Skip to main content
Erschienen in:

Open Access 21.11.2024 | short review

Unraveling (K)RAS in pancreatic ductal adenocarcinoma

verfasst von: OA Dr. Bernhard Doleschal, PhD

Erschienen in: memo - Magazine of European Medical Oncology | Ausgabe 4/2024

Summary

Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal malignancy with a poor 5‑year survival rate. The majority of PDAC cases harbor KRAS mutations, predominantly at codon 12, with G12D being the most common. While selective inhibitors like sotorasib have shown promise in KRAS G12C-mutated PDAC, these mutations are rare, and resistance develops rapidly. Efforts to target more prevalent mutations like KRAS G12D are ongoing, with compounds such as MRTX1133 showing preclinical efficacy. Resistance mechanisms include secondary mutations and pathway reactivation, prompting the development of pan-(K)RAS inhibitors (e.g., RMC-6236) and combination strategies targeting upstream effectors. Novel approaches, such as KRAS-targeted vaccines and T‑cell receptor (TCR) therapies, offer additional potential. Continued clinical trials are crucial to optimizing KRAS-targeted therapies in PDAC.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

Pancreatic ductal adenocarcinoma (PDAC) remains one of the most lethal malignancies, with a persistently low 5‑year survival rate. In phase 3 randomized trials, median overall survival (OS) in the metastatic setting has not exceeded 12 months [1]. Advances in PDAC drug therapy have been limited, particularly with targeted agents and immunotherapy. Outside of germline BRCA mutations, few actionable alterations have been identified, leading to inconsistent adoption of molecular profiling in routine clinical practice for PDAC [2].

KRAS

Overview

One reason PDAC remains challenging to treat is the uniform molecular landscape of the majority (90%) of cases. The most common pathogenic drivers—KRAS, CDKN2A, TP53, and SMAD4—play critical roles in tumor initiation and maintenance. Mutations in KRAS, which occur early in the multistep genetic pathogenesis (PanINs), are the primary oncogenic drivers in PDAC, making it a KRAS-addicted cancer [3]. Most KRAS mutations occur at codon 12, with G12D being the most prevalent (35–40%), followed by G12V (20–30%), G12R (10–20%), Q61 (~5%), G12C (1–2%), and other rare mutations ([4]; Fig. 1).

Mode of action

The KRAS protein is a small GTPase that cycles between active (ON, GTP-bound) and inactive (OFF, GDP-bound) states. In its active state, KRAS promotes downstream signaling through RAS-binding domains (RBDs) of effector proteins, such as the mitogen-activated protein kinase (MAPK) RAF–MEK–ERK pathway and the phosphoinositide 3‑kinase (PI3K) pathways. The transition from the OFF to ON state is facilitated by guanine nucleotide exchange factors (GEFs), which reduce KRAS-GDP affinity and enable GDP to be replaced by GTP upon activation by extracellular signals and scaffolding proteins, such as Src homology‑2 domain-containing protein tyrosine phosphatase (SHP2). To revert to the OFF state, KRAS-GTP undergoes hydrolysis, a process that is intrinsically slow and requires the assistance of GTPase-activating proteins (GAPs; [5]).

Clinical implications of different KRAS mutations

Mutations in KRAS are associated with poorer overall survival (OS) compared with KRAS wild-type (WT) PDACs [4, 6]. The pathomechanism of many KRAS mutations involves altered intrinsic GTPase activity, which slows or eliminates the rate of GTP hydrolysis, keeping KRAS predominantly in the ON state. Additionally, the activation of downstream effector kinases varies depending on the specific KRAS mutation, potentially influencing prognostic outcomes. For example, OS is longer in KRAS G12R-mutated tumors than in KRAS G12D- or KRAS Q61-mutated tumors. KRAS G12D is more prevalent in metastatic disease, while KRAS G12R mutations are found more frequently in well-differentiated tumors. Furthermore, co-mutations may contribute to allele-specific clinical outcomes, such as ARID1A mutations being associated with worse prognosis [4, 6].

