GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: The Lancet, Elsevier BV, Vol. 402, No. 10408 ( 2023-09), p. 1133-1146
    Type of Medium: Online Resource
    ISSN: 0140-6736
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2067452-1
    detail.hit.zdb_id: 3306-6
    detail.hit.zdb_id: 1476593-7
    SSG: 5,21
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 4_suppl ( 2023-02-01), p. 578-578
    Abstract: 578 Background: Immune checkpoint inhibitors (CPIs) targeting PD-1/PD-L1 have become established treatments for advanced hepatocellular carcinoma (aHCC) but yield low objective response rates (ORRs) in treated patients (pts). Dual inhibition of LAG-3 and PD-1 pathways has demonstrated synergy in activating T-cells and improving immune response. Tebotelimab, also known as MGD013, is a bispecific tetravalent DART molecule that can bind both PD-1 and LAG-3. We initiated an open-label, single-arm, phase 1/2 dose escalation and expansion study to assess the safety and efficacy of tebotelimab in pts with aHCC. Methods: Eligible pts with aHCC who received ≥1 prior systemic treatment with or without prior CPI exposure were enrolled. The dose escalation phase evaluated doses at 120, 240, 400, and 600 mg. Tebotelimab was administered intravenously once every two weeks (Q2W) on days 1 and 15 of each 28-day cycle. The dose expansion phase consisted of one CPI-experienced cohort and one CPI-naïve cohort, both treated at recommended phase 2 dose (RP2D). Primary endpoints were safety for the escalation phase, and safety and ORR per RECIST v1.1 for the expansion phase. Investigator-assessed efficacy results are reported. Results: At data cut-off as of 27 April 2022, 13 pts received tebotelimab in the escalation phase. No dose-limiting toxicity was observed and RP2D was determined as 600 mg Q2W. In the expansion phase, 69 pts (CPI-experienced 33, CPI-naïve 36) were enrolled (median age, 57.0 years; male, 87.0%; ECOG 1, 58.0%; BCLC Stage C, 89.9%; and HBV etiology, 84.1%). Thirteen (18.8%) pts had Grade ≥3 treatment-related adverse events (TRAEs), most commonly hepatic function abnormal (n=3), amylase increased (n=2), and aspartate aminotransferase increased (n=2). Serious TRAEs occurred in nine (13.0%) pts, immune-related adverse events in 30 (43.5%), TRAEs leading to treatment discontinuation in five (7.2%), and treatment-related death in one (1.4%). Of the 30 evaluable pts in the CPI-experienced cohort, one achieved confirmed partial response (PR) and 14 achieved stable disease (SD), with a 3.3% ORR and a 50.0% disease control rate (DCR); of the 30 evaluable pts in the CPI-naïve cohort, four achieved confirmed PR and 10 achieved SD, with a 13.3% ORR and a 46.7% DCR. Median progression-free survival was 2.4 and 3.1 months for CPI-experienced and CPI-naïve cohorts, respectively, with median overall survival not reached in both. Conclusions: Tebotelimab demonstrated a manageable safety profile in pts with aHCC. Antitumor activity, mainly as disease stabilization, was observed in both the CPI-naïve setting and the CPI-experienced setting. No additional clinical trials are planned at this time. Clinical trial information: NCT04212221 .
