Anaplastic gliomas: Is there a role for aggressive concomitant treatment?

2013 ◽  
Vol 31 (15_suppl) ◽  
pp. e13050-e13050
Author(s):  
Luiz Gustavo Sueth Berriel ◽  
Fábio Nasser Santos ◽  
Ana Carolina Sigolo Levy Diniz ◽  
Ludmilla Thome Chinen ◽  
Alexandre Andre Balieiro Anastacio da Costa

e13050 Background: Anaplastic glioma is a rare type of glioma, therefore there is no standard treatment after surgery. Adjuvant treatment possibilities are monotherapy with radiotherapy or chemotherapy, or combination therapy done concomitantly or sequentialy. Methods: A comparison of adjuvant treatments in a retrospective analysis of 64 patients with the diagnosis of anaplastic glioma treated at A.C.Camargo Hospital, Sao Paulo-Brazil, from 1994 to 2012. Results: Fifty-eight percent were male, median age 42yo (16-79yo), 76,6% ECOG 0-1, Histopatology was 76,6% astrocytomas, 9,4% oligoastrocytomas and 14% oligodendrogliomas. Adjuvant treatment was concomitant radiochemotherapy in 45, 2%, sequential radiochemotherapy in 14,5%, radiotherapy-only in 16,1%, chemotherary-only in 9,7% and 14,5% received no adjuvant treatment. With a median follow up of 32,2mo (P25-P75 – 13, 3-62, 9mo), median progression free survival (mPFS) was 65mo (CI 29,2 – 100,7mo) and median overall survival (mOS) was 87mo (CI 26,3 – 147,7mo) in the whole group. There was a mOS of 31,4mo for astrocytomas, 89,2mo for oligoastrocytomas and not reached for oligodendrogliomas. Comparing with other treatments, the concomitant therapy group had a longer mPFS (124,8mo vs. 20mo, p: 0,016) and mOS (139,8mo vs. 27,9mo, p = 0.081). When only anaplastic astrocytoma where analyzed, treatment with concomitant radiochemotherapy had better mPFS (p = 0.01) and mOS (p = 0.033). On the other side, treatment with sequential radiochemotherapy didn’t have such impact in either mPFS (p: 0,22) or mOS (p: 0,27). Prognostic factors related with both mPFS and mOS in the whole group were astrocytic histology, multiple lesions and more than 50yo at diagnosis. Non-cortical lesions were also prognostic for mOS. In the multivariate analysis, treatment with concomitant radiochemotherapy had an impact at mPFS (HR 0,28, p = 0.006) and mOS (HR 0,44, p = 0.039). Conclusions: Since there is no standard adjuvant treatment for anaplastic gliomas, evidence for treatment is often taken from GBM studies. This study suggests that an aggressive strategy, like concomitant radiochemotherapy with temozolamide, is useful in grade 3 gliomas, specially in astrocystic histology.

2019 ◽  
Vol 21 (Supplement_3) ◽  
pp. iii87-iii87
Author(s):  
V Villani ◽  
A Fabi ◽  
P Gaviani ◽  
R Rudà ◽  
G Lombardi ◽  
...  

Abstract BACKGROUND Malignant gliomas are aggressive primitive brain tumor in adults. Today, the standard of care is Temozolomide (TMZ) administered daily with radiation therapy, followed by adjuvant TMZ. TMZ treatment has been considered to have a low toxicity profile. However, during concomitant treatment some patient may develop a severe myelosuppression. This toxicity may be in some cases prolonged and lead to treatment discontinuation. MATERIAL AND METHODS We have retrospectively collected data from 5 italian neuro-oncological centers, about glioma patients who developed severe and prolonged hematological toxicity during concomitant chemoradiotherapy with TMZ. The purpouse of this study is to evaluate: percentage of patients receiving adjuvant chemotherapy after severe myelotoxicity; rate of toxicity observed during adjuvant chemotherapy. RESULTS 54 glioma patients who developed myelosuppression of grade 3 or 4 were considered. Hystology was Glioblastoma in 45 patients (83%); 63% of patients were female. Myelotoxicity during concomitant phase occurred at a median of 4 weeks (range 1–8) from the start of treatment.After recovery of myelotoxicity 19 patients did not received any treatment while 35 (65%) were treated with chemotherapy (28 received standard TMZ, one TMZ with metronomic schedule, 2 lomustine and 4 other agents). Among patients treated with TMZ, 13 patients presented hematological toxicity grade 3–4 which required treatment discontinuation in 7 cases (20%). CONCLUSION we observed that 80 % of glioma patients presenting severe myelotoxicity during concomitant radiochemotherapy may be treated with maintenance TMZ after blood value recovery.