Targeting (K)RAS in PDAC

Mutant selective inhibitors

Targeting KRAS G12C

Although the KRAS G12C mutation is found in only 1.6% of PDAC patients, it is the only KRAS mutation for which selective inhibitors have reached clinical application. Historically, the KRAS protein was considered “undruggable” until 2013, when Shokat and colleagues identified a novel cysteine-containing switch II pocket in KRAS G12C [7, 8]. The CodeBreaK 100 trial, a multicenter phase I/II study, evaluated sotorasib, a selective covalent inhibitor of KRAS G12C. Among 38 patients with KRAS G12C-mutated metastatic PDAC, the confirmed objective response rate (ORR) was 21%, with a median progression-free survival (mPFS) of 4.0 months and a median overall survival (mOS) of 6.9 months [9]. The KRYSTAL-1 study investigating adagrasib showed an ORR of 33.3%, a disease control rate (DCR) of 81%, an mPFS of 5.4 months, and an mOS of 8 months for 21 PDAC patients [10]. Several other KRAS G12C inhibitors, such as divarasib, olomorasib, and glecirasib, are currently in (pre)clinical evaluation, demonstrating ORRs of up to 40% in pancreatic cancer cohorts (Table 1; [1113]). Further studies are needed to validate the clinical benefit of these targeted therapies in the PDAC population and to assess the efficacy of combining them with other treatments.
Table 1
Outcome of reported efficacy and safety data of various KRAS G12C inhibitors in PDAC
 
N
Phase
ORR
DCR
DOR
PFS
OS
TRAE
NCT
Sotorasib
38
I/II
21
84
5.7
4
6.9
TRAE: 57%
G3: 11.6%
ELE, Diarrhea, Anemia
NCT03600883 [9, 14]
Adagrasib
21
II
33
NR
5.3
5.4
8
TRAE: 97%
G3: 27%
Nausea, diarrhea, fatigue
NCT03785249 [10]
Divarasib
7
I
43
100
NR
NR
NR
TRAE: 93%
G3:12%
Nausea, diarrhea, vomiting
NCT04449874 [11]
Glecirasib
28
I/II
46
96
4.1
5.5
NR
TRAE:90%
G3: 25%
Anemia, ELE, diarrhea
NCT05002270 [12]
Olomorasib
24
I
33
92
NR
NR
NR
TRAE: 62%
G3: 5%
Diarrhea, fatigue, nausea
NCT04956640 [13]
HS-10370
NR
I
Responses observed
NR
NR
NR
NR
TRAE: 87%
G3: 27%
ELE, anemia, diarrhea
NCT05367778 [15]
MK-1084
1
I
100
NR
NR
NR
NR
TRAE: 57%
G3: 9%
ELE, diarrhea, pruritus
NCT05067283 [16]
ORR overall response rate, DCR disease control rate, DOR duration of response, PFS progression-free survival, OS overall survival, TRAE treatment-related adverse events, G grade, ELE elevated liver enzymes, NR not reported, NCT national clinical trial

Targeting other KRAS mutants

The most prevalent KRAS mutation in PDAC is KRAS G12D, making it a critical target for therapeutic intervention. However, the switch II pocket of the KRAS G12D protein lacks a reactive cysteine, making it more difficult to target with covalent inhibitors. In 2021 Wang et al. developed MRTX1133, a non-covalent KRAS G12D inhibitor based on the structure of adagrasib. Preclinical data demonstrate that MRTX1133 can reverse early PDAC growth and positively influence the tumor microenvironment (TME). Additionally, RMC-9805, a covalent tri-complex RAS(ON) G12D-selective inhibitor, has shown promise in preclinical PDAC models and was presented at AACR 2023 [17]. Moreover HRS-4642, another KRAS G12D inhibitor, has entered a first-in-human phase 1 trial, with early findings presented at ESMO 2023 [18].

Development of rapid resistance in mutant-specific targeting

The response duration to KRAS G12C inhibition in PDAC is typically modest, with resistance developing early. Acquired resistance to KRAS inhibition occurs primarily through three mechanisms [19, 20]:
1.
Acquisition of additional KRAS mutations: For example, in KRAS codons 12, 13, or 61, leading to enhanced RAS-MAPK pathway activation or disruption of binding at the switch II pocket.
 