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4089-4089
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4089-4089
    Abstract: Cholangiocarcinoma is a highly malignant tumor with limited therapeutic options. Gemcitabine (GMT) is currently used as a first line chemotherapeutical reagent for many cancers, including cholangiocarcinoma. However, it is often used in combination with other therapeutical strategies, due to high toxicity of GMT. We have previously reported that antagonists of calmodulin (CaM), including tamoxifen (TMX), trifluoperazine (TFP) and W7, induce apoptosis of cholangiocarcinoma cells and reduce cholangiocarcinoma tumorigenesis in mice. In the present studies, we determined the effect of combination therapy of TMX and GMT on cholangiocarcinoma tumorigenesis and investigated the mechanisms responsible for their efficacy. The effect of TMX and GMT on cholangiocarcinoma cell growth and apoptosis in vitro was determined by MTS and Annexin V staining analysis. The expression/activation of apoptotic signaling molecules were assessed by Western blot analysis. A nude mouse xenograft model was utilized for in vivo cholangiocarcinoma tumorigenesis as we previously reported. TMX (15mg/kg), GMT (15mg/kg) and combination of both were administrated by intraperitoneal injection. Tumor growth was measured every 3 days. Apoptosis in xenograft tumors was determined by TUNEL and cleaved caspase 3 staining. GMT inhibited cell growth and induced apoptosis of cholangiocarcinoma cells in a concentration-dependent manner. TMX enhanced GMT-induced apoptosis and GMT inhibition of cell growth in cholangiocarcinoma cells. GMT (15mg/kg, every third day) inhibited cholangiocarcinoma tumorigenesis in nude mice by 50%. TMX (15mg/kg, two out of three days) enhanced the inhibitory effect of GMX on tumorigenesis by 33%. The inhibition of tumor growth correlated with enhanced apoptosis in tumor tissues. To elucidate the mechanisms underlying the additive effects of TMX on GMT-induced apoptosis, we determined activation of caspases in cholangiocarcinoma cells exposed to GMT, TMX or both. Activation of caspase 9 and caspase 3, as well as cytochrome c release to the cytosol were demonstrated in cells exposed to either reagent alone. By contrast, TMX but not GMT activated caspase 2. Pharmacologic inhibition of caspase 2 activation decreased TMX-, but not GMT-, induced activation of caspase 3 and apoptosis of cholangiocarcinoma cells. Similarly, activation of caspase 2 was found in tumors from TMX-treated mice, but not GMT-treated mice. Therefore, the enhanced effect of TMX on GMT-induced cholangiocarcinoma cell death is partially mediated by activation of caspase 2. TMX and GMT both induce apoptosis and inhibit cholangiocarcinoma tumorigenesis, which may be attributed to the activation of distinct apoptosis signals by TMX and GMT. Our studies provide in vivo evidence and molecular insight to support the use of TMX and GMT in combination as an effective therapy for cholangiocarcinoma. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4089. doi:10.1158/1538-7445.AM2011-4089
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: European Journal of Cancer, Elsevier BV, Vol. 148 ( 2021-05), p. 1-13
    Type of Medium: Online Resource
    ISSN: 0959-8049
    RVK:
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2021
    detail.hit.zdb_id: 1120460-6
    detail.hit.zdb_id: 1468190-0
    detail.hit.zdb_id: 82061-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 3275-3275
    Abstract: Background: GLS-010 is a novel fully human anti-PD-1 mAb developed by the OMT transgenic rat platform. In Phase 1a study, GLS-010 exhibited good tolerance and 240mg (Q2W) was selected. Phase 1b study was conducted to evaluate the safety,anti-tumor activity and explore biomarkers of GLS-010 in pts with advanced solid tumors or lymphomas. Methods: All pts enrolled received GLS-010 240 mg every 2 weeks. Tumor response was assessed by RECIST 1.1 every 8 weeks. Adverse events (AEs) were graded by NCI CTCAE v4.03. Several biomarkers were explored, including PD-L1 by SP263 assay, tissue tumor mutation burden (tTMB) by whole exome sequencing (WES) from FFPE tissue, blood TMB (bTMB) by multi-gene panel based next-generation sequencing (NGS) from blood ctDNA. Results: As of 19 JUL 2019, 213 pts, median age of 55 (range: 21-75) years, were enrolled and 109 of 213 pts were still in treatment. The median dosing number was 7.5 (range: 1~41). Treatment-related AEs (TRAEs) occurred in 185 patients (70%), of which mostly were CTCAE grade 1-2. The most frequent TRAEs were related to hepatotoxicity, included “ALT increased” (32/213), “AST increased” (32/213), “blood bilirubin increased” (25/213). 