Intraoperative 32P High-Dose Rate Brachytherapy of the Dura for Recurrent Primary and Metastatic Intracranial and Spinal Tumors

Neurosurgery ◽  
2012 ◽  
Vol 71 (5) ◽  
pp. 1003-1011 ◽  
Author(s):  
Michael R. Folkert ◽  
Mark H. Bilsky ◽  
Gil'ad N. Cohen ◽  
Marco Zaider ◽  
Lawrence T. Dauer ◽  
...  

Abstract BACKGROUND: Treatment of spinal and intracranial tumors with dural involvement is complicated by radiation tolerance of sensitive structures, especially in the setting of previous treatment. OBJECTIVE: To evaluate whether intraoperative brachytherapy with short-range sources allows therapeutic dose delivery without damaging sensitive structures. METHODS: The median doses of previous treatment were 3000 cGy (range, 1800-7200 cGy) for 8 patients with primary/recurrent and 17 patients with metastatic spinal tumors and 5040 cGy (range, 1300-6040 cGy) for 5 patients with locally recurrent and 2 patients with metastatic intracranial tumors. Patients underwent gross total or maximal resection of the tumor and were then treated with an intraoperative brachytherapy plaque consisting of a flexible silicone film incorporating 32P. A dose of 1000 cGy was delivered to a depth of 1 mm; the percent depth dose was less than 1% at 4 mm from the prescription depth. Median postoperative radiation doses of 2700 cGy (range, 1800-3000 cGy) were delivered to 15 spinal tumor patients and 3000 cGy (range, 1800-3000 cGy) to 3 intracranial tumor patients. The median follow-up was 4.4 months (range, 2.6-23.3 months) for spinal tumor patients and 5.3 months (range, 0.7-16.2) for intracranial tumor patients. RESULTS: At 6-month follow-up, for all spinal tumor patients, local progression-free survival and overall survival rates were both 83.3% (95% confidence interval [CI]: 62.3%-94.3%); for all intracranial tumor patients, the local progression-free survival rate was 62.5% (95% CI: 23.8%-90.9%) and the overall survival rate was 66.7% (95% CI: 26.7%-92.9%). There were no intraoperative or postoperative complications secondary to radiotherapy. CONCLUSION: Use of the 32P brachytherapy plaque is technically simple and not associated with increased risk of complications, even after multiple radiation courses. Local control rates were more than 80% in patients with proven radiation-resistant spinal disease.

Blood ◽  
2016 ◽  
Vol 128 (22) ◽  
pp. 489-489 ◽  
Author(s):  
Philippe Moreau ◽  
Jonathan L. Kaufman ◽  
Heather J. Sutherland ◽  
Marc Lalancette ◽  
Hila Magen ◽  
...  

