Open Access

Targeting KRAS for the potential treatment of pancreatic ductal adenocarcinoma: Recent advancements provide hope (Review)

  • Authors:
    • Joshua Zhang
    • Lily Darman
    • Md Sazzad Hassan
    • Urs Von Holzen
    • Niranjan Awasthi
  • View Affiliations

  • Published online on: October 4, 2023     https://doi.org/10.3892/or.2023.8643
  • Article Number: 206
  • Copyright: © Zhang et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Kirsten rat sarcoma viral oncogene homolog (KRAS) is one of the most frequently mutated oncogenes in solid tumors. More than 90% of pancreatic ductal adenocarcinoma (PDAC) are driven by mutations in the KRAS gene, suggesting the importance of targeting this oncogene in PDAC. Initial efforts to target KRAS have been unsuccessful due to its small size, high affinity for guanosine triphosphate/guanosine diphosphate, and lack of distinct drug‑binding pockets. Therefore, much of the focus has been directed at inhibiting the activation of major signaling pathways downstream of KRAS, most notably the PI3K/AKT and RAF/MAPK pathways, using tyrosine kinase inhibitors and monoclonal antibodies. While preclinical studies showed promising results, clinical data using the inhibitors alone and in combination with other standard therapies have shown limited practicality, largely due to the lack of efficacy and dose‑limiting toxicities. Recent therapeutic approaches for KRAS‑driven tumors focus on mutation‑specific drugs such as selective KRASG12C inhibitors and son of sevenless 1 pan‑KRAS inhibitors. While KRASG12C inhibitors showed great promise against patients with non‑small cell lung cancer (NSCLC) harboring KRASG12C mutations, they were not efficacious in PDAC largely because the major KRAS mutant isoforms in PDAC are G12D, G12V, and G12R. As a result, KRASG12D and pan‑KRAS inhibitors are currently under investigation as potential therapeutic options for PDAC. The present review summarized the importance of KRAS oncogenic signaling, challenges in its targeting, and preclinical and clinical targeted agents including recent direct KRAS inhibitors for blocking KRAS signaling in PDAC.

Introduction

RAS oncogenes [HRAS, NRAS, Kirsten rat sarcoma viral oncogene homolog (KRAS)] comprise the most frequently mutated gene family in cancer, identified in ~30% of cancers. The RAS family plays a critical role in major cellular processes such as growth and proliferation; thus, an activating mutation in these genes can lead to tumor formation. Within this group, KRAS is the most frequently mutated isoform in cancer, identified in ~84% of all RAS-mutant cancers. NRAS mutation (11–17%) is also relatively prevalent, while HRAS is quite rare among RAS-mutated cancers (1).

KRAS is a small GTPase signal transduction protein that cycles between active guanosine triphosphate (GTP)-bound and inactive guanosine diphosphate (GDP)-bound states. In normal quiescent cells, RAS is largely GDP-bound and inactive, but the GTP-bound state is formed through extracellular stimuli activation of receptor tyrosine kinases (RTKs) as well as other cell-surface receptors. Activated KRAS maintains the engagement of effector proteins that then regulate several intracellular signaling networks that control mitogenic processes. KRAS has been shown to play a central role in controlling tumor metabolism (2). Oncogenic KRAS genes are characterized by missense mutations that encode single amino acid substitutions at three primary locations: Glycine-12 (G12), glycine-13 (G13), or glutamine-61 (Q61). Among these, G12 mutations comprise 83% of all KRAS mutations, followed by G13 (14%) and Q61 (2%) mutations (3). The mutation subtypes at KRAS are mainly classified as KRAS G12D, G12V, G12C, G13D, G12R and G12A. KRAS mutations are most common in pancreatic cancer, NSCLC and colorectal cancer, and the profiles of KRAS mutation subtypes differ in different types of cancer. For example, G12C mutation is the most common subtype in NSCLC (41%), whereas G12D and G12V are the major subtypes in pancreatic cancer and colorectal cancer (4). All KRAS mutations render KRAS constitutively bound to GTP and active, overstimulating effector signaling pathways to drive uncontrolled growth of cells leading to cancer formation. This suggests that blocking KRAS has high therapeutic potential for several cancers. In the present review, the critical role of oncogenic KRAS signaling, challenges in its targeting, and preclinical and clinical studies targeting KRAS signaling, including its downstream signaling effectors or direct inhibitors, in pancreatic ductal adenocarcinoma (PDAC) were investigated. In the present review, a comprehensive literature search was performed using three databases, PubMed, Medline and Web of Science.

KRAS in pancreatic cancer

Comprising >90% of cases, the most common form of pancreatic cancer is PDAC, which has a 5-year overall survival (OS) rate of 11% (5). PDAC is the 3rd leading cause of cancer-related deaths in the US and has the highest frequency of KRAS mutations (>90%). PDAC displays two major KRAS mutations at G12D (41%) and G12V (34%), while other less frequent mutations are G12R (16%), Q61H (4%) and G12C (~1%) (6).

PDAC has four types of pre-neoplastic precursors: Intraductal papillary mucinous neoplasm, pancreatic mucinous cystic neoplasm, intraductal tubular papillary neoplasm, and pancreatic intraepithelial neoplasia (PanIN). PanIN is the most common precursor lesion and is classified as low-grade (PanIN-1A and PanIN-1B), intermediate-grade (PanIN-2), or high-grade (PanIN-3) before progressing to PDAC. KRAS mutation is an early event in the genetic onset of PDAC. The progression from normal pancreatic tissue to PDAC is typically initiated by advancing stages on noninvasive microscopic ductal lesions, PanINs. KRAS mutations are considered to be the driving force in the development of human PanINs and >90% frequency of KRAS mutations are identified in PanIN-1 lesions. Furthermore, Aguirre et al reported that the KRASG12D mutation alone formed PanIN and the protracted onset of PDAC (7).

PanIN-to-PDAC progression is initiated by subsequent inactivation of tumor suppressor gene cyclin-dependent kinase inhibitor 2A, identified in PanIN-2, and then followed by inactivation of two other tumor suppressor genes, TP53 and SMAD4, proteins unique to PanIN-3. KRAS-mediated signaling then further leads to the development of PDAC through several metabolic processes, providing the energy and biosynthetic building blocks necessary to drive uncontrolled tumor growth. Furthermore, previous studies revealed that KRAS-mutant PDAC tumor cells are capable of regulating autophagy to meet metabolic demand (8,9). Although KRAS mutations are the initiating genetic step, mutant KRAS is still involved in every step to maintain the growth of metastatic PDAC (2). The gradual progression of KRAS-mediated PDAC progression ultimately results in rapid growth and metastasis. Thus, there is high therapeutic potential in targeting the KRAS pathway for improving clinical PDAC therapy.

KRAS and metabolic reprogramming in pancreatic cancer

The presence of KRAS mutations creates an ideal environment for the thriving of cancer cells, as it elevates the levels of glucose and glycolytic intermediates, cellular redox potential, and uptake of fatty acids and glutamate. Furthermore, mutant KRAS enhances micropinocytosis and promotes amino acid turnover (2,1012). Autophagy becomes a vital mechanism for KRAS mutant PDAC cells, supporting tumor growth and survival during starvation (2,1011). Additionally, mutant KRAS plays a role in reprogramming the tumor microenvironment (TME) by influencing the expression of enzymes that regulate glucose metabolism including HK1/2, phosphofructokinase 1, and LDHA (2,11,13,14). The KRAS/MAPK signaling pathway further amplifies the expression of MYC, leading to increased nonoxidative pentose phosphate pathway gene RPIA expression and sustained nucleotide biosynthesis (15). Furthermore, mutant KRAS orchestrates the reprogramming of noncanonical glutamine metabolism, resulting in a cellular redox state that strongly favors tumor growth (16,17). Collectively, these studies establish the crucial role of mutant KRAS in maintaining PDAC growth through the selective activation and adaptation of biosynthetic metabolic pathways. Consequently, exploring the metabolic reprogramming occurring in KRAS mutant PDAC presents an opportunity to gain insights into the underlying mechanisms of tumor progression and therapeutic resistance. Such knowledge can be harnessed to develop innovative therapeutic strategies.

