scholarly journals Case Report: Anlotinib Therapy in a Patient With Recurrent and Metastatic RAIR-DTC Harboring Coexistent TERT Promoter and BRAFV600E Mutations

2021 ◽  
Vol 11 ◽  
Author(s):  
Yanjun Su ◽  
Shaohao Cheng ◽  
Jun Qian ◽  
Min Zhang ◽  
Tuanli Li ◽  
...  

We describe a case of recurrent and metastatic radioactive iodine-refractory differentiated thyroid cancer (RAIR-DTC) treated with anlotinib in this report. The patient was randomized to placebo initially, after disease progressed at C8 (C is the treatment cycle), the patient was referred to the open label therapy of anlotinib, 12 mg p.o. daily with a 2-week on/1-week off regimen. Partial response was achieved at C2 with anlotinib treatment. To date, over 37 months of progression-free survival (PFS) has been achieved. Adverse effects were tolerable and manageable in this patient. Molecular characterization revealed coexistent C228T mutation of TERT promoter and BRAFV600E mutations. Favorable clinical outcome in this patient suggests that anlotinib might provide a novel effective therapeutic option for patients with RAIR-DTC. TERT and BRAFV600E mutations may represent as biomarker for predicting salutary effects of anlotinib.

2012 ◽  
Vol 30 (4_suppl) ◽  
pp. 503-503
Author(s):  
Sun Young Kim ◽  
Mi Song I Jang ◽  
Hyun Mi Kim ◽  
Ji Yeon Baek ◽  
Yong Sang Hong ◽  
...  

503 Background: The three-drug regimen with oxaliplatin, irinotecan and fluoropyrimidine is an effective therapeutic option for patients with mCRC, but is associated with high rate of toxicity. Pharmacogenetic profile might be helpful for selection of proper patients for this intensive regimen. Methods: Forty-two patients were enrolled to our phase II trial and were given irinotecan 150mg/m2 plus oxaliplatin 85mg/m2 on D1 and S-1 80mg/m2/day on D1-14 every 21 days as their front-line therapy for mCRC. Genomic DNA was extracted from peripheral blood, where the presence of germline polymorphisms of UGT including UGT1A1*6, UGT1A1*28, UGT1A1*60, UGT1A6*2, and UGT1A7*3 were tested. Results: Patients with UGT1A1*6 allele had a tendency of more frequent grade 2-3 vomiting (p = 0.06) compared to those without UGT1A1*6. The presence of a haplotype containing UGT1A6*2 and UGT1A7*3 was associated with grade 2-3 vomiting (p = 0.014) and grade 2-3 diarrhea (p = 0.063). Higher objective response rate (18/20, 90%) was noted in patients without UGT1A*60 compared to those with UGT1A1*60 (11/22, 50%; p = 0.008). The absence of UGT1A1*60 was also associated with marginally improved progression-free survival (10.3 mo v 7.7 mo, p = 0.081) and overall survival (26.8 mo v 15.1 mo, p= 0.044) compared to the presence of the variant allele. Conclusions: UGT1A1*6 and a haplotype containing UGT1A6*2 and UGT1A7*3 may be associated with irinotecan-related gastrointestinal toxicities. Phenotypic association of UGT1A1*60 and efficacy of the three-drug regimen requires further investigation.


Blood ◽  
2018 ◽  
Vol 132 (Supplement 1) ◽  
pp. 3265-3265
Author(s):  
Krystal Bergin ◽  
Flora Yuen ◽  
Craig Thomas Wallington-Beddoe ◽  
Anna Kalff ◽  
Shreerang Sirdesai ◽  
...  