2.
Bypassing KRAS inhibition through alterations in other RAS isoforms: Secondary mutations in other RAS family members (NRAS, HRAS) can bypass KRAS inhibition, sustaining oncogenic signaling.
 
3.
Alterations in downstream effector kinases: Mutations in downstream signaling components, such as BRAF, MEK1, NF1, PTEN, or PI3K, can also contribute to resistance by reactivating the signaling pathways despite KRAS inhibition.
 
In addition to acquired resistance mechanisms, different KRAS G12 mutations exhibit innate resistance to inhibitors based on the cycling velocity between ON and OFF states. Mutations with a very slow or negligible rate of GTP hydrolysis, such as KRAS G12V or G12D, may be “locked” in the ON state, potentially rendering them refractory to OFF-state KRAS inhibitors. ON-state inhibitors block effector engagement more rapidly, while OFF-state inhibitors may require longer to achieve complete target engagement and suppression, raising concerns about their efficacy in these cases [21].

Strategies to overcome mechanisms of acquired resistance

Targeting KRAS upstream pathways

Indirect pan-KRAS inhibition can be achieved by targeting upstream signaling proteins involved in KRAS activation, specifically SHP2 and SOS1 (Son of Sevenless 1). Several SHP2 inhibitors are in development, with some advancing to clinical trials. These include AB-331, presented at ASCO 2024, as well as RMC-4630, TNO155, and JAB-3068, which are being evaluated in phase I/II clinical trials (NCT03634982, NCT04330664, and NCT05288205; [22]). Additionally, BI1701963, a SOS1 inhibitor, is under investigation in a phase I trial to determine dosing as monotherapy and in combination with trametinib for KRAS-mutated solid tumors (NCT04111458).

Pan-KRAS inhibitors

Isoform-selective targeting of RAS may offer a promising approach by inhibiting all KRAS mutants, potentially addressing secondary activating KRAS mutations as well. BI-2865, the first non-covalent pan-KRAS inhibitor, has demonstrated preclinical efficacy against tumors harboring common KRAS alterations. BI-2865 specifically binds to the GDP-bound OFF state of both KRAS WT and its mutant variants, while sparing NRAS WT and HRAS WT isoforms, which may be important for maintaining tolerability in patients [23]. However, a critical challenge for the clinical application of pan-KRAS-selective inhibitors is the limited understanding of the effects of inhibiting KRAS WT in humans, raising concerns about potential adverse consequences.

Pan-RAS inhibitors

The primary challenge with pan-RAS inhibition is the potential toxicity from off-target effects on RAS WT in normal tissues, which could involve all three RAS isoforms (KRAS, HRAS, and NRAS). Preliminary data presented at ESMO 2023 from a phase I trial of the first-in-class pan/multi-KRAS inhibitor RMC-6236, a RAS-selective tri-complex, showed promising results in patients with previously treated metastatic PDAC (n = 46), with an ORR of 20% and a DCR of 87% [24]. Additionally, RMC-7977 has demonstrated efficacy across various preclinical pancreatic cancer models [25]. Both RMC-6236 and RMC-7977 function as tri-complex inhibitors that require cyclophilin A to target the GTP-bound active (ON) state of wild-type and mutant isoforms of KRAS, NRAS, and HRAS. Safety concerns might be mitigated by the enrichment of cyclophilin A in tumor cells or the ability of RMC-7977 to specifically target the GTP-bound ON state of RAS.

Other (K)RAS-directed approaches

Cellular therapies

Encouraging data are emerging for TCR therapy in highly select patients with PDAC. A case report of autologous T‑cell therapy expressing HLA-C * 08:02–restricted TCRs targeting KRAS G12D indicated efficacy in two patients with chemotherapy-refractory PDAC, with one patient maintaining a durable PR lasting more than 6 months [26]. There are several ongoing phase I/II clinical trials evaluating TCR therapy directed against KRAS G12V (NCT04146298, NCT03190941) and KRAS G12D (NCT03745326) in PDAC.