Challenges in KRAS direct targeting

In theory, targeting KRAS signaling could potentially provide an effective approach to block the growth of KRAS-dependent tumors, but this was deemed unrealistic due to several barriers. First, KRAS is a member of a large family of related proteins sharing similar GTP/GDP binding domains, causing the development of specific targeted drugs very difficult. Second, KRAS has a very high picomolar affinity for GTP and GDP, both of which have very high intracellular concentrations. Third, the KRAS protein lacks accessible binding sites for the high-affinity binding of small-molecule inhibitors (18). Thus, not only do KRAS protein inhibitors have limited effectiveness due to chemical affinity, but they are also hindered by the structure of the target proteins. Due to these challenges, KRAS is widely considered undruggable, shifting focus to blocking downstream effector signaling.

Targeting downstream effector signaling has emerged as a promising alternative approach to block oncogenic KRAS signaling pathways. Oncogenic-activating KRAS mutations regulate an intricate complex of cytoplasmic signaling networks. There are at least 11 different effector families, and tumorigenesis is likely the result of multiple integrated effector-signaling pathways (19). Thus, while it shows great promise, targeting downstream KRAS effector signaling is more complex than it seemed at first glance. Therapeutic approaches must decide which effector pathways are the best to target and if co-inhibition of multiple effectors is required. Based on the prominent role of KRAS in PDAC progression and difficulties in the direct targeting of KRAS, much of the efforts have centered on indirect strategies in hopes of finding a more effective treatment for PDAC.

Preclinical studies targeting KRAS signaling in pancreatic cancer

KRAS-GTP is known to activate several downstream oncogenic signaling pathways including PI3K/AKT/mTOR, MAPK, RAL-PLD1 and T1AM1-Rac, and crosstalk exists among these pathways (Fig. 1). Thus, the concept of targeting KRAS effector signaling is not without complications.

PI3K/AKT/mTOR pathway inhibition

Previous studies have shown that PI3K subunits play a critical role as effectors of mutant KRAS-driven oncogenesis (20,21). PI3K phosphorylates PIP2 and stimulates the formation of PIP3, which then phosphorylates and activates a multitude of proteins, including AKT 1/2/3, localizing it in the plasma membrane. AKT phosphorylates numerous other proteins that promote cell growth, in particular the mammalian target of rapamycin (mTOR) (22,23). The PI3K/AKT/mTOR pathway plays a pivotal role in a number of cellular processes such as proliferation, survival, and growth. Abnormalities in this pathway such as PI3K mutation/amplification, loss of PTEN, AKT mutation or RTK activation, have been implicated in cancer cell proliferation, invasion, survival, metastasis, epithelial-mesenchymal transition (EMT) and drug resistance (24,25). There have been conflicting studies regarding PI3K activation in driving PDAC development. A previous study revealed that 93% of rare PIK3CA mutations co-occur with a KRAS mutation, suggesting that activated KRAS is not sufficient to effectively activate PI3K (26). However, another study revealed that KRAS suppression did not alter AKT activation levels in a majority of KRAS-mutated PDAC cell lines (27).

Several drugs have been developed to target this pathway, including PI3K, PI3K/mTOR, mTOR, and AKT inhibitors (Table I). Wortmannin was the first PI3K inhibitor developed which was identified to be extremely effective in inhibiting PI3K, but it was revealed to be nonspecific, unstable and toxic in animals (28). In orthotopic PDAC xenografts, wortmannin promoted the antitumor activity of gemcitabine (tumor weight reduction relative to control was 1.4-fold by gemcitabine and 5-fold by gemcitabine plus wortmannin; P<0.001) supporting its potential as an adjuvant to conventional chemotherapy treatments of PDAC (29). LY294002, however, is a very specific synthetic inhibitor of PI3K and is chemically more stable but has been identified to be less potent than wortmannin in preclinical studies. In PDAC models, LY294002 inhibited in vitro cell proliferation, induced apoptosis and reduced in vivo tumor growth. Furthermore, LY294002 enhanced the effects of cisplatin both in vitro and in vivo (tumor volume decreased to 77, 70 or 44% of the volume in the controls by cisplatin, LY294002 or combination; P<0.05) (30). Subsequently, Wang et al reported the opposite effect of LY294002 by demonstrating that LY294002 (but not wortmannin) enhanced AKT phosphorylation in the gemcitabine-resistant PDAC cell lines (31). This study suggested that the PI3K inhibitors can be counterproductive with gemcitabine-resistant PDAC cells.

Table I.

Preclinical results with KRAS downstream effector inhibitors in PDAC.

Table I.

Preclinical results with KRAS downstream effector inhibitors in PDAC.

DrugTargetEffect as a single agentEffect in combination therapy(Refs.)
WortmanninPI3KEffective in inhibiting PI3K, but relatively non-specific, unstable, and toxicPromoted antitumor activity of Gem in orthotopic xenografts(28,29)
LY294002PI3KSpecific, stable but less potent, antiproliferative, opposite effect in Gem-resistant PDAC cell linesSynergistic antitumor effects with cisplatin (CDX)(30,31)
MK-2206AKTAntiproliferative and pro-apoptotic effects in PDAC cell linesSynergistic antitumor response with CDK inhibitor dinaciclib (PDX); MEK inhibitor trametinib and chemotherapy (in vitro, CDX)(32,33,73)
Perifosine (KRX-0401)AKTInhibited growth of PDAC cells including primary culturesEnhanced activity in combination with Gem (in vitro)(34)
API-2AKTRadiosensitization of PDAC cellsSynergistic antitumor effects with MEK inhibitor mirdametinib (CDX)(70)
Sirolimus (Rapamycin)mTORAntiproliferative and radiosensitization effects (in vitro); efficacy in KC PTEN mice but not in KPC mice (GEMM)Synergistic tumor growth inhibition with metformin (CDX); and EGFR inhibitor gefitinib (in vitro)(3540)
EverolimusmTORInhibited growth of Gem-resistant PDAC cell linesHigher efficacy in combination with Gem (in vitro); trametinib (PDX); sorafenib (PDX)(4146)
Temsirolimus (CCI-779)mTORAntiproliferative effects on PDAC cell linesSynergistic antitumor efficacy in combination with Gem (CDX)(47,48)
NVP-BEZ235PI3K-mTORAntiproliferative activity (in vitro); tumor growth inhibition (orthotopic PDX)Synergistic antitumor activity with Gem (in vitro, CDX); panobinostat (CDX); inRas37 antibody (CDX); PD0325901 (in vitro)(4953)
Tipifarnib (R115777)RasAntiproliferative and proapoptotic activity (in vitro)N/A(54)
Lonafarnib (SCH66336)RasAntiproliferative activity (in vitro)Synergistic antitumor activity in combination with taxanes (in vitro)(55)
Sorafenib (BAY43-9006)RafAntiproliferative and proapoptotic activity (in vitro)Synergistic antitumor activity with vitamin K (in vitro); melatonin (in vitro, CDX); Gem (in vitro, CDX)(5659)
GW5074c-RafAntiproliferative activity (in vitro); tumor growth inhibition (in vivo)Synergistic antitumor activity with DFMO (in vitro, CDX)(60)
TrametinibMEKAntiproliferative and pro-apoptotic effects (in vitro)Increased the antitumor effects of lapatinib (orthotopic PDX); NPT plus Gem with or without MK-2206 (CDX); HDAC inhibitors MPT0E028 or SAHA (in vitro, CDX)(6163,73)
Mirdametinib (PD-0325901)MEKTumor growth inhibition (CDX); radiosensitization of PDAC cellsSynergistic antitumor effects with clusterin inhibition (in vitro); AKT inhibitor API-2 (CDX)(64)
SCH772984ERKAntiproliferative effect on PDAC cellsSynergistic antitumor effects with cucurbitacin (in vitro, CDX); autophagy-inhibiting agents (in vitro, PDX, organoid); CDK4/6 inhibitor palbociclib (in vitro, organoid)(27,6568)
Ulixertinib (BVD-523)ERKInhibition of PDAC cell viability (in vitro)Synergistic effect with Gem, pan-HER1/2/3 inhibitor afatinib or pan-PI3K inhibitor GDC-0941 (CDX)(69)
TIC10 (ONC201)AKT/ERKAntiproliferative, proapoptotic activity (in vitro); tumor growth inhibition (in vivo)Synergistic antitumor effects with Gem (CDX)(72)

[i] KRAS, Kirsten rat sarcoma viral oncogene homolog; PDAC, pancreatic ductal adenocarcinoma; Gem, gemcitabine; CDX, cell-derived xenografts; PDX, patient-derived xenografts; GEMM, genetically engineered mouse model; DFMO, difluoromethylornithine; NPT, nab-paclitaxel.