53 of 163 pts, who received ≥1 response evaluation, achieved response (PR+CR, unconfirmed response included), including GC (4/21), EC (5/25), BTC (3/10), NSCLC (8/32), nasopharynx cancer (10/26),UC (2/8), HCC (0/12), cHL (18/21) and peripheral T/NK cell lymphoma (3/8). In all solid tumor types, pts with PD-L1-positive tumors experienced clinical benefit with significantly higher ORR (39.6% vs 14.5%, p=0.0025) and longer PFS (p=0.0241). ORR was higher for PD-L1 positive (TC≥25%) pts with lung cancer (66.7% vs 20.0%, p=0.051). For pan-caner analysis, ORR benefits of GLS-010 were also enhanced (51.9% versus 26.4%, p=0.0298) in pts with tTMB-high ( & gt;75th percentile, of each tumor type) versus tTMB-low. Pairing tTMB and survival data were analyzed in 129 pts, and tTMB-high pts benefit more with a significantly improved PFS (p=0.011). Also, improved PFS was observed in tTMB-high pts with EC (p=0.0059). For bTMB-high group, pts with EC achieved higher ORR (42.9% vs 6.3%, p=0.0672), which translated into PFS benefits (p=0.03). Conclusion: In conclusion, it is suggested that GLS-010 was well tolerated and had durable antitumor activity in Chinese tumor pts. PD-L1 has showed to be predictive of GLS-010 activity in solid tumors, especially in lung cancer. For pan-cancer analysis, it is preliminarily demonstrated that tTMB may be valuable biomarkers to predict the treatment response and benefit of GLS-010. For Chinese EC pts, tTMB and bTMB may be of value in predicting the response to PD-1 inhibitor. Citation Format: Lin Shen, Jifang Gong, Yuqin Song, Dingwei Ye, Zhihao Lu, Siyang Wang, Peijian Peng, Jianhua Chen, Ou Jiang, Guojun Zhang, Yuxian Bai, Jianji Pan, Chunguang Ma, Li Chen, Yi Ba, Qi Li, Ping Lu, Lingli Zhang, Xianli Yin, Shanzhi Gu, Huilai Zhang, Hang Su, Yongsheng Jiang, Bangwei Cao, Weiqing Han, Yan Sun, Feng Zhang, Weiwei Ouyang, Haiying Dong, Jianming Guo, Yabing Guo, Chongyuan Xu, Junyuan Qi, Li Wang, Jun Lv, Xiang Wang, Chris Chen, Jing Li, Yong Zheng, Ge Jin, Yining Yang, Guodong Zhao, Fan Yang, Kehui Xu, Xiangying Liang, Zhaoyang Pan, Haijin Meng. GLS-010, a novel fully human anti-PD-1 mAb in patients with advanced tumor: Preliminary results of a Phase Ib clinical trial [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 3275.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2011
    In:  Cancer Research Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4776-4776
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 4776-4776
    Abstract: Pancreatic cancer remains a devastating malignancy with poor prognosis, which is largely resistant to current therapies. Death receptor-initiated apoptotic pathways have been implicated both in pancreatic cancer pathogenesis and therapy. To understand the resistance of pancreatic tumor to Fas death receptor-induced apoptosis, we investigated the molecular mechanisms of Fas-activated survival signaling in pancreatic cancer cells. Using lentivirus-mediated RNA interfering strategy, we generated pancreatic cancer cell lines MiaPaCa-2 and BxPc-3 with stable knockdown of the Fas associated protein with death domain (FADD), the adaptor that mediates downstream signaling upon Fas activation. Knockdown of FADD rendered these Fas-sensitive pancreatic cells resistant to apoptosis induced by the Fas agonist antibody, CH-11. Consistently, CH-11 induced recruitment of caspase-8 into the Fas-activated death inducing signaling complex was blocked in FADD knockdown cells, which was associated with no activation of caspase-8 and caspase-3. By contrast, CH-11 promoted the survival of the FADD knockdown MiaPaCa2 and BxPc3 cells in a concentration-dependant manner, as measured by MTS assay. Increased phosphorylation of ERK was demonstrated in the FADD knockdown cells in response to CH-11 treatment. The pharmacological inhibitor of ERK, PD98059, abrogated CH-11-promoted cell survival in FADD knockdown MiaPaCa-2 and BxPC-3 cells. Furthermore, increased phosphorylation of Src was demonstrated in FADD knockdown cells in response to CH-11. The Fas-induced activation of Src was blocked by the specific Src inhibitor, PP2, which further inhibited the Fas-induced activation of ERK and cell survival of the FADD knockdown cells. To elucidate the mechanisms underlying Fas-induced Src activation, we analyzed Fas-associated proteins by immunoprecipitaion of Fas in the FADD knockdown cells. Increased Src and phosphorylated Src was indentified in the Fas-associated protein complex activation by CH-11. Concomitantly, increased recruitment of calmodulin (CaM) into the Fas-associated protein complex was demonstrated as well. Trifluoperazine, a CaM antagonist, inhibited the Fas-induced recruitment of CaM, Src and phosphorylated Src. Consistently, trifluoperazine inhibited Fas-induced activation of Src and blocked Fas-promoted cell survival in the FADD knockdown cells. These results demonstrate that Fas induces FADD-independent survival signaling in pancreatic cancer cells via CaM-mediated activation of Src-ERK signaling pathways. Understanding the molecular mechanisms responsible for the resistance of pancreatic cells to apoptosis induced by Fas-death receptor signaling may provide molecular insights into designing novel therapies to treat pancreatic tumors. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 4776. doi:10.1158/1538-7445.AM2011-4776