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 1564-1564 ◽  
Author(s):  
V. K. Puduvalli ◽  
P. Giglio ◽  
M. D. Groves ◽  
K. R. Hess ◽  
M. Gilbert ◽  
...  

1564 Background: Patients with recurrent anaplastic glioma (AG) have few treatment options after initial alkylating agent therapy. In this study, the efficacy of thalidomide and irinotecan against recurrent AG was tested to assess if synergistic activity of cytotoxic and antiangiogenic agents could affect clinical outcome. Methods: Patients with recurrent AG with a KPS≥70 not on enzyme inducing anticonvulsants and with fewer than three relapses after radiation therapy and chemotherapies were eligible; the total planned enrollment is 39 patients. Irinotecan is administered at 125 mg/m2 weekly for 4 weeks followed by 2 weeks rest; thalidomide is initiated at 100 mg daily and escalated weekly up to 400 mg daily. Warfarin (1 mg) is given for prevention of venous thromboembolism (VTE). Patients undergo clinical and radiologic evaluations every 6-weeks. The primary endpoint is progression free survival at 6 months (PFS-6). To determine possible radiologic correlates to treatment effects, DCE- MR perfusion-imaging studies are obtained at baseline and subsequent follow up visits. Results: 17 are evaluable for response; the remainder were inevaluable. All evaluable patients had previously failed temozolomide and 9 had also failed nitrosourea therapy. The median age is 44 yrs and median KPS is 90. Four patients are alive and progression free at 6-months whereas 9 have progressed; the median progression free survival is 23 weeks and the PFS-6 is 34%. The best response was a CR in one patient, PR in 2 and stable disease in 9. Two patients have died of unspecified causes probably related to treatment. Median overall survival has not been reached; the 12-month and 18 month survivals by Kaplan Meier analysis are 73% and 26% respectively. Grade 3 and 4 toxicities included fatigue (29%), leukopenia (29%), nausea/vomitting (24%), and diarrhea (18%) requiring dose reductions. Two patients had VTE. Conclusions: The preliminary results of this ongoing study suggest that the combination of irinotecan and thalidomide has activity in patients with recurrent anaplastic gliomas; the ongoing assessment of this combination in patients with AG will more definitively define whether the combination can be an effective second line therapy for this population. [Table: see text]


2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 11528-11528
Author(s):  
P. Costa ◽  
F. Braga ◽  
C. Sottomayor ◽  
M. Honavar ◽  
M. Resende ◽  
...  

11528 Background: The authors analyse retrospectively the impact on tumour control and toxicity of concomitant radiotherapy (RT) and Temozolomide (TMZ) in high grade gliomas (HGG) of the CNS, in patients treated in the Clínica de Radioterapia do Porto - Portugal. Methods: This cohort represents all patients with HGG treated between January 2002 and January 2006, with concomitant RT a median total dose of 60 Gy, 2 Gy per treatment given once daily 5-days/week and TMZ 75mg/m2 for 42 days, followed by adjuvant treatment with TMZ 5-days schedule every 28 days (150 mg/m2 for the first cycle increased to 200mg/m2). The cohort was retrospectively analyzed for gender distribution, age, extent of surgical resection, initial KPS, median overall survival (OS) and haematological toxicity. Results: 23 with HGG (6 females, 17 males) were treated with concomitant RT (44–72 Gy) and TMZ followed by adjuvant TMZ; median age was 58 (ranging from 17–72); median KPS was 80 (ranging from 40–90); 3 patients had complete resection, 17 partial resection and 3 biopsy. All patients except one, who had treatment interruption for thrombocytopenia, completed the concomitant phase of treatment; 19 patients continued to received adjuvant treatment with TMZ (median number of cycles was 5 (ranging from 1–20). Median OS (measured from the date of diagnosis to the date of death) was 20.4 month and 1 yr OS was 54%. During concomitant phase, one patient had grade 3–4 thrombocytopenia. During the adjuvant TMZ therapy 4 patients had grade 3–4 haematological toxicity (anaemia: 1; thrombocytopenia: 1; leucopoenia and thrombocytopenia: 2). Conclusions: The results of concomitant RT+TMZ followed by TMZ in these patients with HGG showed values in accordance with the latest data published on literature for this association. Differences observed might be due to the small sample size.RT+TMZ followed by adjuvant TMZ is a well tolerated treatment with better results in median OS comparatively with previous results of RT only treatment in HGG. Treatment related toxicity was within acceptable levels, and this approach became routine practice in this set of patients. No significant financial relationships to disclose.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 2079-2079
Author(s):  
Herwig Matthias Strik ◽  
Hans Rock ◽  
Kai Kallenberg