MK-2206, a novel AKT inhibitor, inhibited in vitro proliferation and induced apoptosis of PDAC cells. When combined with gemcitabine, it both enhanced the cytotoxic efficacy of gemcitabine and inhibited AKT phosphorylation (32). In KRAS-mutant patient-derived xenograft (PDX) models, a combination of MK-2206 with dinaciclib, an inhibitor of cyclin-dependent kinases, demonstrated a reduction in PDAC growth (by >90%; P<0.001) and the number of metastatic lesions (by >88%; P<0.001) (33). Another AKT inhibitor perifosine (KRX-0401) was revealed to inhibit in vitro cell growth and interacted synergistically with gemcitabine in PDAC cells including primary cultures (34).

There are three major mTOR inhibitors currently approved by the FDA: Sirolimus, everolimus and temsirolimus. Sirolimus (rapamycin) induced autophagy and apoptosis in PDAC cell lines (35) but it also led to resistance mediated by AKT phosphorylation (36). The second-generation mTOR inhibitors, such as KU63794 and PP242, also led to treatment resistance due to an increase in ERK activation (36). Rapamycin exhibited a dose-dependent radiosensitization effect (37) and synergistic antiproliferative and antiangiogenic effects in combination with EGFR inhibitor gefitinib on PDAC cells (38). In vivo studies in mice demonstrated that rapamycin was specifically effective on KRAS-mutant PDAC tumors that have loss of PTEN (median survival in controls, gemcitabine, rapamycin and gemcitabine plus rapamycin was 10, 14, 56 and 32 days, respectively), while KRAS-mutant tumors with mutant p53 (KPC) did not respond (39). Metformin causes diverse effects on PDAC tumorigenesis in both mTOR-dependent and -independent manner. In the syngeneic mouse model using C57BL/6 mice and Pan02 cells, metformin and rapamycin both exhibited significant tumor growth reduction (tumor burden compared with control 0.90 g was 0.62 g with metformin and 0.25 g with rapamycin) (40). Everolimus, an analog of rapamycin, sensitized PDAC cells to the effects of gemcitabine in an in vitro study (41). In vitro studies demonstrated that gemcitabine-resistant PDAC cells were more sensitive to everolimus, in contrast to various EGFR, AKT and PI3K inhibitors (42). In another in vitro study in PDAC cell lines, everolimus had an antiproliferative effect, while its combination with sorafenib exhibited an antagonistic effect (43). Wei et al demonstrated that neither everolimus nor AZD8055, a 2nd-generation mTOR inhibitor, exerted any cell viability inhibitory effect on PDAC cell lines (44). In PDAC PDX models, everolimus displayed higher antitumor efficacy when combined with either sorafenib (the tumor/control ratio was 0.6, 0.5 and 0.2 for treatment with everolimus, sorafenib and the combination) (45) or trametinib (46). Temsirolimus (CCI-779), a water-soluble, more stable and specific mTOR inhibitor, showed a significant antiproliferative effect on PDAC cells (47) and exhibited synergistic antitumor response in combination with gemcitabine in PDAC xenograft models (compared with control, decrease in tumor volume was 68%, P=0.0009 with a high dose of CCI-779; and 41%, P=0.0002 with CCI-779 plus gemcitabine (48).

Dual inhibition of PI3K-mTOR holds arguably the most potential. NVP-BEZ235, the primary proponent of this class of drugs, demonstrated significant delay in tumor growth (56, 36 and 46%) in three different orthotopic PDX models (P<0.05) (49). In the peritoneal dissemination animal survival model, NVP-BEZ235 enhanced gemcitabine response (median survival in days was 16, 21, 28 and 30 in control, NVP-BEZ235, gemcitabine, and combination; P<0.05) (50). In PDAC subcutaneous xenografts, NVP-BEZ235 exhibited synergistic tumor growth inhibition in combination with pan-histone deacetylase inhibitor panobinostat (51) or inRas37 antibody (52). In vitro study using PANC-1 and MIA PaCa-2 PDAC cells revealed that NVP-BEZ235 markedly induced the ERK/MEK pathway. The MEK inhibitors U126 and PD0325901 prevented ERK overactivation induced by NVP-BEZ235 and the combination of MEK inhibitors with NVP-BEZ235 produced a further inhibition of PDAC cell proliferation (53).

RAS/RAF/MEK/ERK pathway inhibition

Another hallmark downstream pathway through which KRAS signaling occurs is the Ras-Raf-MEK-ERK (MAPK) pathway which is the key effector in the initiation, progression, and maintenance of KRAS-dependent tumors. Thus, several drugs targeting different components of this pathway have been explored extensively for therapeutic intervention in KRAS-mutant PDAC (Table I).

Ras proteins undergo farnesylation by the enzyme farnesyl-protein transferase for their biological or transforming functions. Therefore, farnesyltransferase inhibitors (FTIs), tipifarnib (R115777) and lonafarnib (SCH66336) have been assessed in PDAC preclinical models. Tipifarnib suppressed the growth of human PDAC cell lines through modulation of the STAT3 and ERK pathways (54). The FTI, lonafarnib, synergized with taxanes to inhibit cell proliferation in KRAS-mutant and KRAS-wild-type PDAC cells (55).

Raf kinases (ARaf, BRaf, and CRaf/Raf1) comprise the most significant effectors of KRAS-driven PDAC. In PDAC cell lines, Raf inhibitor sorafenib (BAY 43-9006) that also targets VEGFR2 and PDGFR-b, demonstrated strong antiproliferative and proapoptotic effects, either alone (56) or in combination with vitamin K (57). Sorafenib also synergized with melatonin to suppress the growth of PDAC both in vitro and in vivo (58). Sorafenib alone or in combination with gemcitabine and EMAP inhibited PDAC cell proliferation. This study also showed enhancement in animal survival by combination treatment of sorafenib, gemcitabine and EMAP (median survival in controls, gemcitabine, sorafenib, EMAP and gemcitabine + sorafenib + EMAP was 22, 29, 23, 25 and 36 days; P=0.004) (59). In PDAC orthotopic xenografts, a selective c-RAF inhibitor, GW5074, exhibited a significant decrease in tumor weight, either alone or in combination with polyamine biosynthesis inhibitor difluoromethylornithine, but it had no effect on improving animal survival (60).