KRAS vaccines

Vaccination offers potential to expand endogenous KRAS G12-directed T cells across diverse HLA backgrounds, with simplified manufacturing and off-the-shelf availability. So far, conventional peptide vaccines suffered from poor immunogenicity. By contrast, modifying antigens—KRAS G12D and KRAS G12R peptides—and adjuvants—Toll-like receptor 9 agonists—with diacyl lipids that associate with fatty-acid-binding pockets on endogenous albumin after injection (creating Amph vaccines) results in efficient delivery into antigen-presenting cells. The first results in the adjuvant setting (ELI-002 2P vaccine; AMPLIFY-201 trial) led by Prof. O’Reilly from the Memorial Sloane Kettering Cancer Center showed a biomarker reduction in 16 out of 20 and MRD biomarker or circulating DNA (ctDNA) clearance in 3 out of 20 cases [27]. Vaccines utilizing mRNA technology for KRAS-mutated PDAC include mRNA-5671, which encodes KRAS G12D-, G12V-, G13D-, and G12C-specific peptides, are currently under investigation in combination with pembrolizumab in a phase I trial in KRAS-mutant advanced cancer (NCT03948763).

Conclusion

The optimal strategy for RAS targeting in pancreatic ductal adenocarcinoma (PDAC) remains unresolved. Mutant or isoform-selective KRAS inhibitors face challenges due to the emergence of de novo or acquired resistance. On the other hand, broad-spectrum pan-RAS (ON) inhibitors, which could be applicable to over 90% of PDAC patients, may disrupt physiological RAS wild-type signaling. The potential consequences and tolerability of such inhibition are still uncertain, highlighting the need for further investigation into balancing efficacy and safety in RAS-targeted therapies.