Abstract Introduction: Daratumumab is an anti-CD38 IgGκ monoclonal antibody that has been combined successfully with lenalidomide and dexamethasone. The combination of daratumumab with lenalidomide and dexamethasone (DRd) has been compared with lenalidomide and dexamethasone alone (Rd) in patients (pts) with relapsed or refractory multiple myeloma (RRMM) in a randomized phase 3 study (Dimopoulos MA, et al. N Engl J Med 2016; in press). In a pre-specified interim analysis, the DRd combination demonstrated significantly longer progression-free survival (PFS) in addition to deep and durable responses compared with the Rd arm. We performed subgroup analyses to further examine these efficacy data according to prior treatment exposure. Methods: Pts who received ≥1 prior line of therapy were randomized (1:1) to Rd (lenalidomide: 25 mg PO on Days 1-21 of each 28-day cycle; dexamethasone: 40 mg PO weekly) with or without daratumumab (16 mg/kg IV qw for 8 weeks, q2w for 16 weeks, then q4w until progression). The primary endpoint was PFS. Pts who were refractory to lenalidomide were not eligible. All analyses were performed in pts who received 1 to 3 prior lines of therapy. Results: Median follow-up was 13.5 months. Pts who were lenalidomide-naive prior to the start of study treatment (DRd, n=226; Rd, n=219) demonstrated significantly longer PFS with DRd vs Rd (median: not reached [NR] vs 18.4 months; HR, 0.36; 95% CI, 0.25-0.52; P<0.0001), with estimated 12-month PFS rates of 83.0% vs 59.9%, respectively. ORR was significantly higher with DRd vs Rd (96% vs 79%), with ≥VGPR rates of 76% vs 47% and ≥CR rates of 44% vs 21%, respectively (P<0.0001 for all). In the lenalidomide-exposed subgroup (DRd, n=46; Rd, n=45), median PFS was NR in both treatment groups (HR, 0.49; 95% CI, 0.22-1.12; P=0.0826); estimated 12-month PFS rates were 84.1% vs 63.1%, respectively. ORR was higher with DRd vs Rd but did not reach statistical significance (87% vs 71%; P=0.0729); however, rates of ≥VGPR (78% vs 38%; P=0.0001) and ≥CR (44% vs 12%; P=0.0011) were significantly improved with DRd vs Rd, respectively. For bortezomib-naive pts (DRd, n=44; Rd, n=45), PFS was significantly longer with DRd vs Rd (median: NR vs 15.8 months; HR, 0.34; 95% CI, 0.13-0.86; P=0.0170), with estimated 12-month PFS rates of 85.4% vs 69.2%, respectively. ORR was significantly higher with DRd vs Rd (98% vs 82%; P=0.0158), with trends toward increased rates of ≥VGPR (74% vs 55%; P=0.0544) and ≥CR (42% vs 23%; P=0.0576). In the bortezomib-exposed pts (DRd, n=228; Rd, n=219), median PFS was NR in DRd vs 18.4 months in Rd (HR, 0.35; 95% CI, 0.24-0.50 P<0.0001); estimated 12-month PFS rates were 82.8% vs 58.7%, respectively. Significant differences in ORR (93% vs 77%), rate of ≥VGPR (77% vs 43%) and rate of ≥CR (44% vs 19%) were observed with DRd vs Rd, respectively (P<0.0001 for all). Among bortezomib-refractory patients (DRd, n=54; Rd, n=49), the PFS benefit of DRd compared with Rd was maintained (median: NR vs 10.3 mo, respectively; HR, 0.46; 95% CI, 0.25-0.85; P=0.0117; Figure). The estimated 12-month PFS rates were 70.8% vs 44.4%, respectively. Similar to bortezomib-exposed pts, ORR (92% vs 68%; P=0.0024), rate of ≥VGPR (75% vs 36%; P=0.0001), and rate of ≥CR (46% vs 13%; P=0.0003) were all significantly higher with DRd vs Rd for bortezomib-refractory pts. Updated data will be presented at the meeting. Conclusions: Among pts who received 1 to 3 prior lines of therapy, significantly longer PFS and higher ORR were observed with DRd vs Rd among pts who previously received bortezomib or were refractory to bortezomib or were lenalidomide-naive. Higher rates of deeper responses were observed in pts who previously received lenalidomide or bortezomib. Follow-up is ongoing to assess PFS in pts who received 1 to 3 prior lines of therapy and previously received lenalidomide. These results further strengthen the significant benefit of combining daratumumab with Rd for RRMM. Figure Progression-free Survival in Bortezomib-refractory Patients who Received 1 to 3 Prior Lines of Therapy Figure. Progression-free Survival in Bortezomib-refractory Patients who Received 1 to 3 Prior Lines of Therapy Disclosures Moreau: Janssen: Honoraria, Speakers Bureau; Novartis: Honoraria; Takeda: Honoraria; Celgene: Honoraria; Amgen: Honoraria; Bristol-Myers Squibb: Honoraria. Kaufman:Pharmacyclics: Consultancy; Incyte: Consultancy; Novartis: Consultancy, Research Funding; Celgene: Consultancy, Research Funding. Sutherland:Celgene: Consultancy, Honoraria; Amgen: Consultancy, Honoraria; Janssen: Consultancy, Honoraria. Lalancette:Celgene: Honoraria; BMS: Honoraria. Iida:Celgene: Honoraria, Research Funding; Janssen Pharmaceuticals: Honoraria, Research Funding. Prince:Janssen: Honoraria; Celgene: Honoraria. Cochrane:BMS: Other: Received sponsorship to attend international meetings; Novartis: Other: Received sponsorship to attend international meetings; Celgene: Other: Received sponsorship to attend international meetings; Takeda: Other: Received sponsorship to attend international meetings. Khokhar:Janssen: Employment. Guckert:Johnson & Johnson: Equity Ownership; Janssen: Employment. Qin:Janssen: Employment. Oriol:Janssen: Honoraria, Membership on an entity's Board of Directors or advisory committees; Celgene: Honoraria, Membership on an entity's Board of Directors or advisory committees; Amgen: Honoraria, Membership on an entity's Board of Directors or advisory committees.