Downstream to RAF, inhibitors for MEK and ERK have been assessed extensively. In KRAS-mutant and wild-type orthotopically implanted patient-derived tumors, the MEK-inhibitor, trametinib, showed a significant reduction in tumor growth and liver metastasis that was increased by the EGFR/HER-2 inhibitor lapatinib (61). In KRAS-mutant PDAC cell-derived xenograft (CDX) models, trametinib increased the effects of nab-paclitaxel-based chemotherapy by inhibiting tumor growth and enhancing animal survival (compared with controls, increase in animal survival was 95 and 145% by nab-paclitaxel + gemcitabine and nab-paclitaxel + gemcitabine + trametinib) (62). Chao et al observed that MEK inhibitors (PD98059 or trametinib) increased the antiproliferative and proapoptotic effects of HDAC inhibitors (MPT0E028 or SAHA) in KRAS-mutant and wild-type PDAC cell in vitro studies. Furthermore, in AsPC-1 subcutaneous xenografts, MPT0E028 and PD98059 combination revealed enhanced antitumor activity (63). Recently, MEK inhibitor mirdametinib (PD0325901) exerted antitumor efficacy in MIA PaCa-2 PDAC xenografts at day 4 and became refractory within a week after treatment due to the involvement of clusterin expression (64). SCH772984 was the first ERK inhibitor studied in PDAC, and it displayed the ability to suppress PDAC xenograft growth (27). SCH772984 in combination with cucurbitacin, a natural tetracyclic triterpene, synergized to induce growth inhibition and apoptosis of PDAC cells in vitro and reducing tumor growth in PDAC subcutaneous xenografts (percent inhibition in tumor volume by cucurbitacin, SCH772984 and combination treatment was 63.8, 54.7 and 85, respectively) (65). Furthermore, SCH772984, in combination with autophagy inhibition, synergistically reduced PDAC growth in vitro and in vivo models (66,67). Recently, SCH772984 has been demonstrated to combine synergistically with CDK4/6 inhibitor palbociclib in KRAS-mutant PDAC cell lines and organoid models (68). Another ERK inhibitor, ulixertinib, has been shown to effectively inhibit the growth of multiple PDAC cell lines and potentiate the cytotoxic effect of gemcitabine in vitro and in vivo. This study also showed that concurrent inhibition of HER (with afatinib) or PI3K proteins (with GDC-0941) synergizes with ulixertinib in suppressing PDAC cell growth in subcutaneous xenograft models (69). Thus, MEK and ERK inhibitors showed promise in preclinical studies in mitigating the progression of PDAC.

Due to the extensive crosstalk and compensatory mechanism between PI3K/AKT and MAPK pathways, simultaneous targeting of these two pathways has been explored in PDAC. In PDAC subcutaneous xenografts, a combination treatment with MEK inhibitor mirdametinib and AKT inhibitor (API-2) induced activation of apoptotic pathways, radiosensitized pancreatic cancer cells and maximized tumor growth inhibition (70). Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has been shown to induce apoptosis of cancer cells by engaging its cell surface death receptors (71). A TRAIL-inducing compound TIC10 (ONC201) that causes dual inhibition of AKT and ERK-inducing TRAIL pathway-mediated cell death, inhibited PDAC cell survival and proliferation in vitro, and demonstrated potent antitumor activity in PDAC xenografts as well as synergized gemcitabine activity (72). Subsequently, Awasthi et al demonstrated that the combined inhibition of PI3K and MAPK signaling with MK-2206 and trametinib, respectively, displayed an enhanced nab-paclitaxel plus gemcitabine (NPT+Gem) chemotherapy antitumor response in PDAC in vitro and in vivo models. In this study, compared with controls, enhancement in animal survival by NPT + Gem and NPT + Gem + MK-2206 + trametinib treatment was 67 and 129%, respectively (73). These findings indicate the potential of dual inhibition of downstream targets of KRAS-mutation-driven signaling in combination with current treatments for clinical PDAC therapy.

Clinical studies targeting KRAS signaling in pancreatic cancer

Dense desmoplastic stroma which comprises >90% of the tumor mass is one of the hallmarks of PDAC. This stroma plays a major role in PDAC progression, metastasis, and therapy resistance (74). The TME in PDAC preclinical models and patients with PDAC have several differences including composition, oncogenic mutations and expression of oncogenic growth factors and cytokines (75,76). Therefore, inhibiting the major signaling pathways downstream of KRAS sometimes has different effects on PDAC tumors in preclinical settings compared with their effect on the progression and survival of patients with PDAC (76).

PI3K/AKT/mTOR pathway inhibition

Considering the activation of the PI3K/AKT/mTOR signaling pathway in KRAS-mutant PDAC progression and survival, several clinical trials evaluated the antitumor efficacy of inhibitors of this pathway in clinical studies (Table II).

Table II.

Clinical results with KRAS downstream effector inhibitors.

Table II.

Clinical results with KRAS downstream effector inhibitors.

DrugTargetEffect as single agentEffect in combination therapy(Refs.)
Copanlisib (BAY 80-6946)PI3KN/APhase I, with Gem or Gem plus cisplatin: manageable safety profile, favorable PK and clinical efficacy(77)
Alpelisib (BYL719)PI3KaN/APhase I, with NPT plus Gem: manageable safety profile; PFS, 5.36 mo; OS, 8.74 mo(78)
Buparlisib (BKM120)PI3KN/APhase I, minimum activity with trametinib or binimetinib, significant toxicity with mFOLFOX6(79,80,117)
IdelalisibPI3KPhase I, no conclusion: study terminated earlyPhase I, with NPT or mFOLFOX6: early termination of the study due to severe toxicity concerns(81)
Perifosine (KRX-0401)AKTPhase II, halted due to high toxicity and low efficacyN/A(82,83)
MK-2206AKTN/APhase II, with selumetinib vs. mFOLFOX, OS 3.9 vs. 6.7 mo, PFS 1.9 vs. 2.0 mo; phase I, with dinaciclib, OS 2.2 mo(84,85)
SirolimusmTORN/AWith vismodegib: minimum activity, only SD in a subgroup of patients(92)
EverolimusmTORPhase II, Gem refractory patients, OS 4.5 mo, PFS 1.8 moPhase II, with erlotinib (OS 87 days, PFS 49 days); phase II, with capecitabine (OS 8.9 mo, PFS 3.6 mo); phase I, with ribociclib (OS 3.7 mo, PFS 1.8 mo); cetuximab plus capecitabine (phase I/II, OS 5 mo)(8688,91,95)
Temsirolimus (CCI-779)mTORPhase II, Gem refractory patients, OS 44 days, PFS 19 daysIneffective with Gem (phase II, OS 4.95 mo, PFS 2.69 mo); docetaxel (phase I); erlotinib (phase I)(87,89,90,94)
Tipifarnib (R115777)RasPhase II, untreated patients: OS 19.7 weeks, PFS 4.9 weeks; Phase II, OS 2.6 mo, PFS 1.4 moNo benefit: Monotherapy vs. with Gem (phase III, OS 193 vs. 182 days, PFS 112 vs. 109 days); CXRT vs. CXRT + tipifarnib (phase II, OS 11.5 vs. 8.9 mo)(9699)
Sorafenib (BAY 43-9006)c/b-RafPhase II, well tolerated but no clinical activity (OS 4.3 mo, PFS 2.3 mο)Phase III, with or without Gem, OS 9.2 vs. 8 mo, PFS 5.7 vs. 3.8 mo; phase II, cisplatin plus Gem with and without sorafenib, OS 7.5 vs. 8.3 mo, PFS 4.3 vs. 4.5 mo; phase I, with concurrent RT and Gem, OS 12.6 mo, PFS 10.6 mo(100104)
TrametinibMEKN/APhase II, with Gem vs. Gem alone, OS 8.4 vs. 6.7 mo (p=0.453), PFS 16 vs. 15 weeks (p=0.349); phase I, with lapatinib, no activity; phase II, with erlotinib, OS 7.3 mo, PFS 1.9 mo; phase II, with RT plus pembrolizumab vs. RT plus Gem, OS 14.9 vs. 12.8 mo, PFS 8.2 vs. 5.4 mo; phase II, with GSK2256098, OS 3.6 mo, PFS 1.6 mo(105109,116)
SelumetinibMEKPhase II, Gem refractory patients, compared with capecitabine (OS 5.4 vs. 5.0 mo); phase II, KRASG12R-mutant patients with ≥2 chemotherapy, OS 9 mo, PFS 3 moPhase II, with MK-2206 vs. mFOLFOX, OS 3.9 vs. 6.7 mo, PFS 1.9 vs. 2.0 m(84,110,111)
BinimetinibMEKN/APhase I, terminated due to toxicity issues with avelumab; study ongoing with hydroxychloroquine(112,113)
Ulixertinib (BVD-523)ERKN/APhase I, with NPT plus Gem, OS 12.23 mo, PFS 5.46 mo, study terminated due to toxicity; study ongoing with palbociclib(114,115)

[i] KRAS, Kirsten rat sarcoma viral oncogene homolog; N/A, not applicable; Gem, gemcitabine; NPT, nab-paclitaxel; RT, radiotherapy; PK, pharmacokinetics; PFS, progression-free survival; OS, overall survival; mo, months; CXRT, concurrent radiation with gemcitabine and paclitaxel; PR, partial response; SD, stable disease.