Conflict of interest

B. Doleschal declares that he/she has no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Unsere Produktempfehlungen

Abo für kostenpflichtige Inhalte

Literatur
1.
Zurück zum Zitat Hu ZI, O’Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2024;21(1):7–24.CrossRefPubMed Hu ZI, O’Reilly EM. Therapeutic developments in pancreatic cancer. Nat Rev Gastroenterol Hepatol. 2024;21(1):7–24.CrossRefPubMed
2.
Zurück zum Zitat O’Kane GM, Lowery MA. Moving the Needle on Precision Medicine in Pancreatic Cancer. J Clin Oncol. 2022;40(24):2693–705.CrossRefPubMed O’Kane GM, Lowery MA. Moving the Needle on Precision Medicine in Pancreatic Cancer. J Clin Oncol. 2022;40(24):2693–705.CrossRefPubMed
3.
Zurück zum Zitat Kanda M, Matthaei H, Wu J, Hong S, Yu J, Borges M, et al. Presence of Somatic Mutations in Most Early-Stage Pancreatic Intraepithelial Neoplasia. Gastroenterology. 2012;142(4):730–733.e9.CrossRefPubMed Kanda M, Matthaei H, Wu J, Hong S, Yu J, Borges M, et al. Presence of Somatic Mutations in Most Early-Stage Pancreatic Intraepithelial Neoplasia. Gastroenterology. 2012;142(4):730–733.e9.CrossRefPubMed
4.
Zurück zum Zitat Yousef A, Yousef M, Chowdhury S, Abdilleh K, Knafl M, Edelkamp P, et al. Impact of KRAS mutations and co-mutations on clinical outcomes in pancreatic ductal adenocarcinoma. Npj Precis Oncol. 2024;8(1):27.CrossRefPubMedPubMedCentral Yousef A, Yousef M, Chowdhury S, Abdilleh K, Knafl M, Edelkamp P, et al. Impact of KRAS mutations and co-mutations on clinical outcomes in pancreatic ductal adenocarcinoma. Npj Precis Oncol. 2024;8(1):27.CrossRefPubMedPubMedCentral
5.
Zurück zum Zitat Singhal A, Li BT, O’Reilly EM. Targeting KRAS in cancer. Nat Med. 2024;30(4):969–83.CrossRefPubMed Singhal A, Li BT, O’Reilly EM. Targeting KRAS in cancer. Nat Med. 2024;30(4):969–83.CrossRefPubMed
6.
Zurück zum Zitat Boilève A, Rousseau A, Hilmi M, Tarabay A, Mathieu JRR, Cartry J, et al. Codon-specific KRAS mutations predict survival in advanced pancreatic cancer. ESMOGastrointest Oncol. 2024;3:100030. Boilève A, Rousseau A, Hilmi M, Tarabay A, Mathieu JRR, Cartry J, et al. Codon-specific KRAS mutations predict survival in advanced pancreatic cancer. ESMOGastrointest Oncol. 2024;3:100030.
7.
Zurück zum Zitat Vasta JD, Peacock DM, Zheng Q, Walker JA, Zhang Z, Zimprich CA, et al. KRAS is vulnerable to reversible switch-II pocket engagement in cells. Nat Chem Biol. 2022;18(6):596–604.CrossRefPubMedPubMedCentral Vasta JD, Peacock DM, Zheng Q, Walker JA, Zhang Z, Zimprich CA, et al. KRAS is vulnerable to reversible switch-II pocket engagement in cells. Nat Chem Biol. 2022;18(6):596–604.CrossRefPubMedPubMedCentral
9.
Zurück zum Zitat Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I, et al. Sotorasib in KRAS p.G12C—Mutated Advanced Pancreatic Cancer. N Engl J Med. 2022;388(1):33–43.CrossRefPubMedPubMedCentral Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I, et al. Sotorasib in KRAS p.G12C—Mutated Advanced Pancreatic Cancer. N Engl J Med. 2022;388(1):33–43.CrossRefPubMedPubMedCentral
10.
Zurück zum Zitat Bekaii-Saab TS, Yaeger R, Spira AI, Pelster MS, Sabari JK, Hafez N, et al. Adagrasib in Advanced Solid Tumors Harboring a KRASG12C Mutation. J Clin Oncol. 2023;41(25):4097–106.CrossRefPubMedPubMedCentral Bekaii-Saab TS, Yaeger R, Spira AI, Pelster MS, Sabari JK, Hafez N, et al. Adagrasib in Advanced Solid Tumors Harboring a KRASG12C Mutation. J Clin Oncol. 2023;41(25):4097–106.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Sacher A, LoRusso P, Patel MR, Miller WH, Garralda E, Forster MD, et al. Single-Agent Divarasib (GDC-6036) in Solid Tumors with a KRAS G12C Mutation. N Engl J Med. 2023;389(8):710–21.CrossRefPubMed Sacher A, LoRusso P, Patel MR, Miller WH, Garralda E, Forster MD, et al. Single-Agent Divarasib (GDC-6036) in Solid Tumors with a KRAS G12C Mutation. N Engl J Med. 2023;389(8):710–21.CrossRefPubMed