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 40, No. 4_suppl ( 2022-02-01), p. TPS487-TPS487
    Abstract: TPS487 Background: In 2020, there were ̃905,000 new cases and ̃830,000 deaths from liver cancer globally, and a respective 410,038 and 391,152 in China (5-year overall survival [OS]: 14.1%). 1,2,3 Hepatocellular carcinoma (HCC) accounts for 75%–85% of liver cancers. 1 Transarterial chemoembolization (TACE) is the preferred treatment for patients with intermediate-stage unresectable HCC (uHCC); however, there are issues with the need for repeated dosing and the heterogenous target population (median OS: 13–43 mo). Treatment with TACE + systemic therapy may improve this. Although several trials reported negative results for TACE + sorafenib (sor) vs TACE alone in patients with uHCC, the TACTICS trial showed improved untreatable (unTACEable) progression-free survival (PFS) (25.2 vs 13.5 mo; P= 0.006). 4 While promising, the unmet need for combination therapies remains. The recent IMbrave150 trial showed significantly improved OS with atezolizumab (atezo) + bevacizumab (bev) vs sor (19.2 vs 13.4 mo; P= 0.0009) in patients with uHCC 5 , making it an attractive option for combination with TACE. Methods: TALENTACE (NCT047126430) is a phase III, open-label, randomized study to evaluate the efficacy and safety of TACE + atezo + bev or TACE alone in patients with uHCC. Key inclusion criteria are ≥18 years old, HCC diagnosis, no prior systemic treatment, no prior locoregional treatment of target lesions, eligible for TACE treatment, tumor max diameter + tumor number ≥6, and not a candidate for curative therapy. Patients with tumors that have macrovascular invasion (MVI) or extrahepatic spread (EHS) are excluded. Patients will be randomized (1:1) to receive TACE + atezo + bev or TACE alone. TACE will be performed by clinical demand; atezo (1200 mg) and bev (15 mg/kg) will be administered by intravenous infusion on Day 1 of each 21-day cycle. Co-primary endpoints are independent review committee-determined TACE-PFS (randomization to unTACEable progression, TACE failure/refractoriness, or death) and OS. Secondary endpoints include investigator-determined TACE-PFS; time to unTACEable progression, progression, MVI, EHS, and MVI/EHS; objective response rate, duration of response, patient reported outcomes, and incidence of adverse events. References: Sung, H., et al. CA Cancer J Clin. 2021;71(3):209–249. World Health Organisation: Globocan 2020 – China Factsheet. Available at: https://gco.iarc.fr/today/data/factsheets/populations/160-china-fact-sheets.pdf [accessed 1 June 2021] Allemani, C., et al. Lancet. 2018, 391(10125): 1023–1075. Kudo, M., et al. Gut. 2020;69(8):1492–1501. Finn, RS., et al. J Clin Oncol. 2021;39(no. 3_suppl):267–267. Clinical trial information: NCT04712643.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2022
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. e16106-e16106
    Abstract: e16106 Background: Porustobart (HBM4003) is a fully human heavy chain only monoclonal antibody (HCAb) targeting CTLA-4. In addition to blocking the CTLA-4 pathway, it has been shown to effectively deplete intratumoral T reg cells in patients due to its engineered enhanced antibody-dependent cellular cytotoxicity (ADCC). Toripalimab is a recombinant, humanized anti-PD-1 monoclonal antibody. Here we reported the results of a phase Ib study that evaluated porustobart plus toripalimab in patients with hepatocellular carcinoma (HCC). Methods: This is a phase Ib dose expansion study (NCT05149027). Patients with advanced HCC (n=28) received porustobart 0.45 mg/kg plus toripalimab 240 mg every three weeks (Q3W) in both Cohort 1 and Cohort 2. Cohort 1 recruited pts who failed previous anti-VEGFR multikinase inhibitor(s) treatment while have not received anti-PD-(L)1 treatment (n=16); Cohort 2 recruited patients who failed previous anti-PD-(L)1 and anti-VEGF(R) treatments (n=12). The primary endpoint was ORR per RECIST 1.1. Results: As of 9 Dec 2022, median follow-up time was 3.6 months (range: 1-7 months). In Cohort 1, ORR and DCR were 46.7% (95%CI: 21.3-73.4) and 73.3% (95%CI: 44.9-92.2) respectively