Copanlisib (BAY 80-6946), a pan-class I PI3K inhibitor, in combination with gemcitabine or cisplatin plus gemcitabine in solid tumors including PDAC demonstrated favorable clinical efficacy (out of 4 evaluated patients with PDAC, 1 partial response, 2 stable disease) with an acceptable toxicity profile in a phase I study (77). In a phase I trial of advanced patients with PDAC who have not received any cytotoxic chemotherapy except as adjuvant therapy, alpelisib (BYL719), an α-specific PI3K inhibitor, in combination with nab-paclitaxel plus gemcitabine, was safely administered and the median progression-free survival (PFS) and OS were 5.36 and 8.74 months (78). Another pan-class I PI3K inhibitor buparlisib (BKM120), in a phase I study which included patients with metastatic PDAC with RAS- or BRAF-mutation, in combination with trametinib showed minimum clinical activity (best overall response was stable disease) (79). In a phase I study of patients with refractory solid tumors including PDAC, buparlisib combination with mFOLFOX6 caused an increase in toxicity and one patient with stage IV PDAC exhibited a 47% decrease in measurable disease from baseline (80). A phase Ib study of PI3K inhibitor idelalisib alone or in combination with nab-paclitaxel or mFOLFOX6 in patients with PDAC was terminated prematurely due to severe toxicity issues observed in a phase III clinical study of idelalisib for hematological malignancies (81).

Perifosine (KRX-0401) and MK-2206 are the most studied AKT inhibitors for PDAC clinical therapy. Although perifosine exhibited significant activity in PDAC preclinical studies, two phase II clinical trials using this drug in patients with advanced PDAC who were previously untreated (82) or had one prior systemic therapy (83) were halted prematurely due to a lack of efficacy and high toxicity. A phase II study compared MK-2206 plus MEK inhibitor selumetinib with mFOLFOX in gemcitabine-refractory patients with metastatic PDAC (84). The median OS was shorter in the MK-2206 plus selumetinib versus mFOLFOX (3.9 vs. 6.7 months; P=0.15) and the median PFS was also inferior (1.9 vs. 2.0 months; P=0.02) (84). Subsequently, a phase I study of MK-2206 plus CDK inhibitor dinaciclib in patients with previously treated metastatic PDAC demonstrated disappointing results (median survival 2.2 months; survival rates at 6 and 12 months 11 and 5%, respectively) (85).

The mTOR inhibitor everolimus in a phase II study in gemcitabine refractory metastatic PDAC patients demonstrated minimal clinical activity and median OS and PFS were 4.5 and 1.8 months, respectively (86). A phase II study of temsirolimus in gemcitabine refractory PDAC patients was closed to accrual due to significant adverse effects and median OS and PFS were 44 and 19 days, respectively (87). Considering the disappointing results of mTOR inhibitors as monotherapy, several clinical studies evaluated mTOR inhibitors in combination with other cytotoxic or targeted agents.

A phase II study of everolimus plus erlotinib in patients with advanced PDAC who received at least one prior gemcitabine-based regimen showed minimal clinical activity with an OS of 87 days and a PFS of 49 days (87). The oral regimen with the combination of everolimus and capecitabine in a phase II trial of patients with advanced PDAC who were untreated (first-line) or had prior chemotherapy (second-line) demonstrated a moderate activity (median OS of 8.9 months and a PFS of 3.6 months) with an acceptable toxicity profile (88). Temsirolimus plus gemcitabine in previously untreated patients with advanced PDAC failed to show any meaningful clinical response in a phase II study (OS, 4.95 months; PFS, 2.69 months) (89). Furthermore, the combination of temsirolimus and docetaxel in patients with refractory solid tumors including PDAC did not meet its primary objective due to dose-limiting toxicities in a phase I study (90).

The combination of PI3K/AKT/mTOR inhibitors with other targeted therapies has also been evaluated in several clinical studies. A phase I study evaluated everolimus and the CDK4/6 inhibitor ribociclib as a third-line therapy in patients with PDAC after disease progression on both 5-fluorouracil and gemcitabine. This study demonstrated favorable tolerability with a decrease in CDK4/6-regulated genes but median OS and PFS were only 3.7 and 1.8 months, respectively (91). A phase I clinical study is currently ongoing to evaluate the combination of the PI3K/mTOR inhibitor, gedatolisib, and the CDK4/6 inhibitor, palbociclib, in patients with solid tumors (including PDAC) that is metastatic or unresectable and resistant to standard therapy (NCT03065062). Based on suggestive crosstalk between PI3K/AKT/mTOR and hedgehog (Hh) signaling, the mTOR inhibitor sirolimus was assessed in combination with the Hh inhibitor vismodegib in patients with advanced PDAC. However, in contrast with preclinical results, the combination of vismodegib and sirolimus only showed stable disease in a subgroup of patients (92). EGFR overexpression has been reported in up to 95% of patients with PDAC. The small molecule EGFR inhibitor erlotinib in combination with gemcitabine is an approved therapy for PDAC (93). Because PI3K/mTOR signaling is a well-established resistance mechanism to erlotinib, the combination of mTOR inhibitors and erlotinib was assessed in PDAC clinical trials. However, the combination of erlotinib with everolimus (87) or temsirolimus (94) did not demonstrate any meaningful clinical benefit. In addition, a phase I/II study of the combinations of temsirolimus with the EGFR monoclonal antibody cetuximab and capecitabine in patients with advanced PDAC demonstrated no clinical benefit with a median OS of 5 months (95).

RAS/RAF/MEK/ERK pathway inhibition

Mutations of KRAS or BRAF play a major role in the activation of the MAPK pathway. The role of inhibitors of the MAPK pathway has been investigated extensively in clinical studies for improving PDAC therapy (Table II).

Among FTIs, tipifarnib (R115777) has been assessed in PDAC clinical trials. In a phase II study, treatment with tipifarnib in previously untreated patients with metastatic PDAC resulted in partial inhibition of farnesyltransferase activity but it did not exhibit antitumor activity (median OS, 19.7 weeks; PFS, 4.9 weeks; and the estimated 6-month survival rate was 25%) (96). Another phase II study of tipifarnib in previously untreated patients with metastatic PDAC did not show any clinical efficacy and the median OS, PFS and 6-month survival rate were 2.6 months, 1.4 months and 19%, respectively (97). In a phase III trial, the tipifarnib combination with gemcitabine did not exhibit any clinical benefit compared with gemcitabine alone (median OS, 193 vs. 182 days; median PFS, 112 vs. 109 days; 6-month and 1-year survival rates were 53 and 27 vs. 49 and 24%, respectively) (98). In a phase II trial, weekly paclitaxel, gemcitabine and radiation (CXRT) was compared with CXRT plus maintenance tipifarnib. This study demonstrated that the addition of tipifarnib was associated with a broad range of toxicities and there was no clinical benefit (median OS, 11.5 vs. 8.9 months for the CXRT and CXRT + tipifarnib, respectively) (99).

Raf, a signaling protein downstream of Ras, is an important target for cancer therapy. Sorafenib (BAY 43-9006) was extensively assessed in combination with chemotherapy in several clinical studies of metastatic PDAC. A phase I study of sorafenib plus gemcitabine in advanced unresectable or metastatic PDAC revealed favorable tolerability with 56.6% of patients (n=13) achieving disease stabilization (100). Subsequently, a phase II trial of sorafenib alone versus sorafenib plus gemcitabine in patients with metastatic PDAC who received no prior chemotherapy or completed prior adjuvant chemotherapy >6 months before study entry, demonstrated no clinical benefit (median OS, 4.3 vs. 6.5 months; median PFS, 2.3 vs. 2.9 months) (101). A phase III trial of gemcitabine plus sorafenib and gemcitabine plus placebo in previously untreated patients with advanced PDAC exhibited a median OS of 9.2 and 8 months, and a median PFS of 5.7 and 3.8 months (102). Another phase II study of cisplatin plus gemcitabine with and without sorafenib in patients with metastatic PDAC also showed no clinical activity of sorafenib addition (median OS, 7.5 vs. 8.3 months; median PFS, 4.3 vs. 4.5 months) (103). A phase I study demonstrated modest clinical activity of sorafenib with concurrent radiation therapy and gemcitabine in advanced PDAC patients. The median OS and PFS for 25 evaluable patients were 12.6 and 10.6 months, respectively (104).

