A multicenter phase II study of pemetrexed and cisplatin in patients with advanced gastric cancer (AGC)

2006 ◽  
Vol 24 (18_suppl) ◽  
pp. 14008-14008
Author(s):  
Y. Bang ◽  
Y. Kim ◽  
H. C. Chung ◽  
W. Kang ◽  
S. Park ◽  
...  

14008 Background: Pemetrexed is a novel folate antimetabolite, and it inhibits a number of folate-dependent enzymes. This agent has demonstrated activity in a variety of tumor types including AGC. This study was performed to evaluate the combination of pemetrexed and cisplatin in the treatment of AGC. The primary endpoint was response rate, and secondary endpoints were duration of response, time to progressive disease, time to treatment failure, overall survival, and toxicity. Methods: Patients with stage IV AGC not to be amendable to curative surgery and measurable disease were eligible. Pemetrexed 500 mg/m2 and cisplatin 75 mg/m2 were given on day 1, every 21 days. Treatment was supplemented with folic acid, vitamin B12, and dexamethasone. Response was assessed by RECIST, and toxicity was assessed by NCI-CTC v 2.0. Results: From October 2003 to September 2004, 51 patients were enrolled, but 1 did not meet the eligibility criteria. There were 37 men and 13 women with a median age of 56 years (range, 24–69) and an ECOG PS 0/1 for 14/36 patients; all had metastatic disease. Of 50 evaluable patients, there were no complete responses, and 13 had confirmed partial responses (26%; 95% CI, 14.6%-40.3%). Fifteen patients (30%) had stable disease, and 21 (42%) progressed, and 1 (2%) was unknown. Among 13 responders, the median durarion of response was 3.60 months (95% CI, 2.80–9.40). Median time to progressive disease was 2.8 months (95% CI, 2.20–4.40), and median overall survival was 6.6 months (95% CI, 4.80–10.40). The median time to treatment failure was 2.10 months (95% CI, 1.00–2.80). Survival estimates were 32.0% at 3 months and 7.0% at 6 months. A total of 212 cycles were administered to 51 patients (median 4 [range, 1–13]). Based on 51 patients, most common grade 3/4 hematologic toxicities were neutropenia (49.0%), leukopenia (19.7%), and anemia (13.7%); the most common grade 3/4 nonhematologic toxicities were hyponatremia (15.7%), anorexia (9.8%), nausea (7.8%), and vomiting (7.8%). Conclusions: : The combination of pemetrexed and cisplatin in the current dose and schedule has a modest activity and a mild toxicity profile in patients with AGC. Further study is warranted using a different dose and treatment schedule. [Table: see text]

Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 936-936 ◽  
Author(s):  
Rebecca L. Elstrom ◽  
Charalambos Andreadis ◽  
Nicole Aqui ◽  
Donald E. Tsai

Abstract Background: PTLD is a rare but life-threatening side effect of organ transplantation. While some patients respond to reduction in immunosuppression (RI), in many patients this approach is ineffective or impractical. Chemotherapy and anti-B cell monoclonal antibody therapy have been explored for treatment of such patients, but most reports include small numbers of patients. We report the results of treatment of patients with PTLD with rituximab and/or chemotherapy at the University of Pennsylvania Medical Center. Methods: Data were collected using retrospective chart and database review. Patients were evaluated for baseline characteristics as well as response to therapy, time to treatment failure, and overall survival. Prognostic factors were identified by univariate analysis. Results: 35 of 117 adult solid organ transplant patients diagnosed with PTLD that were refractory to RI underwent treatment with rituximab and/or chemotherapy. Median time to PTLD was 4 years (range 3 months to 23 years), and most patients presented with advanced stage (57%) and extranodal involvement (74%). 71% had elevated LDH. There were no significant baseline differences between patients receiving rituximab and those receiving chemotherapy. 22 patients underwent treatment with rituximab. The ORR was 68% (CR 59% and PR 9%). Median time to treatment failure (TTF) was not reached at median follow up of 19 months (range 0.8–51 months), and estimated OS was 31 months (1.5–51 months). EBV positivity was a favorable prognostic factor for achievement of response and TTF (p<0.01). LDH elevation predicted shorter overall survival (0.04). No patient died due to rituximab toxicity, and all 8 patients who either did not respond or relapsed were able to undergo further treatment with chemotherapy. 23 patients received chemotherapy, 22 of whom were evaluable. ORR was 74% (CR 57% and PR 17%). At a median follow up of 27 months, median TTF was 10.5 months (0.4–54 months), and estimated OS was 42 months (0.4–70 months). Adverse prognostic factors for response included advanced stage (p=0.02), elevated LDH (p=0.01) and allograft involvement by tumor (p<0.01). These factors, in addition to lack of achievement of CR, also predicted poor overall survival (p<0.05 for all factors). 26% of patients receiving chemotherapy died due to treatment-related toxicity. Conclusion: Rituximab and chemotherapy are effective treatments in patients with PTLD who fail or do not tolerate RI. While rituximab is generally well tolerated, chemotherapy is associated with marked toxicity. When possible, PTLD patients requiring therapy beyond RI should be considered for rituximab, especially those with EBV-positive disease. Chemotherapy should be reserved for those patients who fail rituximab, have EBV-negative tumors, or need a rapid response.