2079 Background: Recent data have been raising doubt if dose-dense temozolomide is more efficient against malignant gliomas than with conventional dosing. We report here updated results of combined near-continuous temozolomide, aimed at depleting anti-alkylating MGMT, with once weekly application of low-dose lomustine (CCNU) at first or second relapse of malignant gliomas. Methods: 25 consecutive patients with recurrent malignant gliomas (12 anaplastic gliomas WHO III, 13 glioblastomas or gliosarkomas WHO IV) were treated: 15 males (60%), 10 females (40%); mean age at start of chemotherapy was 52 (20-78) years; Eight patients were treated at first, 15 at second and two at third recurrence. All patients were pretreated with temozolomide. Four patients received fractionated, stereotactically guided re-irradiation (FSRT). The treatment regimen consisted of near-continuous temozolomide 50mg/m2 day 1‑5/7 and low-dose CCNU 40mg absolute dose day 6/7. In cases of bone-marrow depression, temozolomide was reduced in steps of 20mg total dose or – in more severe cases – chemotherapy was interrupted until normalization of blood counts. Results: In total, 89 cycles (months) of chemotherapy were applied. The combination was well tolerated in terms of nausea and fatigue. Blood counts usually decreased continuously, enabling a gradual dose adaptation. Hematological WHO grade 3+4 toxicity occurred in 5/25 patients (20%), two of them were symptomatic. Three patients had prolonged elevation of liver enzymes. Best responses after ≥ 3 months (23 patients) were: 2 complete and 1 partial remissions (13%, ), 13 stable diseases (57%), and 7 progressive diseases (30%). Progression-free survival at six months from start of chemotherapy of the 16 patients treated > 6 months or deceased (PFS 6) was 37%, median overall survival 6.3 months. Conclusions: Although some hematotoxicity was observed, the regimen presented here is well tolerated and safe if carefully controlled. The objective responses and considerable rate of stable diseases indicate that this combination is active in malignant gliomas after pretreatment with temozolomide alone. The results have to be controlled in a prospective trial.


2012 ◽  
Vol 30 (15_suppl) ◽  
pp. 2050-2050
Author(s):  
Johnny Duerinck ◽  
Stephanie Du Four ◽  
An Van Binst ◽  
Hendrik Everaert ◽  
Alex Michotte ◽  
...  