Abstract Introduction: Despite improved survival outcomes in multiple myeloma (MM), disease relapse is inevitable and further therapeutic options are required. Therapy with bortezomib (a proteasome inhibitor (PI)), thalidomide and dexamethasone (VTD) has been shown to be highly effective in relapsed/refractory MM (RRMM). Ixazomib (an oral PI) in combination with lenalidomide and dexamethasone (IRD) has been shown to increase progression-free survival (PFS) in RRMM. Currently there are limited data on the efficacy/tolerability of the combination of ixazomib, thalidomide and dexamethasone (ITD). ITD may prove to be an attractive "all oral" therapeutic alternative particularly in jurisdictions with less access to high cost therapeutics. Aim: To evaluate the efficacy and safety of the combination of ixazomib, thalidomide and dexamethasone (ITD) in RRMM. Methods: This is a planned preliminary analysis of a multi-centre, single arm, open label, phase 2 study of the combination of ITD in RRMM patients with 1-3 prior lines of therapy. ITD (Ixazomib: 4mg orally D1, 8, 15, Thalidomide 100mg orally nocte D1-28, Dexamethasone 40mg orally D1, 8, 15, 22 of a 28-day cycle) was continued until disease progression or unacceptable toxicity. We plan to enrol 45 patients and to date data for 33 patients who have received 4 cycles of therapy or are off study were available for analysis. Baseline characteristics, therapy/safety, response and outcome data were analysed. The primary endpoint is overall response rate (ORR) defined as at least a partial response (PR). As specified in the protocol, we calculated the posterior distribution for ORR using the currently available data and a minimally informative prior distribution (Bayesian updating of the posterior could commence after 12 patients became evaluable) and we report a 95% credible interval (CI) for ORR and also for the clinical benefit rate (CBR) defined as at least a minimal response (MR). We also report an interim summary of PFS. Analyses were performed using R and SAS9.4. Results: Thirty-three patients were enrolled from 2 Australian sites (M:F 24:9), median age: 66 years (41-85). Median number of prior lines of therapy was 1 (1-3). Patients received a median of 6 cycles (1-20). Nine patients (27.3%) received <4 cycles. ISS stage at diagnosis: 1: 12, 2: 14, 3: 4 (3 unknown). Of patients with known cytogenetic/FISH data (n=22) 8 had high risk features (del17p, +1q21, t(14;16), t(4;14)). Twenty patients had IgG disease (IgA: 7, Light chain only: 6). The ORR was 18/33 (54.5%; 95% CI: 37.6 - 70.1%) including complete response (CR) in 2 patients (6.1%). The posterior probability that the true ORR exceeds 45% is currently 86%. Median time to best response was 83 days for responders and 99 days when non-responders were censored at their dates of last contact. The CBR was 23/33 (69.7%; 95% CI: 52.4 - 82.5%). The posterior probability that the true CBR exceeds 45% is currently > 99%. Both median PFS and OS have not been reached. Safety/tolerability: Fourteen patients (42.4%) tolerated ITD dosing as per protocol with thalidomide dose reduced/ceased in 15 patients (reduced: 8, ceased: 7) and dexamethasone dose reduced in 6 patients. No patients remaining on study have dose reduced/ceased ixazomib. Eighteen patients remain on study therapy with 15 ceasing therapy (progressive disease (PD): 9, adverse event (AE): 2, consent withdrawal: 2, lack of response: 1, new diagnosis of AML: 1). Twenty-two patients experienced a total of ninety-two (all-grade) AEs regardless of relatedness to study treatment with new/worsening peripheral neuropathy (12/92, 13.0%) and infection (11/92, 12.0%) most common. Two patients had grade 3 adverse events (epistaxis/thrombocytopenia:1, constipation: 1). Conclusion: The combination of ITD is efficacious and tolerable in RRMM and would represent an attractive all oral therapeutic option for RRMM particularly in jurisdictions with limited access to other therapeutics such as lenalidomide. Figure 1. Progression-free survival (PFS) and conventional 95% confidence intervals in months from the date of day 1 of the first cycle of ITD (FAS1). 1 Up to the cutoff date (23JUL2018) Disclosures Bergin: Celgene: Consultancy; AMGEN: Other: Travel to education meeting. Kalff:Janssen: Honoraria; Amgen: Other: travel to preceptorship; Celgene: Honoraria; Takeda: Honoraria. Reynolds:Novartis: Equity Ownership, Other: former employee of Novartis AG and holds stock in the company. .