A phase II trial of gemcitabine plus trametinib or placebo in previously untreated patients with PDAC revealed no significant clinical benefits with an observed median OS of 8.4 vs. 6.7 months (P=0.453), a median PFS of 16 vs. 15 weeks (P=0.349), and an overall response rate 22 vs. 18%. These outcomes were independent of KRAS mutations determined by circulating free DNA (105). A phase I study of patients with KRAS-mutant solid tumors including PDAC showed manageable toxicity of trametinib plus lapatinib but there was no preliminary sign of antitumor activity in patients with PDAC (106). Based on preclinical evidence of synergistic antitumor response of MEK and EGFR inhibition in PDAC, a phase II study evaluated trametinib plus erlotinib in chemotherapy-refractory PDAC patients. This trial demonstrated a modest clinical efficacy of this combination in PDAC (median OS, 7.3 months; median PFS, 1.9 months) (107). Recently, a phase II study in locally recurrent, KRAS-mutant and PD-1 positive PDAC patients after surgery followed by chemotherapy (mFOLFIRINOX or 5-FU) demonstrated promising clinical efficacy after receiving the combination of radiotherapy plus pembrolizumab and trametinib (median OS and PFS of 14.9 and 8.2 months, respectively) compared with radiotherapy plus gemcitabine (median OS and PFS of 12.8 and 5.4 months, respectively) (108). Recently, a phase II trial of patients with advanced PDAC whose disease progressed after first-line chemotherapy with the combination treatment of trametinib and an oral FAK inhibitor, GSK2256098, demonstrated dismal clinical activity with a median PFS of 1.6 months and an OS of 3.6 months (109). A phase I trial is currently ongoing to evaluate trametinib and hydroxychloroquine in patients with PDAC (NCT03825289).

The MEK inhibitor, selumetinib, was compared with capecitabine in a phase II study in patients with PDAC who failed first-line gemcitabine therapy. In this trial, selumetinib was well tolerated with a manageable safety profile but showed no significant difference in OS compared with capecitabine (5.4 vs. 5.0 months) (110). A recent phase II study assessing selumetinib in KRASG12R-mutant PDAC patients who received two or more lines of systemic chemotherapy (~87.5% patients) demonstrated dismal clinical activity, revealing a 3-month median PFS and a 9-month median OS (111). A phase I study evaluating the MEK inhibitor, binimetinib plus the anti-PD-L1 antibody avelumab in patients with metastatic PDAC after 1–2 prior lines of therapy was terminated due to toxicity issues (112). A phase I study of binimetinib plus hydroxychloroquine in patients with metastatic PDAC who had at least one line of systemic therapy and harbor a KRAS mutation is currently ongoing (113).

The ERK inhibitor ulixertinib in combination with gemcitabine plus nab-paclitaxel in untreated patients with metastatic PDAC showed potentially similar efficacy (the median PFS and OS were 5.46 and 12.23 months, respectively) as chemotherapy and the study was terminated due to treatment-related adverse events (114). A phase I study of ulixertinib and palbociclib is currently ongoing in patients with advanced solid tumors including patients with PDAC who have received at least one line of therapy in the metastatic setting (115).

Attributed to the synergistic antitumor response in preclinical studies, simultaneous targeting of MAPK and PI3K pathways was assessed in several clinical studies in patients with KRAS-mutant PDAC. A phase I clinical trial of patients with solid tumors including PDAC with the PI3K/mTOR inhibitor GSK2126458 and trametinib demonstrated poor tolerability and limited antitumor efficacy (only 2 out of 7 patients with PDAC had stable disease) (116). As aforementioned in the previous section, a phase II study on gemcitabine-refractory PDAC patients demonstrated no clinical benefit of MK-2206 plus selumetinib therapy compared with mFOLFOX (84). Furthermore, a phase I study evaluating the combination of BKM120 plus binimetinib did not report any clinical efficacy in patients with advanced PDAC (117).

Direct KRAS targeting: New developments

Until recently, direct KRAS targeting by small molecule inhibitors was not possible and indirect therapeutic strategies including targeting RAS signaling and metabolic pathways demonstrated disappointing results in clinical trials in treating patients with KRAS-mutant cancers. Therefore, recently, numerous drugs with the potential of direct KRAS targeting have been evaluated in KRAS-mutant cancers including PDAC (Table III).

Table III.

Efficacy of direct KRAS inhibitors in PDAC.

Table III.

Efficacy of direct KRAS inhibitors in PDAC.

DrugTargetEffects in preclinical studiesEffects in clinical studies(Refs.)
Sotorasib (AMG510) KRASG12CAntitumor efficacy, synergistic effects with a proapoptotic agent DT2216 (in vitro, CDX)Phase I/II, KRASG12C-mutant PDAC patients who had at least 1 systemic therapy: OS 6.9 mo, PFS 4 mo(122,123)
Adagrasib (MRTX849) KRASG12CN/APhase I/II, KRASG12C-mutant previously treated PDAC patients: PFS 6.6 mo, treatment ongoing in 50% of patients(124)
MRTX1133 KRASG12D KRASG12D-mutant PDAC: tumor regression (≥30%) in CDX and PDX models; antitumor activity in immunocompetent modelsN/A(127,128)
RMC-6236Cyclophilin ASignificant antitumor efficacy in multiple CDX and PDX modelsOngoing phase I study in solid tumor patients with specific KRAS mutations(131, NCT05379985)
RMC-4550SHP-2Antitumor efficacy in combination with ERK inhibitor LY3214996 in vitro and multiple CDX and PDX modelsOngoing phase I study in solid tumor patients with KRAS mutations(132, NCT04916236)
BI 1701963SOS1N/APhase I, KRAS-mutant patients: with trametinib, study ongoing; with adagrasib, study completed, results awaited(133, NCT04975256)
mRNA-5671/V941 KRASG12D KRASG12V KRASG13D KRASG12CN/APhase I, KRAS-mutant solid tumor patients including PDAC: monotherapy and in combination with pembrolizumab, study completed, results awaited(NCT03948763)

[i] KRAS, Kirsten rat sarcoma viral oncogene homolog; PDAC, pancreatic ductal adenocarcinoma; CDX, cell-derived xenografts; PDX, patient-derived xenografts; N/A, not applicable; PFS, progression-free survival; OS, overall survival; mo, months.

Advancements in drug discovery and significant efforts by several groups led to a breakthrough by successfully developing compounds to target the KRASG12C-mutant allele (118), paving the way for the development of two inhibitors of this class, sotorasib (AMG510) (119) and adagrasib (MRTX849) (120). These two inhibitors are more relevant in NSCLC where the mutation frequency of KRASG12C is 13.8% but less relevant in colorectal and pancreatic cancer where KRASG12C mutation is 3.2% and <1%, respectively.

Sotorasib demonstrated significant clinical activity in a phase II trial in KRASG12C-mutated lung cancer patients and received FDA approval for this indication (121). In preclinical models (in vitro tumor cell lines and in vivo tumor cell-derived xenografts) of KRASG12C-mutant cancers including PDAC, sotorasib exhibited antitumor activity and significantly synergized with the proapoptotic agent DT2216 (122). In a phase I/II trial in patients with KRASG12C-mutant PDAC who had received at least one previous systemic therapy, sotorasib demonstrated an acceptable safety profile and promising antitumor activity (median PFS, 4 months; median OS, 6.9 months) (123). Adagrasib also exhibited clinical efficacy in a proof-of-concept study in KRASG12C-mutated lung and colon adenocarcinoma patients (120). In a phase I/II (KRYSTAL-1) trial in previously treated patients with metastatic PDAC harboring KRASG12C mutation, adagrasib demonstrated clinical efficacy in all 10 evaluable patients. Median PFS was 6.6 months, and treatment was ongoing in 50% of patients with PDAC at the time of data reporting (124). These findings are highly encouraging given that the median PFS was <3 months and a response rate of 10% with approved second-line therapy for advanced PDAC (125).