2050 Background: Recurrent low-grade and anaplastic gliomas eventually transform to a higher grade that is characterized by neo-angiogenesis. Sunitinib inhibits multiple tyrosine kinase receptors (including VEGFR, PDGFR and c-Kit) but has no meaningful activity as a mono-therapy for recurrent glioblastoma when given at a daily dose of 37,5 mg (Neyns et al. J Neurooncol. 2011). We investigated sunitinib in combination with lomustine (CCNU) for the treatment of patients (pts) with temozolomide (TMZ) refractory recurrent low-grade or anaplastic glioma. Methods: Pts received a daily dose of 25 mg sunitinib for 28 consecutive days followed by a 14 days treatment-free interval. CCNU was administered as a single dose (80 mg/m²) on day 14 of a 6w cycle. T1 ± Gd and T2/FLAIR weighted MRI images were obtained q6 wks. 18FET-PET was performed at baseline and reassessed in responding pts. Results: 13 pts were enrolled (mean age 40 [range 33-49]; M/F 8/5; KPS 80-90/70-60: 8/5 pts). All pts had PD following surgery, RT and TMZ. In 7 pts, treatment was initiated at 2nd recurrence, in 4 pts at 3rd recurrence and in 2 pts at the 4th recurrence. Most frequent AEs where fatigue (gr 2: n= 3; gr 3: n= 1), thrombopenia (gr 2: n= 1 gr 3: n= 3 gr4: n= 1), neutropenia (gr 2: n= 2; gr3: n= 2; gr4: n= 2) and lymphopenia (gr 2: n=1; gr 3: n=2; gr 4: n=1). In 5/13 pts CCNU had to be discontinued because of AEs. Treatment with sunitinib was continued in these cases without reoccurrence of AEs. BOR according to RANO criteria: 1 CR, 1 PR (not-confirmed) and 2 SD (DCR: 4/13= 31%). In the patient with CR, FET-PET indicated a complete metabolic response. After a mean FU of 7 mths (range 2 -19), 5 pts are alive. The 6-month PFS is 23% (3/13 pts); median PFS is 1,8 mths (95%CI 1.0 - 2,7). A durable disease control was obtained in 3 pts (TTP respectively 14.8, 11.8+ and 19.2+ mths). Conclusions: in this heavily pretreated population with recurrent low-grade and anaplastic glioma the combination of sunitinib and CCNU is associated with acceptable toxicity and offers a durable progression-free survival in a subgroup of pts. Molecular predictive factors identifying the sensitive population would be needed to justify further clinical investigation of this combination.


2019 ◽  
Vol 37 (15_suppl) ◽  
pp. e15643-e15643
Author(s):  
Jyothika Mamadgi ◽  
Kristina Lundeen ◽  
Shaakir Hasan ◽  
Sunita Patruni ◽  
Ahmed Khattab ◽  
...  

e15643 Background: Biliary tract cancers are rare cancers with an annual incidence of about 8,000. In resected non-metastatic BTCs (NM-BTCs), cure rates decrease significantly due to local and distant recurrence. However, guidelines to use adjuvant therapies are not well established due to the lack of well-structured phase three randomized trials, given the rarity of the disease. We present a retrospective study analyzing outcomes of NM-BTCs managed within Allegheny Health Network (AHN) with specific focus on patients who underwent resection and received different adjuvant treatment modalities. Methods: Data of all patients with NM-BTCs treated at AHN from January 2012 to December 2017 was collected and outcomes analyzed for patients treated with surgery vs no surgery We then sub-stratified the patients who received surgery into three groups based on the modality of adjuvant therapy. Group 1 included patients with no additional therapy , Group 2 with adjuvant chemotherapy and Group 3 with adjuvant chemotherapy+radiation. We compared Mean Progression-Free Survival (PFS) and Overall Survival (OS) in these three groups. Results: Of the 59 NM-BTCs treated at AHN, 32(52.4%) underwent surgery. The OS for surgical vs non-surgically treated patients was 23 vs 17 months (p = 0.008) and PFS 26 vs 19 months (P = 0.06) respectively. Of the resected –patients, PFS for patients in Group1 vs Group2 vs Group3 was 16 vs 21 vs 34 months (pooled P = 0.05) and OS was 19 vs 22 vs 19 months respectively (pooled P = 0.5). PFS for patients in Group1 vs Group (2+3),i.e, patients treated surgery only vs surgery+ any adjuvant treatment was 16 vs 28 months (P = 0.1), OS was 19 vs 22 months (P = 0.7)respectively. PFS in patients in Group 3 vs Group (1+2),i.e, patients treated with adjuvant chemotherapy+radiation vs others was 34 vs 19 months. (p = 0.0183) respectively. Conclusions: This analysis revealed a statistically significant improvement in OS in patients with NM-BTCs who received surgery. There was a trend towards improvement in PFS/OS in patients who received adjuvant therapy, though not statistically-significant. Use of adjuvant chemotherapy+radiation resulted in statistically significant improvements in PFS as compared to other treatment modalities.