2021 ◽  
Vol 11 (1) ◽  
pp. 37-46
Author(s):  
Ya. A. Zhulikov ◽  
E. I. Kovalenko ◽  
V. Yu. Bohyan ◽  
M. V. Khoroshilov ◽  
M. M. Gabrava ◽  
...  

Introduction. Adrenocortical carcinoma (ACC) is an orphan disease with an unfavorable prognosis. The most effective therapeutic option in the treatment of ACC is EDP plus mitotane combination chemotherapy. However, no studies comparing the efficacy of the EDP regimen with or without mitotane have been published.Materials and methods. A retrospective analysis of health records of patients with histologically confirmed metastatic ACC, who received at least one chemotherapy cycle with EDP ± mitotane. The study included 73 patients, 49 of which received a combination of EDP and mitotane and 24 were treated with EDP chemotherapy.Results. The objective response rate was 18,4 % in the EDP + mitotane group versus 4,1 % in the EDP group. Disease control was reported in 25 (51 %) and 13 (54,2 %) patients, respectively. No significant differences were found in progression-free survival (PFS) rates between the EDP and EDP + mitotane groups; the median PFS rate was 6,5 and 6,0 months, respectively. The median overall survival (OS) in the total population was 20,9 months; no significant differences were found between the groups. However, an increase in median PFS was observed in patients who achieved a therapeutic concentration of mitotane. Moreover, the achievement of therapeutic mitotane concentrations was the only factor significantly associated with improved PFS (HR 0.44, p = 0.006). Significant unfavorable prognostic factors associated with lower OS were Ki-67 level in the primary tumor > 20 % (HR 10.5, p = 0.006) and more than 1 site of metastases (HR 3.82, p = 0.02).Conclusions. This study showed that the addition of mitotane to EDP chemotherapy does not improve the median PFS and OS in the total patient population, however, the achievement of therapeutic mitotane concentrations is significantly associated with improved progression-free survival.


2014 ◽  
Vol 32 (3_suppl) ◽  
pp. LBA382-LBA382 ◽  
Author(s):  
Pompiliu Piso ◽  
Michael Koller ◽  
Dirk Arnold ◽  
Hans J. Schlitt ◽  
Gabriel Glockzin

LBA382 Background: Cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) provide a promising therapeutic option as part of an interdisciplinary multimodal treatment regimen for selected patients with peritoneal metastasis arising from colorectal cancer. The COMBATAC trial evaluates the feasibility, safety, and efficacy of perioperative cetuximab-containing systemic polychemotherapy, CRS and bidirectional oxaliplatin-based HIPEC. Methods: The COMBATAC trial is a prospective, multicenter, open-label, single-arm, single-stage phase II trial recruiting patients with histologically proven wild-type KRAS colorectal or appendiceal adenocarcinoma and synchronous or metachronous peritoneal metastasis. The planned total number of patients to be recruited is 60. The primary endpoint is progression-free survival (PFS). Results: Preliminary data of the first 16 documented patients show a total number of 51 adverse events. According to CTCAE v4 the distribution of AE was 23 grade 1, 14 grade 2, 13 grade 3, and one grade 4, respectively. Of the 14 grade 3/4 adverse events three were chemotherapy-related, four surgery-related, and seven not directly treatment-related. Most common grade 3/4 complications were infection, thrombosis, and urinary tract obstruction. Conclusions: First data indicate that perioperative systemic polychemotherapy including cetuximab in combination with CRS and bidirectional HIPEC is feasible and might not lead to increased morbidity and toxicity rates. Nevertheless, the COMBATAC trial is still recruiting, and data regarding progression-free survival (PFS) and overall survival (OS) is not yet available. Clinical trial information: NCT01540344.


Author(s):  
Takeshi Kato ◽  
Yoshinori Kagawa ◽  
Yasutoshi Kuboki ◽  
Makio Gamoh ◽  
Yoshito Komatsu ◽  
...  