in 15 patients with post-treatment tumor assessments. In Cohort 2, the ORR and DCR were 9.1% (95%CI: 0.2-41.3) and 54.5% (95%CI: 23.4-83.3) respectively in 11 patients with post-treatment tumor assessments. PFS or OS data were not mature by the cut-off date. Treatment-related adverse events (TRAEs) were reported in 89.3% (25/28) patients, and Grade 3 TRAEs were reported in 39.3% (11/28) patients. The most common (≥20%) TRAEs with all grades by pooled term were liver function test abnormal 46.4% (13/28), thrombocytopenia 42.9% (12/28), pyrexia 32.1% (9/28), asthenia 28.6% (8/28), anemia 25% (7/28), decreased appetite 25% (7/28), blood bilirubin increased 21.4% (6/28), leucopenia 21.4% (6/28), lymphopenia 21.4% (6/28), nausea 21.4% (6/28) and neutropenia 21.4% (6/28). TRAEs leading to permanent discontinuation of porustobart were reported in 3 (10.7%) patients. No Grade 4 or Grade 5 TRAE was reported. Porustobart promoted the clearance of T reg cells and the proliferation of CD4 + T cells and CD8 + T cells in peripheral blood that attested to its MOA. Greater effects were observed in Cohort 1. Taken together with the clinical observations these results suggested that PD-(L)1 inhibitor naïve patients have a larger available pool of effectors to induce anti-tumor activity in the presence of effective T reg depletion. No noticeable differences in PK between Cohort 1 and Cohort 2 were observed. Conclusions: Porustobart 0.45 mg/kg in combination with toripalimab 240mg Q3W showed promising anti-tumor activity in patients with advanced HCC, as well as an acceptable safety profile. Clinical trial information: NCT05149027 .
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 41, No. 16_suppl ( 2023-06-01), p. 4077-4077
    Abstract: 4077 Background: Cancer immunotherapy has expanded the treatment options for advanced HCC (aHCC). Atezolizumab plus bevacizumab is approved as the standard of care for patients (pts) with aHCC. QL1706 is a novel dual immune checkpoint blockade containing a mixture of anti-PD-1 IgG4 and anti-CTLA4 IgG1 antibodies (Abs). QL1604 is a humanized mAb against PD-1. The anti-PD-1 Ab of QL1706 has the same protein sequence and was produced by using the same single clone selection method with QL1604. Here we report the safety and efficacy results from a Ph Ib/II study of QL1706 or QL1604 plus bevacizumab (beva) as first-line treatment in pts with aHCC. Methods: Eligible pts had histologically or clinically confirmed HCC; had BCLC stage B/C disease and Child-Pugh class A and B liver function; were systemic therapy naive; and were not suitable for radical surgery and/or locoregional therapy, or progressed after surgery and/or locoregional therapy. This study included 3 parts. Part 1 included a safety run-in phase and an expansion phase. Patients were enrolled to receive QL1706 5 mg/kg plus beva 15 mg/kg Q3W. In part 2, pts were randomized to receive QL1604 200 mg or QL1706 5 mg/kg plus beva 15 mg/kg Q3W. In part 3, pts were enrolled to receive QL1706 7.5 mg/kg plus beva 15 mg/kg Q3W. Initiation of part 3 was to be determined based on factorial analysis on the results from parts 1 and 2. Each treatment cycle lasted for 21 days. Treatment continued until disease progression or other discontinuation events. The primary endpoint was safety. Secondary endpoints included efficacy etc. Results: As of data cutoff (18 Nov 2022), 76 pts were enrolled in parts 1 and 2 (QL1706: 50; QL1604: 26). The baseline characteristics were balanced between the 2 groups. A total of 43 (86%) pts and 23 (88.5%) pts in QL1706 and QL1604 groups experienced TRAEs. For both groups (QL1706 vs QL1604), the most common TRAEs were platelet count decreased (26% vs 23.1%); followed by AST increased (22% vs 19.2%). A total of 17 (34%) pts and 10 (38.5%) pts in QL1706 and QL1604 groups experienced Gr≥3 TRAEs. A total of 25 (50%) pts and 5 (19.2%) pts in QL1706 and QL1604 groups experienced irAEs. The most common irAE in the QL1706 group was rash (16%) (vs 3.8% for QL1604). A total of 8 (16%) pts and 6 (23.1%) pts in QL1706 and QL1604 groups experienced TRSAEs. In the efficacy evaluable population, the ORR was 38.3% (18/47) and 15.4% (4/26) in QL1706 and QL1604 groups. The DCR was 74.5% and 69.2% in QL1706 and QL1604 groups. The mPFS was 6.7 months (95% CI: 3.0-11.4) and 5.4 months (95% CI:2.4-8.5) in QL1706 and QL1604 groups. The mOS was not reached. Conclusions: QL1706 5 mg/kg showed comparable safety profile and numerically higher ORR and longer mPFS vs QL1604 when combined with beva as first-line treatment in pts with aHCC. Ph III head-to-head clinical trial to compare QL1706 or anti-PD-1/PD-L1 Ab plus beva in this population has been planned. Clinical trial information: NCT05603039 .