2013 ◽  
Vol 31 (15_suppl) ◽  
pp. 10029-10029 ◽  
Author(s):  
Eric Bouffet ◽  
Katrin Scheinemann ◽  
Shayna M. Zelcer ◽  
Juliette Hukin ◽  
Beverley Wilson ◽  
...  

10029 Background: Vinblastine has shown promising activity in a phase II study in children with recurrent/refractory LGG. The aim of this study was to assess the activity of vinblastine in chemotherapy naïve children. Methods: Patients < 18 years old with unresectable or progressive LGG were eligible if they had not received any previous treatment with chemotherapy or radiation. Vinblastine was administered weekly at a dose of 6 mg/m2over a period of 70 weeks. Patients who showed progression on 2 consecutive imaging studies or evidence of clinical progression were removed from treatment. Results: 54 patients (23 female) were enrolled between 2007 and 2010. Median age at inclusion was 7 years, 13 patients were < 3 years. 32 had chiasmatic/hypothalamic tumours, 6 had evidence of dissemination. 13 had neurofibromatosis type 1. Histology was pilocytic astrocytoma (25), pilomyxoid astrocytoma (4), low grade astrocytoma variant (8); 17 patients had no histological diagnosis. Treatment was well tolerated; however, only 14 patients received full dose for the duration of the study. Most common toxicity was haematological: 40 patients who experienced grade 3+ neutropenia. There were only 6 episodes of febrile neutropenia, 3 RBC transfusions and no toxic death. Best response to chemotherapy was assessed centrally by an independent radiologist: 1 CR, 10 PR, 3 MR, 28 SD, 12 PD, for a response rate of 24.5%. With a median follow-up of 2 years (9-48 months), progression-free survival at 2 years was 72.1% (95%CI: 58.1-82.2). One patient died of progression. Conclusions: Weekly vinblastine is well tolerated in paediatric LGG patients. Although the response rate appears inferior to other common LGG regimens, progression free survival at 2 years favourably compares to most currently used regimens. Supported by a grant from the Ontario Institute Cancer Research. Clinical trial information: 1000011227.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. 5020-5020 ◽  
Author(s):  
Phuoc T. Tran ◽  
C. Leigh Moyer ◽  
Ryan Phillips ◽  
Noura Radwan ◽  
Ashley Ross ◽  
...  

5020 Background: The importance of local treatment in oligometastatic prostate cancer (OPC) is unknown. Stereotactic ablative radiotherapy (SABR) is highly focused, high-dose radiation that is well suited for treatment of oligometastases. Here we report on the safety and preliminary clinical outcomes of SABR in a modern cohort of OPC men. Methods: Eighty four men who satisfied criteria of OPC diagnosed on imaging underwent consolidative SABR were then followed prospectively on our IRB approved registry by our GU multidisciplinary team. We collected demographic, clinical, toxicity and efficacy information. We examined the first 66 men in this preliminary report to allow for a minimum of 4.5 months follow-up. SABR was delivered in 1-5 fractions of 5-18 Gy. Kaplan-Meier method was used to assess local progression-free survival (LPFS), biochemical progression-free survival (bPFS; PSA nadir+2), distant progression free survival (DPFS), ADT-free survival (ADT-FS) and time-to-next intervention (TTNI). Results: Of the 66 OPC patients analyzed, 25 (38%) men presented as synchronous OPC and the remaining 41 had recurrent OPC. Median and mean follow-up was 61 and 66 weeks, respectively. Patient and disease factors as listed in the Table. Crude Grade 1 and 2 acute toxicities were 36% and 11%, respectively, with no Grade > 2 toxicity. SABR was delivered to 134 metastases: 89 bone (66%), 40 nodal (30%) and 5 (4%) visceral metastases. Overall LPFS at 1-year was 92%. The bPFS and DPFS at 1-year were 69% and 69%, respectively. Median TTNI was not reached yet. Of the 18 men with hormone sensitive prostate cancer who had their ADT deferred, 11/18 (56%) remain free of disease following SABR (1-year ADT-FS was 78%) and in 17 castration resistant men, 11 had > 50% PSA declines with 1-year TTNI of 30% with a median of 45 weeks. Conclusions: Consolidative SABRfor OPCis feasible and well tolerated. The preliminary clinical outcomes in our series is limited by heterogeneity and size but our data suggests that this approach is worthy of further prospective study. [Table: see text]


2021 ◽  
Vol 38 (6) ◽  
Author(s):  
Luca Triggiani ◽  
Rosario Mazzola ◽  
Davide Tomasini ◽  
Alessio Bruni ◽  
Giulia Alicino ◽  
...  