Abstract Background We aimed to assess the safety and efficacy of combination treatment with panitumumab plus trifluridine/tipiracil (FTD/TPI) in patients with wild-type RAS metastatic colorectal cancer (mCRC) who were refractory/intolerant to standard therapies other than anti-epidermal growth factor receptor therapy. Methods APOLLON was an open-label, multicentre, phase 1/2 trial. In the phase 1 part, 3 + 3 de-escalation design was used to investigate the recommended phase 2 dose (RP2D); all patients in the phase 2 part received the RP2D. The primary endpoint was investigator-assessed progression-free survival (PFS) rate at 6 months. Secondary endpoints included PFS, overall survival (OS), overall response rate (ORR), disease control rate (DCR), time to treatment failure (TTF), and safety. Results Fifty-six patients were enrolled (phase 1, n = 7; phase 2, n = 49) at 25 Japanese centres. No dose-limiting toxicities were observed in patients receiving panitumumab (6 mg/kg every 2 weeks) plus FTD/TPI (35 mg/m2 twice daily; days 1–5 and 8–12 in a 28-day cycle), which became RP2D. PFS rate at 6 months was 33.3% (90% confidence interval [CI] 22.8–45.3). Median PFS, OS, ORR, DCR, and TTF were 5.8 months (95% CI 4.5–6.5), 14.1 months (95% CI 12.2–19.3), 37.0% (95% CI 24.3–51.3), 81.5% (95% CI 68.6–90.8), and 5.8 months (95% CI 4.29–6.21), respectively. Neutrophil count decreased (47.3%) was the most common Grade 3/4 treatment-emergent adverse event. No treatment-related deaths occurred. Conclusion Panitumumab plus FTD/TPI exhibited favourable anti-tumour activity with a manageable safety profile and may be a therapeutic option for pre-treated mCRC patients.


2017 ◽  
Vol 27 (2) ◽  
pp. 258-266 ◽  
Author(s):  
Patricia Pautier ◽  
Ignace Vergote ◽  
Florence Joly ◽  
Bohuslav Melichar ◽  
Elzbieta Kutarska ◽  
...  

ObjectiveAdvanced/metastatic or recurrent endometrial cancer has a poor prognosis. Malignant endometrial tissue has high steroid sulphatase (STS) activity. The aim of this study was to evaluate STS as a therapeutic target in patients with endometrial cancer.MethodsThis was a phase 2, multicenter, international, open-label, randomized (1:1), 2-arm study of the STS inhibitor oral irosustat 40 mg/d versus oral megestrol acetate 160 mg/d in women with advanced/metastatic or recurrent estrogen receptor–positive endometrial cancer. The primary end point was the proportion of patients without progression or death 6 months after start of treatment. Secondary end points included progression-free survival, time to progression, overall survival, and safety.ResultsSeventy-one patients were treated (36 with irosustat, 35 with megestrol acetate). The study was prematurely stopped after futility analysis. Overall, 36.1% and 54.1% of patients receiving irosustat or megestrol acetate had not progressed or died at 6 months, respectively. There were no statistically significant differences between irosustat and megestrol acetate in response and overall survival rates. Irosustat patients had a median progression-free survival of 16 weeks (90% confidence interval, 9.0–31.4) versus 40 weeks (90% confidence interval, 16.3–64.0) in megestrol acetate patients. Treatment-related adverse events occurred in 20 (55.6%) and 13 (37.1%) patients receiving irosustat or megestrol, respectively. Most adverse events in both groups were grade 1 or 2.ConclusionsAlthough irosustat monotherapy did not attain a level of activity sufficient for further development in patients with advanced/recurrent endometrial cancer, this study confirms the activity of hormonal treatment (megestrol acetate) for this indication.


2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Kuo-Chen Wei ◽  
Peng-Wei Hsu ◽  
Hong-Chieh Tsai ◽  
Ya-Jui Lin ◽  
Ko-Ting Chen ◽  
...  