2019 ◽  
Vol 21 (Supplement_6) ◽  
pp. vi133-vi133
Author(s):  
Veronica Villani ◽  
Alessandra Fabi ◽  
Paola Gaviani ◽  
Roberta Rudà ◽  
Giuseppe Lombardi ◽  
...  

Abstract INTRODUCTION Malignant gliomas are aggressive primitive brain tumor in adults. Today, the standard of care is Temozolomide (TMZ) administered daily with radiation therapy, followed by adjuvant TMZ. TMZ treatment has been considered to have a low toxicity profile. However, during concomitant treatment some patient may develop a severe myelosuppression. This toxicity may be in some cases prolonged and lead to treatment discontinuation. METHODS We have retrospectively collected data from 5 Italian neuro-oncological centers, about glioma patients who developed severe and prolonged hematological toxicity during concomitant chemoradiotherapy with TMZ. The purpose of this study is to evaluate: percentage of patients receiving adjuvant chemotherapy after severe myelotoxicity; rate of toxicity observed during adjuvant chemotherapy. RESULTS 54 glioma patients who developed myelosuppression of grade 3 or 4 were considered. Histology was Glioblastoma in 45 patients (83%); 63% of patients were female. Myelotoxicity during concomitant phase occurred at a median of 4 weeks (range 1–8) from the start of treatment. After recovery of myelotoxicity 19 patients did not received any treatment while 35 (65%) were treated with chemotherapy (28 received standard TMZ, one TMZ with metronomic schedule, 2 lomustine and 4 other agents). Among patients treated with TMZ, 13 patients presented hematological toxicity grade 3–4 which required treatment discontinuation in 7 cases (20%). CONCLUSION the results of our study show that 80% of glioma patients presenting severe myelotoxicity during concomitant radiochemotherapy may be treated with maintenance TMZ after recovery of myelosuppression.


2017 ◽  
Vol 35 (15_suppl) ◽  
pp. TPS9600-TPS9600 ◽  
Author(s):  
Elisa A. Rozeman ◽  
Alexander Christopher Jonathan Van Akkooi ◽  
Emilie Routier ◽  
Alexander M. Menzies ◽  
Hanna Eriksson ◽  
...  

TPS9600 Background: The outcome of high risk stage IIIb and IIIc melanoma patients is poor, with a 5 year overall survival (OS) rate of < 50%. Adjuvant high dose IPI significantly improves 5 year progression free survival (PFS) and OS. In stage IV patients the combination of IPI and NIVO improves response rates (RR) and PFS compared to monotherapy, but at cost of higher toxicity. Neo-adjuvant treatment may be a favorable approach as immune checkpoint inhibition (ICI) is of greatest value at the moment of TCR triggering and therefore dependent on the amount of antigen present. The phase Ib OpACIN study comparing neo-adjuvant and adjuvant IPI and NIVO showed that neo-adjuvant treatment is feasible as all patients underwent surgery on time. The neo-adjuvant pathological RR was 80%, although 18/20 patients (90%) stopped early due to ≥1 grade 3 or 4 immune-related adverse events (irAEs). To date (median follow-up 45 wks), none of the 8 responders in the neo-adjuvant arm have relapsed. This raises the question whether the neo-adjuvant IPI and NIVO schedule can be adjusted to reduce toxicity but preserve efficacy. Methods: The aim of the multi-center phase 2 OpACIN-neo trial is to identify an optimal neo-adjuvant combination scheme of IPI and NIVO. 90 patients with resectable stage III melanoma will be randomized 1:1:1 between three combination schemes of IPI and NIVO. Patients in arm A will receive 2 courses standard regimen IPI 3mg/kg + NIVO 1mg/kg q3wks, in arm B IPI 1mg/kg + NIVO 3mg/kg q3wks, and in arm C 2 courses of IPI 3mg/kg q3wks directly followed (2-24hr) by 2 courses NIVO 3mg/kg q2wks. All patients will undergo surgery at week 6. The primary endpoints are rate of grade 3/4 irAEs, pathological RR, and radiologic RECIST 1.1. An interim analysis is planned after 13 patients have been accrued to each arm (according to the Simon 2-stage design). Major inclusion criteria are: ≥1 measurable lymph node metastases (according to RECIST 1.1) that can be biopsied, no history of in-transit metastases in the last 6 months, and naïve for ICI. Baseline biopsies and blood samples (week 0, 6, 12) will be taken for translational research. The first center started inclusion in December 2016, until now 2 patients have been enrolled. Clinical trial information: NCT02977052.