12.
Zurück zum Zitat Li J, Shen L, Gu Y, Calles A, Wu L, Ba Y, et al. Preliminary activity and safety results of KRAS G12C inhibitor glecirasib (JAB-21822) in patients with pancreatic cancer and other solid tumors. J Clin Oncol. 2024;42(3):604–604.CrossRef Li J, Shen L, Gu Y, Calles A, Wu L, Ba Y, et al. Preliminary activity and safety results of KRAS G12C inhibitor glecirasib (JAB-21822) in patients with pancreatic cancer and other solid tumors. J Clin Oncol. 2024;42(3):604–604.CrossRef
13.
Zurück zum Zitat Hollebecque A, Kuboki Y, Murciano-Goroff YR, Yaeger R, Cassier PA, Heist RS, et al. Efficacy and safety of LY3537982, a potent and highly selective KRAS G12C inhibitor in KRAS G12C-mutant GI cancers: Results from a phase 1 study. J Clin Oncol. 2024;42(3):94–94.CrossRef Hollebecque A, Kuboki Y, Murciano-Goroff YR, Yaeger R, Cassier PA, Heist RS, et al. Efficacy and safety of LY3537982, a potent and highly selective KRAS G12C inhibitor in KRAS G12C-mutant GI cancers: Results from a phase 1 study. J Clin Oncol. 2024;42(3):94–94.CrossRef
14.
Zurück zum Zitat Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med. 2020;383(13):1207–17.CrossRefPubMedPubMedCentral Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRASG12C Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med. 2020;383(13):1207–17.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Dong X, Meng X, Zhang Y, Wang Y, Chen J, Han L, et al. Abstract CT119: Safety and efficacy of HS-10370 in KRAS G12C-mutated solid tumors including non-small cell lung cancer (NSCLC). Cancer Res. 2024;84(7):CT119–CT119. Dong X, Meng X, Zhang Y, Wang Y, Chen J, Han L, et al. Abstract CT119: Safety and efficacy of HS-10370 in KRAS G12C-mutated solid tumors including non-small cell lung cancer (NSCLC). Cancer Res. 2024;84(7):CT119–CT119.
16.
Zurück zum Zitat Rojas C, Lugowska I, Juergens R, Sacher A, Weindler S, Sendur MAN, et al. 663P Safety and preliminary efficacy of the KRAS G12C Inhibitor MK-1084 in solid tumors and in combination with pembrolizumab in NSCLC. Ann Oncol. 2023;34:S466–7.CrossRef Rojas C, Lugowska I, Juergens R, Sacher A, Weindler S, Sendur MAN, et al. 663P Safety and preliminary efficacy of the KRAS G12C Inhibitor MK-1084 in solid tumors and in combination with pembrolizumab in NSCLC. Ann Oncol. 2023;34:S466–7.CrossRef
17.
Zurück zum Zitat Jiang L, Menard M, Weller C, Wang Z, Burnett L, Aronchik I, et al. Abstract 526: RMC-9805, a first-in-class, mutant-selective, covalent and oral KRASG12D(ON) inhibitor that induces apoptosis and drives tumor regression in preclinical models of KRASG12D cancers. Cancer Res. 2023;83(7):526–526.CrossRef Jiang L, Menard M, Weller C, Wang Z, Burnett L, Aronchik I, et al. Abstract 526: RMC-9805, a first-in-class, mutant-selective, covalent and oral KRASG12D(ON) inhibitor that induces apoptosis and drives tumor regression in preclinical models of KRASG12D cancers. Cancer Res. 2023;83(7):526–526.CrossRef
18.
Zurück zum Zitat Zhou C, Li W, Song Z, Zhang Y, Zhang Y, Huang D, et al. LBA33 A first-in-human phase I study of a novel KRAS G12D inhibitor HRS-4642 in patients with advanced solid tumors harboring KRAS G12D mutation. Ann Oncol. 2023;34:S1273.CrossRef Zhou C, Li W, Song Z, Zhang Y, Zhang Y, Huang D, et al. LBA33 A first-in-human phase I study of a novel KRAS G12D inhibitor HRS-4642 in patients with advanced solid tumors harboring KRAS G12D mutation. Ann Oncol. 2023;34:S1273.CrossRef
19.