AbstractAsunercept (company code APG101 [Apogenix AG]; company code CAN008 [CANbridge Pharmaceuticals]) is a novel glycosylated fusion protein that has shown promising effectiveness in glioblastoma. This Phase I study was initiated to evaluate the tolerability and safety of asunercept in combination with standard radiotherapy and temozolomide (RT/TMZ) in Asian patients with newly diagnosed glioblastoma. This was the Phase I portion of a Phase I/II open label, multicenter trial of asunercept plus standard RT/TMZ. Adults with newly-diagnosed glioblastoma received surgical resection followed by standard RT/TMZ plus asunercept 200 mg/week (Cohort 1) or 400 mg/week (Cohort 2) by 30-min IV infusion. The primary endpoint was the safety and tolerability of asunercept during concurrent asunercept and RT/TMZ; dose-limiting toxicities were observed for each dose. Secondary endpoints included pharmacokinetics (PK) and 6-month progression-free survival (PFS6). All patients (Cohort 1, n = 3; Cohort 2, n = 7) completed ≥ 7 weeks of asunercept treatment. No DLTs were experienced. Only one possibly treatment-related treatment emergent adverse event (TEAE), Grade 1 gingival swelling, was observed. No Grade > 3 TEAEs were reported and no TEAE led to treatment discontinuation. Systemic asunercept exposure increased proportionally with dose and showed low inter-patient variability. The PFS6 rate was 33.3% and 57.1% for patients in Cohort 1 and 2, respectively. Patients in Cohort 2 maintained a PFS rate of 57.1% at Month 12. Adding asunercept to standard RT/TMZ was safe and well tolerated in patients with newly-diagnosed glioblastoma and 400 mg/week resulted in encouraging efficacy.Trial registration NCT02853565, August 3, 2016.


2021 ◽  
Vol 39 (3_suppl) ◽  
pp. 270-270
Author(s):  
Masatoshi Kudo ◽  
Kazuomi Ueshima ◽  
Masafumi Ikeda ◽  
Takuji Torimura ◽  
Hiroshi Aikata ◽  
...  

270 Background: To date many trials have been conducted to compare the efficacy and toxicity between TACE plus molecular targeted agents and TACE alone; all of them failed to show its clinical benefit in terms of progression free survival (PFS) or OS. In TACTICS trial (NCT01217034) TACE plus sorafenib significantly improved PFS over TACE alone in patients with unresectable HCC. ( Gut 2020;69:1374-1376). Here we will report a final OS analysis from TACTICS trial with predefined mature OS events. Methods: Patients with unresectable HCC were randomized to TACE plus sorafenib (n=80) or TACE alone (n=76). Patients in the combination group received sorafenib 400 mg once daily for 2–3 weeks before TACE, followed by 800 mg twice daily during on-demand conventional TACE sessions until time to untreatable (unTACEable) progression (TTUP), defined as untreatable tumor progression, transient deterioration to Child-Pugh C or appearance of vascular invasion/extrahepatic spread. Co-primary endpoints were progression-free survival (PFS), which is defined as TTUP, or time to any cause of death and OS. Multiplicity was adjusted by gatekeeping hierarchical testing. Results: At the cut-off date of July 31, 2020, 131 OS events were observed. Median OS was 36.2 mo with TACE plus sorafenib vs 30.8 mo with TACE alone (HR, 0.861 [95%CI, 0.607, 1.223]; P=0.40). ΔOS was 5.4 mo. Updated PFS was 22.8 mo with TACE plus sorafenib vs 13.5 mo with TACE alone (HR, 0.661[95%CI, 0.466, 0.938]; P=0.02). Post-trial treatments with active procedures/agents were observed in 47 (58.8%) in TACE plus sorafenib and in 58 (76.3%) with TACE alone. Anticancer procedures in TACE alone group include resection/ablation in 14, transarterial therapy in 53 and radiation in 7. Anticancer medications in TACE alone include targeted agents in 40 (29 sorafenib, 5 regorafenib, 3 lenvatinib, 3 ramucirumab), other systemic chemotherapy in 5 and immunotherapy in 5. Safety was consistent with the primary analysis, with no new signals identified. Conclusion: In TACTICS, TACE plus sorafenib did not show OS benefit as compared with TACE alone although significantly better PFS was consistently observed. OS in TACE plus sorafenib in TACTICS trial showed the longest OS (36.2 mo) with the longest ΔOS (5.4 mo) as compared with the previous 5 TACE combination trials. The major reason for negative OS result was speculated that many post-trial active treatments were performed in control arm (76.3%), which implies that OS endpoint in TACE combination trial may not be feasible anymore in current era of sequential therapy with many active locoregional and systemic treatments. Clinical trial information: NCT01217034.