2020 ◽  
Vol 14 (Supplement_1) ◽  
pp. S120-S121
Author(s):  
M C Dubinsky ◽  
J W Chou ◽  
C Su ◽  
W Wang ◽  
I Modesto ◽  
...  

Abstract Background Tofacitinib is an oral, small-molecule JAK inhibitor for the treatment of ulcerative colitis (UC). Safety and efficacy of tofacitinib were evaluated in two Phase 3 induction studies (OCTAVE Induction 1 and 2; NCT01465763, NCT01458951), a 52-week, Phase 3 maintenance study (OCTAVE Sustain, NCT01458574) and an ongoing, open-label, long-term extension study (NCT01470612).1,2 Here, we assess time to treatment failure among patients in remission at the end of OCTAVE Induction 1 and 2 who enrolled in OCTAVE Sustain. Methods In OCTAVE Induction 1 and 2, patients received placebo or tofacitinib 10 mg twice daily (BID) for 8 weeks; clinical responders were re-randomised to placebo, tofacitinib 5 or 10 mg BID in OCTAVE Sustain for 52 weeks. Kaplan–Meier method was used to estimate median time to treatment failure (withdrawal due to insufficient clinical response) in patients who received 10 mg BID in induction studies and entered OCTAVE Sustain in remission (total Mayo score ≤2 with no subscore >1, and rectal bleeding subscore [RB] 0). Treatment failure: ≥3-point increase from baseline total Mayo score, plus ≥1-point increase in RB and endoscopic subscore (ES; centrally read) and absolute ES ≥2 after ≥8 weeks of maintenance therapy. Results Following induction, 156 patients in remission entered OCTAVE Sustain and received tofacitinib 5 mg BID (N = 57) or 10 mg BID (N = 50), or placebo (N = 49). Estimated treatment failure rates are reported in the table. At Week 52, Kaplan–Meier rates of treatment failure were higher in patients who switched to placebo (81.8% [95% confidence interval 67.0, 90.4]) vs. those who continued to receive tofacitinib 10 mg BID (25.6% [14.2, 38.6]) or reduced their dose to 5 mg BID (34.4% [21.8, 47.3]). Median time to treatment failure was 169 days for placebo and >52 weeks for tofacitinib 5 and 10 mg BID (due to the low number of treatment failure events, exact median time to treatment failure was not available for tofacitinib 5 or 10 mg BID). Conclusion For patients in remission following 8 weeks of induction treatment with tofacitinib 10 mg BID, median time to treatment failure for those who continued tofacitinib treatment (5 or 10 mg BID) was >52 weeks. After treatment interruption (remission patients who were re-randomised to placebo), median time to treatment failure was 169 days; this was similar to previously published time-to-treatment-failure data for patients with clinical response (including remission) following 8 weeks of induction treatment with tofacitinib who were re-randomised to placebo.3. References


Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3049-3049 ◽  
Author(s):  
Eva Hoster ◽  
Michael Unterhalt ◽  
Bernhard Wörmann ◽  
Ulrich Dührsen ◽  
Bernd Metzner ◽  
...  

