肿瘤瞭望消化时讯 发表时间:2024-10-14 14:33:46
编者按:KRAS基因是结直肠癌中常见的致癌基因突变,约40%的CRC病例存在KRAS突变。KRAS基因突变携带者往往预后不佳。由于特定的空间结构特点,KRAS一直被认为是最难成药的突变基因。2021年5月,美国FDA宣布加速批准sotorasib上市,用于治疗肿瘤携带KRAS G12C突变的晚期非小细胞肺癌患者,打破了其不可成药的“魔咒”。目前,关于KRAS突变的靶向药物研究正在如火如荼的进行中。
2024年9月13~17日,欧洲肿瘤内科学会(ESMO)年会将于西班牙巴塞罗那正式召开。当地时间9月9日00:05大会官网公布了除LBA摘要以外的其他全部摘要内容。本文特此梳理并总结了本次ESMO年会上,在消化肿瘤领域内关于KRAS突变位点抑制剂的最新药物研究进展,以飨读者。
1.第二代KRAS G12C抑制剂D3S-001治疗KRAS G12C突变晚期/转移性实体瘤的Ⅰ/Ⅱ期研究
背景
方法
结果
结论
2.KRAS G12C阳性实体瘤患者接受单药Divarasib治疗的长期随访
背景
方法
结果
结论
3.Sotorasib(soto)、帕尼单抗(pani)和FOLFIRI在KRAS G12C突变转移性结直肠癌(mCRC)一线治疗中的安全性和有效性分析:来自Ⅰb期CodeBreaK 101研究的数据
背景
方法
结果
结论
4.ASP3082在晚期胰腺癌、结直肠癌和非小细胞肺癌成人患者中的初步安全性和临床活性研究:一种首创的KRAS G12D选择性蛋白降解剂
背景
方法
结果
结论
Speakers:Byoung Chul Cho (Seoul, Korea, Republic of)
D3S-001 is a 2nd generation KRAS G12C inhibitor (G12Ci) with faster target engagement kinetics, depletes cellular active G12C at nanomolar levels, a MoA relevant to stronger anti-tumor activity (Cancer Discov. 2024 in press). Preliminary results of FIH dose escalation cohorts were reported at AACR 2024. Here we report the updated analyses.
In the ongoing Phase 1/2 study (NCT05410145), pts were administered D3S-001 (50-900 mg po daily) until intolerable toxicity or disease progression. Endpoints included safety and preliminary efficacy by investigators per RECIST v1.1.
As of 07 May 2024, a total of 42 pts (25 NSCLC, 13 CRC and 4 PDAC) were treated. Median follow-up was 6.8 months (m, range 0.2-18.3m) and 21 pts (50.0%) remain on-study treatment. No treatment-related DLTs or deaths during assessment period. TRAE of any grade occurred in 32 pts (76.2%). Of which, 7 (16.7%) were Grade 3 (no ≥ Grade 4) TRAEs. D3S-001 dose was modified in 12 pts (28.6%) due to TRAE, and none had dose discontinuation. In all 42 dosed pts, 38 had post-baseline tumor assessments, of which 34 were naïve to G12Ci and 4 were G12Ci pre-treated. In 34 G12Ci naïve pts (21 NSCLC, 9 CRC and 4 PDAC), PRs were achieved in 26/34 overall population (76.5%, 95% CI 58.8, 89.3%), with a confirmed ORR (cORR) of 70.6% (95% CI: 52.5%, 84.9%) and DCR of 97.1%. The 6m DoR rate was 70.5% (95% CI: 45.9%, 93.0%). The 6m PFS rate was 65.3% (95 CI%: 45.7%, 79.3%). Among 21 NSCLC pts, 15/21 (71.4%, 95% CI: 47.8, 88.7%) achieved PR, with a cORR of 66.7% (95% CI: 43.0%, 85.4%) and DCR of 100%. The 6m DoR rate was 70.0% (95% CI: 32.9%, 89.2%). The 6m PFS rate was 64.1% (95 CI%: 38.7%, 81.2%). Tumor shrinkage was observed in 2/6 NSCLC pts with stable brain metastases. Among 9 CRC pts, 7/9 (77.8%, 95% CI: 40.0%, 97.2%) achieved PR (all confirmed), with DCR of 88.9% and 6m DoR of 100%. Among 4 PDAC pts, 4/4 (100%) achieved PRs (3/4 confirmed). DCR of 100% achieved in 4 G12Ci pre-treated pts (2 NSCLC and 2 CRC). Over 90% plasma ctDNA clearance as early as at C1D8 was detected in 18 of 31 (58.1%) baseline ctDNA G12C positive pts.
D3S-001, a 2G G12Ci, demonstrated favorable tolerability and consistent promising efficacy across KRAS G12C-mutated NSCLC, CRC, and PDAC.
NCT05410145.
Speakers:Elena Garralda (Barcelona, Spain)
Divarasib (GDC-6036) is an oral, highly potent, and selective KRAS G12C inhibitor. We report the long-term safety and clinical activity of single-agent divarasib in patients with advanced KRAS G12C-positive solid tumors.
As part of an ongoing phase I study (NCT04449874), patients received single-agent oral divarasib 50-400 mg daily until intolerable toxicity or disease progression. Assessments included safety (NCI-CTCAE v5) and preliminary antitumor activity (RECIST v1.1). Confirmed response was defined as complete or partial response on 2 consecutive tumor assessments at ≥4 weeks apart.