AbstractThe aim of the present study was to explore the potential impact of upfront metastases-directed therapy (MDT) in terms of prolongation of castration-sensitive phase in a series of oligorecurrent castration-sensitive prostate cancer (PC) patients. The present article is a multicenter retrospective study. The population of interest was castrate-sensitive oligorecurrent PC, defined as the presence of 1–3 uptakes in non-visceral sites such as bones or nodes detected by means of 18F-Choline PET/CT or 68-Gallium PSMA PET/CT. Primary endpoint was the time to castration resistance. Secondary endpoints were ADT-free survival, local progression-free survival, and overall survival. Eighty-two patients and 118 lesions were analyzed. The median time to castration resistance for the entire population of the study was 49 months (95% CI 43.6–54.4 months). The 1- and 2-year TTCR-free survival rates were 94% and 82%, respectively. At the time of analysis, 52 patients were still in the castration-sensitive phase of the disease. In this cohort of patients, the median ADT-free survival was 20 months (range 3–69 months). On the other hand, during follow-up 30 patients switched to the castration-resistant phase of disease. In this last group of patients, the median ADT-free survival was 20 months (range 4–50 months). After the ADT administration, the median castration-sensitive phase was 29 months (range 5–71 months). Castration resistance generally occurs at a median follow-up of 24–36 months following ADT. In the current study, upfront MDT does not decrease the time from initiation of ADT to castration resistance.


2015 ◽  
Vol 33 (3_suppl) ◽  
pp. 411-411
Author(s):  
Stephen Y Oh ◽  
Alicia M Edwards ◽  
Hejin P Hahn ◽  
Margaret T Mandelson ◽  
Vincent J. Picozzi

411 Background: Natural history (NHx) and overall survival (OS) of patients (pts) with LPC and PPC without gross metastasis (GM) at initial diagnosis are rarely studied. We assessed rate, time and pattern of disease recurrence (REC), impact of local therapy (Rx) and OS in this pt cohort from our institution. Methods: The Virginia Mason Pancreatic Cancer (PC) Database (Picozzi et.al. Proc GI Cancer Symp 2011) was used to identify pt cohort. Inclusion criteria were 1) laparoscopic PPC and 2) no other evidence of GM (radiographic/ laparoscopic). Local progression (LP) was defined as primary tumor growth with change/cessation of chemoRx. Systemic progression (SP) was defined as radiological/pathological metastastic progression. The database provided OS. Results: 39/884 (4.4%) pts initiating Rx at VMMC from 2003-2014 met these criteria. 13/39 pts were included in a prior report (Clark et.al. Am J Surg 2010). Pt characteristics include: 21/39 (54%) male; median (med) age 65 (range 49 – 83) yrs; 25/39 (64%) head lesions; med tumor size 39 (range 25 - 100) mm. Med pt follow-up is 13 mo. All pts received gemcitabine-combination chemoRx as initial Rx. 14/39 (36%) pts also received local Rx (13 consolidative chemoXRT, 1 surgery). 3 month (mo) disease control (DC) was 86% (31/36 pts, 3 pts followed <3 mo). 6 mo progression - free survival (PFS) was 72% (26/36 pts). 6/36 pts (16%, range 9+-15+ mo) are progression – free. 30/36 pts (83%) progressed (med time 8.1 mo). REC pattern was local (13/30 pts, 43%), distant (9/30 pts, 30%) and combined (8/30 pts, 27%). Pts given local Rx did not differ statistically from pts given systemic Rx only with respect to 3 mo DC, 6 mo PFS, REC rate, time, or pattern, or OS. OS for LPC with PPC was significantly better than pts with GM (n = 277) identified from the database from 2003-2014 (med OS 15 vs. 9.3 mo, p = 0.0002; 1-yr OS 68 vs. 32%, p = 0.01; 2-yr OS 34 vs. 12%, p = 0.007). Conclusions: 1)LPC with PPC is a rare PC pt cohort with high response rates to chemoRx and superior OS to those with GM. 2) However, long-term disease free OS was not seen. 3)Local Rx did not clearly alter pt cohort NHx. 4)This is the largest pt series of this pt cohort compiled from a US institution; their further study seems of value.


2021 ◽  
Vol 11 ◽  
Author(s):  
Sung Uk Lee ◽  
Sang Min Yoon ◽  
Jason Chia-Hsien Cheng ◽  
Tae Hyun Kim ◽  
Bo Hyun Kim ◽  
...  