2010 ◽  
Vol 28 (13) ◽  
pp. 2181-2190 ◽  
Author(s):  
Anne Aupérin ◽  
Cecile Le Péchoux ◽  
Estelle Rolland ◽  
Walter J. Curran ◽  
Kiyoyuki Furuse ◽  
...  

Purpose The previous individual patient data meta-analyses of chemotherapy in locally advanced non–small-cell lung cancer (NSCLC) showed that adding sequential or concomitant chemotherapy to radiotherapy improved survival. The NSCLC Collaborative Group performed a meta-analysis of randomized trials directly comparing concomitant versus sequential radiochemotherapy. Methods Systematic searches for trials were undertaken, followed by central collection, checking, and reanalysis of updated individual patient data. Results from trials were combined using the stratified log-rank test to calculate pooled hazard ratios (HRs). The primary outcome was overall survival; secondary outcomes were progression-free survival, cumulative incidences of locoregional and distant progression, and acute toxicity. Results Of seven eligible trials, data from six trials were received (1,205 patients, 92% of all randomly assigned patients). Median follow-up was 6 years. There was a significant benefit of concomitant radiochemotherapy on overall survival (HR, 0.84; 95% CI, 0.74 to 0.95; P = .004), with an absolute benefit of 5.7% (from 18.1% to 23.8%) at 3 years and 4.5% at 5 years. For progression-free survival, the HR was 0.90 (95% CI, 0.79 to 1.01; P = .07). Concomitant treatment decreased locoregional progression (HR, 0.77; 95% CI, 0.62 to 0.95; P = .01); its effect was not different from that of sequential treatment on distant progression (HR, 1.04; 95% CI, 0.86 to 1.25; P = .69). Concomitant radiochemotherapy increased acute esophageal toxicity (grade 3-4) from 4% to 18% with a relative risk of 4.9 (95% CI, 3.1 to 7.8; P < .001). There was no significant difference regarding acute pulmonary toxicity. Conclusion Concomitant radiochemotherapy, as compared with sequential radiochemotherapy, improved survival of patients with locally advanced NSCLC, primarily because of a better locoregional control, but at the cost of manageable increased acute esophageal toxicity.


Author(s):  
Yi-Jou Tai ◽  
Heng-Cheng Hsu ◽  
Ying-Cheng Chiang ◽  
Yu-Li Chen ◽  
Chi-An Chen ◽  
...  

Adjuvant treatment in advanced-stage (stages III /IV) endometrial carcinomas in terms of tumor grades has not yet been explored. We retrospectively analyzed 194 patients with advanced-stage endometrioid endometrial carcinoma who received surgery, followed by adjuvant therapy, at National Taiwan University Hospital between January 1, 2000 and August 31, 2017. Adjuvant therapies included radiation (RT), chemotherapy alone (CT), and combined modality treatment (CMT: radiation and chemotherapy). The prognostic factors were determined from multivariate survival analyses using Cox regression models. Progression-free survival (PFS) and overall survival (OS) times were estimated with the Kaplan–Meier method. The median follow-up was 45.5 months (range: 6.2–207.9). In grade 1/2 endometrioid carcinoma, neither adjuvant CT nor CMT could prolong PFS significantly compared to RT (CT: HR 1.59, 95% CI 0.64–3.97; CMT: HR 2.03, 95% CI 0.72–5.74). Notably, maximal cytoreduction independently improved PFS (HR 0.31, 95% CI 0.10–0.90). No particular adjuvant treatment provided an OS advantage over the others for grade 1/2 endometrioid carcinomas. However, for grade 3 endometrioid carcinoma, CMT showed OS benefits (HR 0.15, 95% CI 0.03–0.89) compared to RT and CT. In conclusion, maximal cytoreduction should be the goal in patients with grade 1/2 advanced-stage endometrioid carcinomas. Based on our results, patients with grade 3 endometrioid carcinomas might benefit from adjuvant CMT.


Sign in / Sign up

Export Citation Format

Share Document