ◽  
pp. v43-v51
Author(s):  
Jianbo Wang ◽  
Hussein A Tawbi

Abstract Brain metastases from solid tumors are increasing in incidence, especially as outcomes of systemic therapies continue to extend patients’ overall survival. The long-held notion that the brain is an immune sanctuary has now been largely refuted with increasing evidence that immunotherapy can induce durable responses in brain metastases. Single agent immune checkpoint inhibition with anti-CTLA4 and anti-PD1 antibodies induces durable responses in 15%–20% in melanoma brain metastases as long as patients are asymptomatic and do not require corticosteroids. The combination of anti-CTLA4 with anti-PD-1 antibodies induces an intracranial response in over 50% of asymptomatic melanoma patients, and much lower rate of otherwise durable responses (20%) in symptomatic patients or those on steroids. Data in other cancers, such as renal cell carcinoma, are accumulating indicating a role for immunotherapy. Emerging immunotherapy approaches will have to focus on increasing response rates, decreasing toxicity, and decreasing steroid dependency. The path to those advances will have to include a better understanding of the mechanisms of response and resistance to immunotherapy in brain metastases, the use of novel agents such as anti-LAG3 checkpoint inhibitors, targeted therapy (oncogene directed or TKIs), and possibly surgery and SRS to improve the outcomes of patients with brain metastases.


Sign in / Sign up

Export Citation Format

Share Document