Given that the KRASG12D mutation is the most predominant variant (~41%) in PDAC, KRASG12D inhibitors have a high therapeutic potential for PDAC clinical therapy. MRTX1133 (126), a small-molecule inhibitor for KRASG12D, demonstrated marked tumor regression (≥30%) in 8 out of 11 KRASG12D-mutated PDAC CDX and PDX (127). Recently, Kemp et al demonstrated the specificity, potency and efficacy of MRTX1133 in immunocompetent KRASG12D mutant PDAC models. In this study, MRTX1133 exhibited marked tumor regression in all in vivo models tested (128). Several other groups are also developing KRASG12D mutant-specific inhibitors (129).

KRAS missense mutation variants in human malignancies are in the following order, G12D (35%), G12V (29%), G12C (21%), G12A (7%) and G12S (3%), indicating that recent advancements in targeting KRASG12C mutated tumors represent a relatively small portion of KRAS-mutant tumors. Therefore, targeting all KRAS mutation alleles with a single drug may be an ideal therapeutic strategy for most solid tumors where relative frequencies of different KRAS mutation subtypes are very diverse (130). A small molecule pan-RAS inhibitor RMC-6236, has been reported, that binds to an intracellular chaperone protein, cyclophilin A, resulting in an inhibitory binary complex that binds active GTP-bound RAS to form an inactive tricomplex, suppressing RAS signaling by disrupting the interaction with signaling partners such as RAF kinases. In a preclinical study, RMC-6236, an inhibitor of multiple RAS mutations and wild-type RAS, demonstrated profound and durable tumor regression in multiple CDX and PDX models of KRAS-mutant PDAC (131). A phase I study of RMC-6236 in patients with solid tumors harboring specific KRAS mutations (including KRAS G12A, G12D, G12V, G12R, G12S; excluding G12C) is currently ongoing (NCT05379985). Recently, the combined inhibition of SHP2, upstream of KRAS, using the inhibitor RMC-4550 and of ERK, downstream of KRAS, using LY3214996, demonstrated significant tumor growth inhibition in multiple PDAC mouse models including CDX, PDX and orthotopic mouse models (132). Based on these results, a phase I trial is currently enrolling patients with KRAS-mutant solid tumors including PDAC to evaluate this drug combination (NCT04916236). In addition, a novel pan-KRAS inhibitor, BI 1701963, an inhibitor of the son of sevenless 1 protein that is a guanine nucleotide exchange factor involved in the activation of KRAS, is currently being investigated alone and in combination with trametinib in a phase I trial of patients with advanced solid tumors including PDAC harboring all major KRAS mutation subtypes (133). Recently, a phase I trial of BI 1701963 in combination with adagrasib has been completed in patients with KRASG12-mutant advanced solid tumors and results are awaited (NCT04975256). Furthermore, results are also awaited for a recently completed phase I study evaluating an mRNA-based vaccine (mRNA-5671/V941), targeting G12D, G12V, G13D or G12C KRAS mutation subtypes, as monotherapy and in combination with pembrolizumab in patients with KRAS-mutant advanced or metastatic NSCLC, colorectal cancer or PDAC (NCT03948763).

Conclusions and future perspectives

Targeting KRAS signaling has great potential for therapeutic intervention in several solid tumors including PDAC. While downstream effector signal inhibitors have shown promise in preclinical studies, these drugs displayed limited clinical benefits, largely due to differences in the TME in these settings leading to enhanced toxicity and induction of adaptive resistance mechanisms in patients. Recent advances in preclinical models such as PDX and tumor organoids which better recapitulate patient tumor biology have helped design promising combination therapies targeting downstream KRAS effectors to overcome toxicity and resistance issues and improve PDAC clinical therapy. A combination of BRAF inhibitor vemurafenib plus sorafenib (phase II), binimetinib plus hydroxychloroquine, and binimetinib plus palbociclib are currently under clinical investigation (NCT05068752, NCT04132505, NCT04870034) in KRAS-mutant PDAC patients. Due to limited success with downstream KRAS inhibition strategy, attention has recently shifted to direct mutant-specific KRAS inhibitors and pan-KRAS inhibitors. Novel drugs such as sotorasib and adagrasib have been effective at inhibiting KRAS at G12C, but these are largely ineffective in PDAC as the major mutant isoforms are found at other locations. Recent advancements in KRASG12D-mutant-specific inhibitors (such as MRTX1133) are very encouraging and their success could be a game changer in PDAC clinical therapy. A phase I study is evaluating mesenchymal stromal cells-derived exosomes with KrasG12DsiRNA (iExosomes) in metastatic PDAC patients with KRASG12D mutation (NCT03608631). In addition, the upstream pan-KRAS inhibitors RMC-6236, RMC-4550 and BI 1701963 are currently under clinical investigation in KRAS-mutant PDAC (NCT05379985, NCT04916236, NCT04975256). These pan-KRAS inhibitors that have the ability to target a broad range of oncogenic KRAS variants, including all major G12 and G13 subtypes, could make a marked clinical impact in the majority of patients with PDAC irrespective of their KRAS mutation subtypes. Additionally, KRAS targeting therapeutic vaccines including peptides, dendritic cells and mRNA vaccines have recently emerged as a promising therapy and these are currently under clinical investigation (NCT05013216, NCT03592888, NCT03948763) for potential improvement of PDAC clinical therapy.

Acknowledgements

Not applicable.

Funding

The present study was financially supported by Indiana University School of Medicine funds.

Availability of data and materials

Not applicable.

Authors' contributions

JZ and NA contributed to the conception and design of the article and interpretation of the relevant literature. JZ and NA prepared the original draft of the manuscript. JZ, LD, MSH and NA performed formal analysis. JZ, UvH and NA wrote, reviewed, and edited the manuscript. NA acquired funding. Data authentication is not applicable. All authors have read and approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

PDAC

pancreatic ductal adenocarcinoma

KRAS

Kirsten rat sarcoma viral oncogene homolog

NSCLC

non-small cell lung cancer

GTP

guanosine triphosphate

GDP

guanosine diphosphate

RTK

receptor tyrosine kinases

PanIN

pancreatic intraepithelial neoplasia

mTOR

mammalian target of rapamycin

PDX

patient-derived xenograft

FTI

farnesyltransferase inhibitor

References

1 

Cox AD and Der CJ: Ras history: The saga continues. Small GTPases. 1:2–27. 2010. View Article : Google Scholar : PubMed/NCBI

2 

Bryant KL, Mancias JD, Kimmelman AC and Der CJ: KRAS: Feeding pancreatic cancer proliferation. Trends Biochem Sci. 39:91–100. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Hobbs GA, Der CJ and Rossman KL: RAS isoforms and mutations in cancer at a glance. J Cell Sci. 129:1287–1292. 2016.PubMed/NCBI

4 

Zhu C, Guan X, Zhang X, Luan X, Song Z, Cheng X, Zhang W and Qin JJ: Targeting KRAS mutant cancers: From druggable therapy to drug resistance. Mol Cancer. 21:1592022. View Article : Google Scholar : PubMed/NCBI

5 

Siegel RL, Miller KD, Fuchs HE and Jemal A: Cancer statistics, 2021. CA Cancer J Clin. 71:7–33. 2021. View Article : Google Scholar : PubMed/NCBI

6 

Waters AM and Der CJ: KRAS: The critical driver and therapeutic target for pancreatic cancer. Cold Spring Harb Perspect Med. 8:a0314352018. View Article : Google Scholar : PubMed/NCBI