Zurück zum Zitat Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical Acquired Resistance to KRASG12C Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS-MAPK Reactivation. Cancer Discov. 2021;11(8):1913–22.CrossRefPubMedPubMedCentral Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical Acquired Resistance to KRASG12C Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS-MAPK Reactivation. Cancer Discov. 2021;11(8):1913–22.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired Resistance to KRASG12C Inhibition in Cancer. N Engl J Med. 2021;384(25):2382–93.CrossRefPubMedPubMedCentral Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired Resistance to KRASG12C Inhibition in Cancer. N Engl J Med. 2021;384(25):2382–93.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Corcoran RB. A single inhibitor for all KRAS mutations. Nat Cancer. 2023;4(8):1060–2.CrossRefPubMed Corcoran RB. A single inhibitor for all KRAS mutations. Nat Cancer. 2023;4(8):1060–2.CrossRefPubMed
22.
Zurück zum Zitat Zhao J, Fang J, Yu Y, Chu Q, Li X, Chen J, et al. Updated safety and efficacy data of combined KRAS G12C inhibitor (glecirasib, JAB-21822) and SHP2 inhibitor (JAB-3312) in patients with KRAS p.G12C mutated solid tumors. J Clin Oncol. 2024;42(16):3008–3008.CrossRef Zhao J, Fang J, Yu Y, Chu Q, Li X, Chen J, et al. Updated safety and efficacy data of combined KRAS G12C inhibitor (glecirasib, JAB-21822) and SHP2 inhibitor (JAB-3312) in patients with KRAS p.G12C mutated solid tumors. J Clin Oncol. 2024;42(16):3008–3008.CrossRef
23.
Zurück zum Zitat Kim D, Herdeis L, Rudolph D, Zhao Y, Böttcher J, Vides A, et al. Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature. 2023;619(7968):160–6.CrossRefPubMedPubMedCentral Kim D, Herdeis L, Rudolph D, Zhao Y, Böttcher J, Vides A, et al. Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature. 2023;619(7968):160–6.CrossRefPubMedPubMedCentral
24.
Zurück zum Zitat Arbour KC, Punekar S, Garrido-Laguna I, Hong DS, Wolpin B, Pelster MS, et al. 652O Preliminary clinical activity of RMC-6236, a first-in-class, RAS-selective, tri-complex RAS-MULTI(ON) inhibitor in patients with KRAS mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). Ann Oncol. 2023;34:S458.CrossRef Arbour KC, Punekar S, Garrido-Laguna I, Hong DS, Wolpin B, Pelster MS, et al. 652O Preliminary clinical activity of RMC-6236, a first-in-class, RAS-selective, tri-complex RAS-MULTI(ON) inhibitor in patients with KRAS mutant pancreatic ductal adenocarcinoma (PDAC) and non-small cell lung cancer (NSCLC). Ann Oncol. 2023;34:S458.CrossRef
25.
Zurück zum Zitat Holderfield M, Lee BJ, Jiang J, Tomlinson A, Seamon KJ, Mira A, et al. Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature. 2024;629(8013):919–26.CrossRefPubMedPubMedCentral Holderfield M, Lee BJ, Jiang J, Tomlinson A, Seamon KJ, Mira A, et al. Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature. 2024;629(8013):919–26.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Leidner R, Silva NS, Huang H, Sprott D, Zheng C, Shih YP, et al. Neoantigen T‑Cell Receptor Gene Therapy in Pancreatic Cancer. N Engl J Med. 2022;386(22):2112–9.CrossRefPubMedPubMedCentral Leidner R, Silva NS, Huang H, Sprott D, Zheng C, Shih YP, et al. Neoantigen T‑Cell Receptor Gene Therapy in Pancreatic Cancer. N Engl J Med. 2022;386(22):2112–9.CrossRefPubMedPubMedCentral
27.
Zurück zum Zitat Pant S, Wainberg ZA, Weekes CD, Furqan M, Kasi PM, Devoe CE, et al. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: the phase 1 AMPLIFY-201 trial. Nat Med. 2024;30(2):531–42.CrossRefPubMedPubMedCentral Pant S, Wainberg ZA, Weekes CD, Furqan M, Kasi PM, Devoe CE, et al. Lymph-node-targeted, mKRAS-specific amphiphile vaccine in pancreatic and colorectal cancer: the phase 1 AMPLIFY-201 trial. Nat Med. 2024;30(2):531–42.CrossRefPubMedPubMedCentral
Metadaten
Titel
Unraveling (K)RAS in pancreatic ductal adenocarcinoma
verfasst von
OA Dr. Bernhard Doleschal, PhD
Publikationsdatum
21.11.2024
Verlag
Springer Vienna
Erschienen in
memo - Magazine of European Medical Oncology / Ausgabe 4/2024
Print ISSN: 1865-5041
Elektronische ISSN: 1865-5076
DOI
https://doi.org/10.1007/s12254-024-01003-6