Abstract Background: The addition of rituximab to chemotherapy (R-CHOP) has been shown to improve response rates in mantle cell lymphoma (MCL), but prolongation of response duration or overall survival was not observed (Lenz et al., JCO 2005). In a similar randomized comparison of 90 patients, again the addition of rituximab to MCP showed a tendency towards higher CR rates, but no improvement of overall response rate, progression free, or overall survival (Herold et al., ICML-10, 2008). Methods: We present an update of a previously published trial randomly comparing efficacy and safety of R-CHOP to CHOP induction in previously untreated patients with advanced stage MCL. Results: Of the 123 evaluable patients, 63 patients were randomized to R-CHOP. Median age was 62 years, and risk profiles of the two treatment arms were comparable. Overall response rates were 92% vs. 75% for R-CHOP vs. CHOP (p = 0.0139) and complete remission rates 33% vs. 8% (p = 0.0008). After a median follow-up of 65 months, median time to treatment failure was prolonged from 14 months for CHOP to 28 months for R-CHOP (p = 0.0003). Similarly, median response duration was prolonged from 18 (CHOP) to 29 months (R-CHOP, p = 0.0052). So far, no significant improvement of overall survival has been observed with median not reached vs. 59 months and 5-years OS rates of 59% and 46% (p = 0.27) after R-CHOP and CHOP, respectively. Toxicity was not significantly higher for R-CHOP treated patients. Conclusions: After longer follow-up, superior remission rates, time to treatment failure, and response duration of combined immuno-chemotherapy were confirmed. However, in contrast to other lymphoma entities, improvement of overall survival has not yet been proven in MCL patients. Therefore new therapeutic options are urgently warranted to further improve the long term outcome in this otherwise dismal disease.


Neurosurgery ◽  
2011 ◽  
Vol 70 (5) ◽  
pp. 1125-1134 ◽  
Author(s):  
Matthew Bender ◽  
Gustavo Pradilla ◽  
Sachin Batra ◽  
Alfred See ◽  
Neal Bhutiani ◽  
...  

Abstract BACKGROUND: Percutaneous glycerol rhizotomy (GR) is used to treat trigeminal neuralgia (TN), with satisfactory pain relief lasting 2 to 3 years in most patients after the first intervention. The efficacy of subsequent GRs, however, has not been studied. OBJECTIVE: To compare the pain relief and durability achieved by the first GR with those obtained after subsequent GRs in a retrospective cohort of TN patients. METHODS: Between 1998 and 2010, 548 patients with TN underwent 708 GRs. After exclusions, 430 initial GRs (GR1) and 114 subsequent GRs (GR2+) were compared in terms of initial pain relief, durability, sensory change, and complications. Durability was assessed by determining median time to treatment failure for all GRs achieving complete pain relief without medications (n = 375: 264 failures, 111 censored). Predictors of initial pain relief were assessed by logistic regression, and predictors of failure were assessed by Cox regression analysis. RESULTS: After GR1, pain relief results were as follows: 285 patients (66%) were pain free without medications, 26 (6%) were pain free with medications, 66 (15%) improved, and 53 (12%) were unchanged. After GR2+, results were as follows: 90 patients (79%) were pain free without medications, 6 (5%) were pain free with medications, 7 (6%) improved, and 11 (10%) were unchanged (P = .03). Median time to treatment failure was 26 months after GR1 and 25 months after GR2+ (P = .34). On multivariate analysis, prior GR was a positive predictor of initial pain relief (odds ratio, 2.067; 95% confidence interval, 1.243-3.437; P = .005) and had no effect on durability. CONCLUSION: TN patients experienced greater pain relief and equivalent durability after GR2+ beyond the initial treatment.


Blood ◽  
2005 ◽  
Vol 106 (11) ◽  
pp. 5129-5129
Author(s):  
Peter Haas ◽  
Ulrich Denz ◽  
Gabriele Ihorst ◽  
Monika Engelhardt