As of 1 January 2024, 153 patients had received single-agent divarasib (non-small cell lung cancer [NSCLC]: n=65; colorectal cancer [CRC]: n=61; other solid tumors [OST]: n=27). Median treatment duration was 9.57 months (range: 0-37.3) and median lines of prior systemic therapy was 2 (range: 0-8). No patients had prior KRAS G12C inhibitor treatment. Among 43 patients (31 NSCLC, 8 CRC, and 4 OST) who continued divarasib beyond 1 year, 20 (47%) experienced a new-onset treatment-related adverse event (TRAE) after a year. One patient (2%) experienced a new-onset Grade 3 TRAE (weight decreased); no patients experienced new-onset Grade 4-5 TRAEs. One patient (2%) experienced a new-onset Grade 2 TRAE (nausea) that led to dose reduction. No new-onset TRAEs led to divarasib withdrawal. Preliminary activity, including objective response rate (ORR), progression-free survival (PFS), and duration of response (DOR), are presented in the table for patients receiving divarasib 400 mg. Table: 616MO
ORR, PFS and DOR for patients receiving divarasib 400 mg
Single-agent divarasib was well tolerated with few new-onset TRAEs after 1 year and continued to demonstrate durable clinical activity in patients with advanced KRAS G12C-positive solid tumors.
NCT04449874.
Speakers:Salvatore Siena (Milan, Italy)
In the phase 1b CodeBreaK 101 (NCT04185883) study, the addition of soto plus pani to FOLFIRI demonstrated an acceptable safety profile and a promising response rate of 60% for patients (pts) with previously treated KRAS G12C–mutated mCRC. Here, we evaluate the safety and efficacy of soto, pani, and FOLFIRI in treatment (Tx)-naïve pts with KRAS G12C–mutated mCRC.
Pts with KRAS G12C–mutated mCRC who had not received any prior systemic Tx for metastatic disease were enrolled at 17 global sites (between Jul 2021 and Mar 2024) and received the recommended phase 2 dose of soto (960 mg, oral, daily), pani (6 mg/kg, intravenous [IV], once every 2 weeks [Q2W]), and FOLFIRI (IV Q2W). Primary endpoint was safety and tolerability. Secondary endpoints included objective response rate, disease control rate, and median time to response.
Forty pts were enrolled and treated (58% male; median age: 60 years). Tx-related adverse events (TRAEs) of any grade occurred in 100% of pts, and grade ≥ 3 TRAEs occurred in 53% of pts; no fatal event was reported. The most common grade ≥ 3 TRAEs included neutropenia (23%), dermatitis acneiform (18%), and diarrhea (10%). 30 pts had confirmed partial responses, with an overall response rate of 75%. Treatment with soto, pani, and FOLFIRI resulted in a disease control rate of 93% and median time to response of 1.5 months. Table: 505O
∗Data are presented as n (%) unless indicated otherwise.
This study provides the first data set on the use of a KRASG12C inhibitor in 1L mCRC. The combination of soto, pani, and FOLFIRI demonstrated a tolerable safety profile and promising response rates in pts with Tx-naïve KRAS G12C–mutated mCRC. CodeBreaK 301 (NCT06252649), a phase 3 study, is currently enrolling to evaluate this combination in 1L mCRC against standard of care.
NCT04185883
Speakers:Wungki Park (New York, United States of America)
ASP3082 is a novel protein degrader selectively targeting KRAS G12D. Here, we describe the preliminary safety and antitumor activity of ASP3082 monotherapy in patients (pts) with previously treated advanced solid tumors.
In this phase 1, dose-escalation, first-in-human study, adults with unresectable or metastatic KRAS G12D-positive solid tumors were included. Pts received escalating doses of ASP3082 monotherapy (10–600 mg) intravenously once weekly over a 21-day cycle. Primary endpoints were incidence of dose-limiting toxicities (DLTs) and adverse events (AEs).
Data cutoff was 1 April 2024, with 98 pts enrolled (median age: 64 years; 56% male; median [range] prior lines of systemic therapy: 2 [1–7]) who had PC (n=67), CRC (n=16), NSCLC (n=13), or other cancers (n=2). Treatment-related AEs (TRAEs) occurred in 68/98 (69.4%) pts, including 5 pts with grade (Gr) 3 and 0 pts with Gr 4 or 5 events. TRAEs occurring in ≥5% of pts were fatigue (15.3%), infusion-related reactions (14.3%), pruritus (9.2%), nausea (7.1%), urticaria (7.1%), aspartate aminotransferase (AST) increased (7.1%), alanine aminotransferase (ALT) increased (6.1%), and vomiting (5.1%). DLTs were observed in 2 pts at 450 mg (Gr 3 ALT increased; Gr 3 ALT/AST increased) and 1 pt at 600 mg (Gr 3 ALT increased). The maximum tolerated dose has not been reached. Based on a preclinical modeling simulation, the predicted lowest dose for clinical efficacy was >100 mg. Efficacy evaluation included 65 pts receiving 10–300 mg. A total of 35 pts received ≤90 mg; objective response rate (ORR) was 0% and disease control rate (DCR) was 25.7%. At 140 mg (n=9), ORR was 11.1% (1 partial response [PR]/5 PC) and DCR was 33.3% (2/5 PC, 1/4 CRC). At 200 mg (n=9), ORR was 0% and DCR was 55.6% (3/7 PC, 1/1 NSCLC, 1/1 CRC). At 300 mg (n=12), ORR was 33.3% (3 PR/7 PC, 1 PR/4 NSCLC, 0 PR/1 CRC) and DCR was 75.0% (5/7 PC, 4/4 NSCLC, 0/1 CRC).
The ongoing study suggests that ASP3082, a novel KRAS G12D degrader, has an acceptable safety profile and promising antitumor activity, especially in pts with heavily pretreated PC. Based on these results, further studies are needed.
NCT05382559.