Background: No studies evaluating the clinical outcomes of radiotherapy (RT) for hepatocellular carcinoma (HCC) in the caudate lobe have been available to date. The purpose of this study was to evaluate the effectiveness and safety of RT for HCC in the caudate lobe.Material and Methods: Seventy patients with HCC in the caudate lobe treated with RT from a multi-institutional database were included in this study. The median equivalent dose in 2 Gy (EQD2) was 80.0 Gy10 (range, 31.3–99.3), and freedom from local progression (FFLP), progression-free survival (PFS), and overall survival (OS) rates were evaluated.Results: The median time of follow-up was 47.9 months (range, 3.4–127), and the 5-year FFLP, PFS, and OS rates were 80.6% [95% confidence interval (CI), 70.8–91.8], 13.8% (95% CI, 7.5–25.4), and 51.3% (95% CI, 39.9–66.1), respectively. In the multivariate analysis, the radiation dose was significantly associated with the FFLP rate [hazard ratio (HR), 0.57 per 10 Gy10 increase, p = 0.001], and the status of FFLP was significantly associated with OS (HR, 2.694, p = 0.014). The overall rate of ≥grade 3 adverse events was 5.7% (4 of 70), and RT-related mortality was not observed.Conclusion: RT for HCC in the caudate lobe showed promising FFLP and OS rates with safe toxicity profiles. These findings suggest that RT may be a promising treatment option for HCC in the caudate lobe.


Blood ◽  
2012 ◽  
Vol 120 (21) ◽  
pp. 2978-2978
Author(s):  
Shameem Mahmood ◽  
Christopher P. Venner ◽  
Thirusha Lane ◽  
Lisa Rannigan ◽  
Darren Foard ◽  
...  

Abstract Abstract 2978 Background: Novel agents have emerged as important therapeutic options for plasma cell dyscrasias and consequently have been adopted into treatment strategies for Systemic AL amyloidosis. Several studies to date have shown the efficacy of immunomodulatory drugs (IMiD) in this disease. However, little data exists exploring the efficacy of these agents especially in the context of previous treatment lines. The primary objective of this study was to evaluate the response, OS and PFS in patients treated with lenalidomide regimens in the relapsed setting. Further analysis was carried out examining outcomes based on previous lines of therapy. The importance of continuous therapy for control of the clonal disease stable disease was also examined. Patients and Methods: Our primary cohort consisted of 66 AL amyloidosis patients who presented to our Centre between July 2007 and July 2012, identified from the database of the UK National Amyloidosis Centre, commencing a lenalidomide regimen in the relapsed setting. It was recommended that treatment continue until disease progression or unacceptable toxicity. Organ involvement and haematological and organ response were defined according to the 2010 consensus criteria. Overall survival (OS) was calculated from the start of lenalidomide treatment until death or last follow-up. Progression-free survival (PFS) was calculated from the start of lenalidomide treatment until relapse, death or last follow-up. Survival endpoints were examined based on previous lines of treatment. A subgroup analysis was carried out in those attaining at least stable disease to examine the impact of continuous therapy. In this cohort PFS was examined based on those remaining on continuous therapy versus those discontinuing treatment due to intolerance or patient preference. Results: The median age was 59.1 years (range 39.9–76.9) and 52% were male. A lambda clonal plasma cell dyscrasia was present in 62%. 44% had ≥ 3 organ involvement with 50% having cardiac involvement by echo criteria. Mayo cardiac staging (at diagnosis) was stage I, II and III in 30%, 62% and 8% respectively. The median number of previous treatment lines was 2 (range 1–5), with thalidomide and bortezomib pre-treatment in 80% and 68% respectively. Grade 3 toxicity was seen in 27% of patients and was mainly haematologic. A thrombotic complication was noted in 1 patient. On an intention-to-treat basis, 56% achieved a haematological response with a complete response in 11%. A dFLC response was observed in 57% and 26% achieved a dFLC-VGPR or better. The median follow-up of 12.4 months. The median OS was 46.3 months and the estimated 2-year PFS was 67% (figure 1). Compared to those who did not receive the drug, previous exposure to thalidomide did not confer a worse survival outcome (estimated 2-year OS 67% vs 67%, p = 0.59). Similarly, previous exposure to Bortezomib did not confer a worse survival (estimated 2-year OS OS 88% vs 79%, p=0.09). 58 patients (88%) achieved stable disease or better. In this subgroup, the median duration on lenalidomide was 17.2 months. The 2-year OS and PFS were 84% and 78% respectively. Compared to those who halted therapy, continuous treatment with lenalidomide correlated with an improved PFS (estimated 2-year PFS 62% vs 80% respectively, p = 0.02). Conclusion: This data further supports the effectiveness of lenalidomide containing regimens in the treatment of relapsed AL amyloidosis. Previous lines of therapy did not negatively impact survival outcomes. Continuous therapy is important in maximizing the duration of the clonal response. Disclosures: No relevant conflicts of interest to declare.