Abstract The central goal in improving multiple myeloma (MM) treatment is the achievement of higher complete remission (CR) rates and prolonged progression free (PFS) and overall survival (OS). Maintenance treatment after high-dose regimens has been one approach to attain this goal. As one of these maintenance agents Thalidomide (Thal) is used, since it leads to the inhibition of fibroblast growth factor (bFGF) and monocyte-derived TNF-alpha, reduced vascularisation, immune response upregulation and others. Response rates with single Thal are 30% and can be increased up to 60–70% with combined Dexamethasone (Dex) use. Maintenance treatment has shown an improved 3-year PFS rate of 15% after tandem autologous stem cell transplantation (auto-SCT). Although Thal is well tolerated compared to regular cytotoxic drugs, side effects occur in about one third of patients (pts), most disturbing being fatigue, polyneuropathy and deep venous thrombosis. As these are dose dependent, a dose-reduction has evolved from 800mg to 200mg/day (d). In this study, we analyzed, whether the Thal dose of 100mg/d is feasible and effective: 38 consecutive MM pts received Thal at our center between 5/01 and 6/05. The time to treatment failure was calculated from treatment start to death, relapse or therapy modification due to side effects. The median age of all pts was 62.4 years; 27 were male and 11 female. All pts had stage II and III MM according the DurieIgG was the most frequent MM subtype in 23 pts. Twelve showed deletion 13q14. The median time from initial diagnosis to Thal therapy was 3.9 years (range; 0–18.7). Pts had received a median of 2 treatment lines (range 0–3), three pts had received Thal as first-line treatment. Twenty-six pts (68.4%) underwent SCT prior to Thal treatment (23 pts single auto-SCT, 1 pt tandem auto SCT, 1 pt sequential auto- and allo-SCT and 1 pt with 2 allo-SCT). The median Thal dose was 125mg/d (range 50–800mg/d), 22 pts received more than 200mg/d and 16 pts less. The median Thal duration was 7 months (range; 0.3–37.8). Nineteen pts received Thal as a single-agent, 16 pts in combination with Dex or melphalan (Alexanian). Remission before Thal was PD in 21 pts, SD in 9 pts, MR in 1 pt, PR in 4 pts and CR in 3 pts. Causes for treatment failure were side effects in 10 pts, relapse in 18 pts and death in 1 pt. The median time to treatment failure (TTF) was 7.6 months. Subgroup analyses showed an advantage for pts without deletion 13q14 (8.2 vs. 2.8 months; p=0.054) and for the combination therapy with Thal/Dex or Thal/Alexanian (8.7 vs. 4.5 months; p=0.0554). Other parameters tested in univariate analysis (age, stage, remission status, prior Tx) showed no difference in TTF. Of note was that different dose-levels (<200 vs. ≥200mg/d) revealed no difference in TTF (7.5 vs. 8.2 months; p=0.087). We conclude that a Thal dose of 100/d is efficient, which can be increased with combined Dex or other effective anti-MM usage. This is of importance since side-effects show a dose-relation and often lead to treatment discontinuation. The strategy to lower the Thal dose with persistence of high efficacy rates is a promising approach to optimize MM-treatment and make it a feasible option for more pts.


Blood ◽  
2006 ◽  
Vol 108 (11) ◽  
pp. 482-482 ◽  
Author(s):  
Christian Buske ◽  
Michael Kneba ◽  
Eva Lengfelder ◽  
Michael Pfreundschuh ◽  
Wolf-Dieter Ludwig ◽  
...  

Abstract Follicular lymphoma (FL) is an indolent disease of the advanced age with more than 40 % of the patients being older than 60 years at diagnosis and an age-specific incidence peaking above 75 years. We now analyzed the treatment outcome of elderly patients in the GLSG multicenter phase III study comprising a prospective randomized comparison of R-CHOP versus CHOP alone in patients with advanced stage FL. 221 patients > 60 years with untreated FL were randomized for therapy with R-CHOP (R-CHOP: Rituximab 375 mg/m2 d0–1; cyclophosphamide 750 mg/m2 d1; doxorubicine 50 mg/m2 d1; vincristine 1.4 mg/m2 d1; prednisone 100 mg/m2 d1–5) (n=109) or CHOP alone (n=112). Patient characteristics were well balanced between the treatment groups, also with regard to the distribution of the FLIPI risk groups (≥ 3 adverse factors 73% and 66 % in the R-CHOP and CHOP arm, respectively). R-CHOP induced higher overall response rates and significantly prolonged the time to treatment failure (TTF)(median 5.0 years versus 2.1 years, respectively; logrank test: p<0.0001) compared to CHOP in the elderly patient group. Furthermore, the estimated 4-years progression free survival was 62.2% for R-CHOP versus 27.9 % after CHOP (logrank: p< 0.0001). Importantly, R-CHOP was able to prolong the overall survival in elderly patients compared to CHOP with an estimated 4-years overall survival of 90% after immunochemotherapy versus 81 % after CHOP alone (logrank test: p=0.039). In the multivariate analysis individual FLIPI risk factors such as elevated serum LDH level, a hemoglobin level below 12 g/dl, the number of nodal areas (> 4) as well as application of CHOP alone were independently associated with a shorter TTF. Treatment related side effects were similar in both patient groups and comprised predominantly myelosuppression. In summary, the addition of Rituximab to CHOP significantly improves the outcome of elderly patients with previously untreated advanced stage FL without adding major side effects.