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2023
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: Journal of Clinical Oncology, American Society of Clinical Oncology (ASCO), Vol. 39, No. 15_suppl ( 2021-05-20), p. 4077-4077
    Abstract: 4077 Background: Many advanced hepatocellular carcinoma (aHCC) patients (pts) are often with more complicated clinical conditions such as damaged liver or blood function, poor physical conditions. Those aHCC pts are not suitable for molecular target drug like sorafenib or systemic chemotherapy and no standard or generally accepted treatment. Icaritin, a single molecule ( 〉 98% purity) derived from Epimedii herba (Traditional Chinese herbal medicine), is a novel immune-modulation anti-tumor agent. Preclinical studies demonstrated that Icaritin induced anti-HCC activities through targeting IL-6/JAK//STAT3 pathways and modulating inflammation-immune systems including Th1 cytokines, and down-regulation of alpha-fetoprotein (AFP). Prior phase II study demonstrated favorable overall survival (OS) improvement in aHCC pts with poor conditions and correlated with the combined serum biomarkers. The current phase III study was designed to confirm above clinical benefits and safety of Icaritin in those patients. Methods: An adaptive enrichment design was used in a multicenter randomized, double-blinded study of comparing Icaritin with Huachashu (a TCM formula commonly used in China) as first line therapy for those aHCC pts (NCT03236636). The primary endpoint was overall survival (OS) and secondary endpoints included time-to-progression (TTP), progression-free-survival (PFS), disease control rate (DCR), and safety. The pts were randomized (1:1) to receive either Icaritin at 600mg or Huachashu. Based on prior studies, a composite biomarker score (CBS) of AFP(≥400 ng/mL), TNF-a( 〈 2.5 pg/mL) and IFN-g(≥7.0 pg/mL) was used for pts selection and a CBS score of 2/3 was predefined positive. Patients with CBS-positive were applied in interim analysis according to the protocol and statistical analysis plan (SAP). Results: A total of 283 aHCC pts were enrolled and randomized from Sept. 2017, and 71 enriched pts was CBS-positive with combined risk/poor prognosis factors such as BCLC stage C, HBV infection, and thrombocytopenia etc.. Thirty-three and 38 CBS-positive aHCC pts were treated with Icaritin or Huachashu, respectively. With a median follow-up of 8.1 mo (cutoff date, Dec.30,2020), the treatment outcomes for Icaritin and Huachashu arm showed following, that is mOS, 13.54 vs. 7.06 mo (HR = 0.40, 95%CI 0.21-0.77, p = 0.0046), mTTP, 3.65 vs. 1.84 mo (HR = 0.67, 95%CI, 0.36-1.22), mPFS, 2.79 vs. 1.84 mo (HR = 0.75, 95%CI, 0.43-1.33), and DCR, 48.5% vs. 26.3, respectively. Treatment-related adverse event (AE≥3 grades) observed were 15.2% vs. 31.6%, respectively. Conclusions: Small molecule immunomodulation agent Icaritin could significantly improve the overall survival with favorable safety in a prospectively CBS-enriched HBV-related advanced HCC pts with poor conditions. Clinical trial information: NCT03236636.
    Type of Medium: Online Resource
    ISSN: 0732-183X , 1527-7755
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Clinical Oncology (ASCO)
    Publication Date: 2021
    detail.hit.zdb_id: 2005181-5
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...