2009 ◽  
Vol 27 (15_suppl) ◽  
pp. 2054-2054
Author(s):  
S. Taillibert ◽  
L. A. Vincent ◽  
B. Granger ◽  
Y. Marie ◽  
C. Carpentier ◽  
...  

2054 Background: Treatment with a regimen of bevacizumab/irinotecan has been shown to be effective in recurrent grade 3 and 4 gliomas, but the effect of this regimen against recurrent oligodendroglial tumors has not been specifically studied. Methods: The bevacizumab/irinotecan regimen was retrospectively evaluated in a consecutive series of 25 patients with recurrent oligodendroglial tumors. All patients had failed previous treatment with radiation therapy and at least one line of temozolomide chemotherapy. Bevacizumab (10 mg/kg) and irinotecan (125 or 340 mg/m2 according to the antiepileptic regimen) were delivered every 14 days. Response was measured clinically and on monthly MRI. Results: The objective response rate was 72% (20% complete response, 52% partial response). After a median follow-up (from the first cycle) of 310 days (95% CI, 47–499), the median progression-free survival was 174 days (95% CI, 116–342), and the median overall survival was 328 days (95%CI, 217-not reached). The 6-month progression-free survival was 42 % (95 % CI, 26% to 67%). Among the 20 patients who progressed at the time of the analysis, the radiological pattern of progression was atypical in seven patients with an isolated multifocal or diffuse spread of the FLAIR signal, or an isolated meningeal spread or FLAIR abnormalities preceding contrast-enhancement recurrence. Among the 10 patients who are still alive, two are still on follow-up since 6 months with a complete response after, respectively, 10 and 12 months of treatment. Among the 17 patients in whom the status of the main molecular alterations of gliomas could be evaluated (search for deletions of chromosomes 1p, 19q, 9p, 10q, and amplification of EGFR, MDM2, CDK4), no relation could be found between the response rate and the type of genetic change (including 1p-19q co-deletion). The profile of tolerance was fair, with treatment discontinuation in 20% of patients. Intratumoral hemorrhages occurred in six patients (24%), but the treatment had to be discontinued because of symptomatic bleeding in only one patient (4%). Conclusions: This regimen is effective in recurrent oligodendrogliomas, and the overall tolerance is acceptable. [Table: see text]


Cancers ◽  
2021 ◽  
Vol 13 (6) ◽  
pp. 1223
Author(s):  
Daniel Pink ◽  
Dimosthenis Andreou ◽  
Sebastian Bauer ◽  
Thomas Brodowicz ◽  
Bernd Kasper ◽  
...  

We aimed to evaluate the efficacy and toxicity of paclitaxel combined with pazopanib in advanced angiosarcoma (AS). The primary end point was progression-free survival (PFS) rate at six months (PFSR6). Planned accrual was 44 patients in order to detect a PFSR6 of >55%, with an interim futility analysis of the first 14 patients. The study did not meet its predetermined interim target of 6/14 patients progression-free at 6 months. At the time of this finding, 26 patients had been enrolled between July 2014 and April 2016, resulting in an overrunning of 12 patients. After a median follow-up of 9.5 (IQR 7.7–15.4) months, PFSR6 amounted to 46%. Two patients had a complete and seven patients a partial response. Patients with superficial AS had a significantly higher PFSR6 (61% vs. 13%, p = 0.0247) and PFS (11.3 vs. 2.7 months, p < 0.0001) compared to patients with visceral AS. The median overall survival in the entire cohort was 21.6 months. A total of 10 drug-related serious adverse effects were reported in 5 patients, including a fatal hepatic failure. Although our study did not meet its primary endpoint, the median PFS of 11.6 months in patients with superficial AS appears to be promising. Taking recent reports into consideration, future studies should evaluate the safety and efficacy of VEGFR and immune checkpoint inhibitors with or without paclitaxel in a randomized, multiarm setting.


Sign in / Sign up

Export Citation Format

Share Document