2016 ◽  
Vol 34 (23) ◽  
pp. 2736-2742 ◽  
Author(s):  
Peter C. Enzinger ◽  
Barbara Ann Burtness ◽  
Donna Niedzwiecki ◽  
Xing Ye ◽  
Kathe Douglas ◽  
...  

Purpose To determine the optimal chemotherapy backbone for testing in future US cooperative group studies for metastatic esophageal and gastroesophageal junction cancers. Cetuximab was added to each treatment arm based on promising preclinical data. Patients and Methods Patients with previously untreated metastatic esophageal or gastroesophageal junction cancer were randomly assigned at a one-to-one-to-one ratio to epirubicin, cisplatin, and continuous-infusion fluorouracil (ECF), irinotecan plus cisplatin (IC), or FOLFOX (oxaliplatin, leucovorin, and bolus and infusional fluorouracil). All treatment programs included cetuximab once per week. The primary end point was response rate. Secondary outcomes included overall survival, progression-free survival, time to treatment failure, and safety. As prespecified, primary and secondary analyses were conducted only among patients with adenocarcinoma. Results This study randomly assigned 245 patients, including 222 with adenocarcinoma. Among patients with adenocarcinoma, response rate was 60.9% (95% CI, 47.9 to 72.8) for ECF plus cetuximab, 45.0% (95% CI, 33.0 to 57.0) for IC plus cetuximab, and 54.3% (95% CI, 42.0 to 66.2) for FOLFOX plus cetuximab. Median overall survival was 11.6, 8.6, and 11.8 months; median progression-free survival was 7.1, 4.9, and 6.8 months; and median time to treatment failure was 5.6, 4.3, and 6.7 months for each of these arms, respectively. FOLFOX plus cetuximab required fewer treatment modifications compared with ECF plus cetuximab and IC plus cetuximab (P = .013), and fewer patients were removed from treatment because of an adverse event or experienced treatment-related death. Conclusion In combination with cetuximab, ECF and FOLFOX had similar efficacy, but FOLFOX was better tolerated. Although differences were nonsignificant, IC plus cetuximab seemed to be the least effective and most toxic of the three regimens tested.


2015 ◽  
Vol 33 (28) ◽  
pp. 3193-3198 ◽  
Author(s):  
Troy Z. Horvat ◽  
Nelly G. Adel ◽  
Thu-Oanh Dang ◽  
Parisa Momtaz ◽  
Michael A. Postow ◽  
...  

Purpose Ipilimumab is a standard treatment for metastatic melanoma, but immune-related adverse events (irAEs) are common and can be severe. We reviewed our large, contemporary experience with ipilimumab treatment outside of clinical trials to determine the frequency of use of systemic corticosteroid or anti-tumor necrosis factor α (anti-TNFα) therapy and the effect of these therapies on overall survival (OS) and time to treatment failure (TTF). Patients and Methods We reviewed retrospectively the medical records of patients with melanoma who had received treatment between April 2011 and July 2013 with ipilimumab at the standard dose of 3 mg/kg. We collected data on patient demographics, previous and subsequent treatments, number of ipilimumab doses, irAEs and how they were treated, and overall survival. Results Of the 298 patients, 254 (85%) experienced an irAE of any grade. Fifty-six patients (19%) discontinued therapy because of an irAE, most commonly diarrhea. Overall, 103 patients (35%) required systemic corticosteroid treatment for an irAE; 29 (10%) also required anti-TNFα therapy. Defining TTF as either starting a new treatment or death, estimated median TTF was 5.7 months. Twelve percent of patients experienced long-term disease control without receiving additional antimelanoma therapy. OS and TTF were not affected by the occurrence of irAEs or the need for systemic corticosteroids. Conclusion IrAEs are common in patients treated with ipilimumab. In our experience, approximately one-third of ipilimumab-treated patients required systemic corticosteroids, and almost one-third of those required further immune suppression with anti-TNFα therapy. Practitioners and patients should be prepared to treat irAEs and should understand that such treatment does not affect OS or TTF.


Sign in / Sign up

Export Citation Format

Share Document