2020 ◽  
Author(s):  
Ke Cheng ◽  
Yu-Wen Zhou ◽  
Ye Chen ◽  
Zhi-Ping Li ◽  
Meng Qiu ◽  
...  

Abstract Background Irinotecan-based doublet chemotherapy strategy was standard second-line backbone treatment for patients with oxaliplatin‑refractory metastatic colorectal cancer(mCRC). The aim of this study was to evaluate tolerability and efficacy of raltitrexed combined with irinotecan biweekly administered as the second-line therapy for mCRC patients.Methods The study was a single-center, non-randomized, open-label phase II trial. Patients with mCRC after failure with first-line treatment of oxaliplatin and fluoropyrimidine or its derivatives were enrolled. Irinotecan (180 mg/m2) and raltitrexed (2.5 mg/m2) were given intravenously on day 1. Cycles were repeated every 2 weeks. The primary endpoint was progression-free survival, and the secondary endpoints included overall response rate, disease control rate, overall survival and treatment related adverse events. Results Between December 2012 and October 2016, 35 patients were enrolled. 33 and 35 patients were assessed for response and safety, respectively. The overall response rate (ORR) was 8.6 %, and the disease control rate (DCR) was 71.4%. The median progression-free survival (PFS) was 4.5 months (95% CI 3.8–5.2). The median overall survival was 12.0 months (95% CI 8.5–15.5). Four patients received conversion therapy to no evidence of disease (NED), and 2 patients were still alive with beyond 24 months survival. The most common grade 3/4 hematological adverse events were leukopenia (8.6%), neutropenia (5.7%). The most common grade 3/4 nonhematological adverse events were anorexia (14.3%), vomiting (14.3%), nausea (11.4%) and fatigue (8.6%). Two patients discontinued the protocol treatment because of treatment-related gastrointestinal adverse events. No one died from treatment-related events. The incidence and severity of toxicity was irrelevant to UGT1A1 status.Conclusions The combination of irinotecan with raltitrexed is an active, convenient and acceptable toxic regimen for second-line treatment for mCRC patients, which needs further study as a chemotherapy backbone to be combined with targeted agents in mCRC.Trial registration No. ChiCTR-ONC-12002767. The study was registered with the Chinese Clinical Trial Registry at 29 Octorber 2012, http://www.chictr.org.cn/index.aspx.


2020 ◽  
Vol 48 (12) ◽  
pp. 030006052097682
Author(s):  
Ping Zhang ◽  
Liangliang Ma ◽  
Xiaojie Wang ◽  
Ruijie Zhang ◽  
Yuting Dong

Ovarian cancer remains the most lethal gynecological malignant tumor, with relapse occurring in approximately 70% of advanced cases. Anlotinib is an oral small-molecule multi-targeted tyrosine kinase inhibitor that can resist neoangiogenesis and inhibit tumor growth. Previous research demonstrated clinical antitumor activity of anlotinib in various cancers. We report the case of an elderly woman with advanced ovarian cancer who received anlotinib after failure of multiple-line chemotherapy. A partial response was observed after six cycles of anlotinib monotherapy, with a reduction in the size of the metastases and significantly decreased serum CA125 levels from 1832.7 U/mL to 118.7 U/mL. She continued to take anlotinib, with a progression-free survival time of more than 4 months. Only mild hypertension was observed during the treatment. Anlotinib monotherapy may be a novel therapeutic option for patients with advanced ovarian cancer.


Sign in / Sign up

Export Citation Format

Share Document