7 

Aguirre AJ, Bardeesy N, Sinha M, Lopez L, Tuveson DA, Horner J, Redston MS and DePinho RA: Activated Kras and Ink4a/Arf deficiency cooperate to produce metastatic pancreatic ductal adenocarcinoma. Genes Dev. 17:3112–3126. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, Lengrand J, Deshpande V, Selig MK, Ferrone CR, et al: Transcriptional control of autophagy-lysosome function drives pancreatic cancer metabolism. Nature. 524:361–365. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Yang S, Wang X, Contino G, Liesa M, Sahin E, Ying H, Bause A, Li Y, Stommel JM, Dell'antonio G, et al: Pancreatic cancers require autophagy for tumor growth. Genes Dev. 25:717–729. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Buscail L, Bournet B and Cordelier P: Role of oncogenic KRAS in the diagnosis, prognosis and treatment of pancreatic cancer. Nat Rev Gastroenterol Hepatol. 17:153–168. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Dey P, Kimmelman AC and DePinho RA: Metabolic Codependencies in the tumor microenvironment. Cancer Discov. 11:1067–1081. 2021. View Article : Google Scholar : PubMed/NCBI

12 

Shen X, Niu N and Xue J: Oncogenic KRAS triggers metabolic reprogramming in pancreatic ductal adenocarcinoma. J Transl Int Medicine 0. -. 2022. View Article : Google Scholar

13 

Muthalagu N, Monteverde T, Raffo-Iraolagoitia X, Wiesheu R, Whyte D, Hedley A, Laing S, Kruspig B, Upstill-Goddard R, Shaw R, et al: Repression of the type I interferon pathway underlies MYC- and KRAS-dependent evasion of NK and B cells in pancreatic ductal adenocarcinoma. Cancer Discov. 10:872–887. 2020. View Article : Google Scholar : PubMed/NCBI

14 

Dey P, Li J, Zhang J, Chaurasiya A, Strom A, Wang H, Liao WT, Cavallaro F, Denz P, Bernard V, et al: Oncogenic KRAS-driven metabolic reprogramming in pancreatic cancer cells utilizes cytokines from the tumor microenvironment. Cancer Discov. 10:608–625. 2020. View Article : Google Scholar : PubMed/NCBI

15 

Santana-Codina N, Roeth AA, Zhang Y, Yang A, Mashadova O, Asara JM, Wang X, Bronson RT, Lyssiotis CA, Ying H and Kimmelman AC: Oncogenic KRAS supports pancreatic cancer through regulation of nucleotide synthesis. Nat Commun. 9:49452018. View Article : Google Scholar : PubMed/NCBI

16 

Son J, Lyssiotis CA, Ying H, Wang X, Hua S, Ligorio M, Perera RM, Ferrone CR, Mullarky E, Shyh-Chang N, et al: Glutamine supports pancreatic cancer growth through a KRAS-regulated metabolic pathway. Nature. 496:101–105. 2013. View Article : Google Scholar : PubMed/NCBI

17 

Raho S, Capobianco L, Malivindi R, Vozza A, Piazzolla C, Leonardis FD, Gorgoglione R, Scarcia P, Pezzuto F, Agrimi G, et al: KRAS-regulated glutamine metabolism requires UCP2-mediated aspartate transport to support pancreatic cancer growth. Nat Metab. 2:1373–1381. 2020. View Article : Google Scholar : PubMed/NCBI

18 

Kessler D, Gmachl M, Mantoulidis A, Martin LJ, Zoephel A, Mayer M, Gollner A, Covini D, Fischer S, Gerstberger T, et al: Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci USA. 116:15823–15829. 2019. View Article : Google Scholar : PubMed/NCBI

19 

Vigil D, Cherfils J, Rossman KL and Der CJ: Ras superfamily GEFs and GAPs: Validated and tractable targets for cancer therapy? Nat Rev Cancer. 10:842–857. 2010. View Article : Google Scholar : PubMed/NCBI

20 

Cuesta C, Arévalo-Alameda C and Castellano E: The importance of being PI3K in the RAS signaling network. Genes (Basel). 12:10942021. View Article : Google Scholar : PubMed/NCBI

21 

Krygowska AA and Castellano E: PI3K: A crucial piece in the RAS signaling puzzle. Cold Spring Harb Perspect Med. 8:a0314502018. View Article : Google Scholar : PubMed/NCBI

22 

Hemmings BA and Restuccia DF: PI3K-PKB/Akt pathway. Cold Spring Harb Perspect Biol. 4:a0111892012. View Article : Google Scholar : PubMed/NCBI

23 

Fritsch R and Downward J: SnapShot: Class I PI3K isoform signaling. Cell. 154:940–940.e941. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Peng Y, Wang Y, Zhou C, Mei W and Zeng C: PI3K/Akt/mTOR pathway and its role in cancer therapeutics: Are we making headway? Front Oncol. 12:8191282022. View Article : Google Scholar : PubMed/NCBI

25 

Rascio F, Spadaccino F, Rocchetti MT, Castellano G, Stallone G, Netti GS and Ranieri E: The pathogenic role of PI3K/AKT pathway in cancer onset and drug resistance: An updated review. Cancers (Basel). 13:39492021. View Article : Google Scholar : PubMed/NCBI

26 

Witkiewicz AK, McMillan EA, Balaji U, Baek G, Lin WC, Mansour J, Mollaee M, Wagner KU, Koduru P, Yopp A, et al: Whole-exome sequencing of pancreatic cancer defines genetic diversity and therapeutic targets. Nat Commun. 6:67442015. View Article : Google Scholar : PubMed/NCBI

27 

Hayes TK, Neel NF, Hu C, Gautam P, Chenard M, Long B, Aziz M, Kassner M, Bryant KL, Pierobon M, et al: Long-term ERK inhibition in KRAS-mutant pancreatic cancer is associated with MYC degradation and senescence-like growth suppression. Cancer Cell. 29:75–89. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Workman P, Clarke PA, Raynaud FI and van Montfort RL: Drugging the PI3 kinome: From chemical tools to drugs in the clinic. Cancer Res. 70:2146–2157. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Ng SS, Tsao MS, Nicklee T and Hedley DW: Wortmannin inhibits pkb/akt phosphorylation and promotes gemcitabine antitumor activity in orthotopic human pancreatic cancer xenografts in immunodeficient mice. Clin Cancer Res. 7:3269–3275. 2001.PubMed/NCBI

30 

Fujiwara M, Izuishi K, Sano T, Hossain MA, Kimura S, Masaki T and Suzuki Y: Modulating effect of the PI3-kinase inhibitor LY294002 on cisplatin in human pancreatic cancer cells. J Exp Clin Cancer Res. 27:762008. View Article : Google Scholar : PubMed/NCBI

31 

Wang Y, Kuramitsu Y, Baron B, Kitagawa T, Tokuda K, Akada J, Maehara SI, Maehara Y and Nakamura K: PI3K inhibitor LY294002, as opposed to wortmannin, enhances AKT phosphorylation in gemcitabine-resistant pancreatic cancer cells. Int J Oncol. 50:606–612. 2017. View Article : Google Scholar : PubMed/NCBI

32 

Wang Z, Luo G and Qiu Z: Akt inhibitor MK-2206 reduces pancreatic cancer cell viability and increases the efficacy of gemcitabine. Oncol Lett. 19:1999–2004. 2020.PubMed/NCBI

33 

Hu C, Dadon T, Chenna V, Yabuuchi S, Bannerji R, Booher R, Strack P, Azad N, Nelkin BD and Maitra A: Combined inhibition of cyclin-dependent kinases (Dinaciclib) and AKT (MK-2206) blocks pancreatic tumor growth and metastases in patient-derived xenograft models. Mol Cancer Ther. 14:1532–1539. 2015. View Article : Google Scholar : PubMed/NCBI

34 

Massihnia D, Avan A, Funel N, Maftouh M, van Krieken A, Granchi C, Raktoe R, Boggi U, Aicher B, Minutolo F, et al: Phospho-Akt overexpression is prognostic and can be used to tailor the synergistic interaction of Akt inhibitors with gemcitabine in pancreatic cancer. J Hematol Oncol. 10:92017. View Article : Google Scholar : PubMed/NCBI

35 

Utomo WK, Narayanan V, Biermann K, van Eijck CHJ, Bruno MJ, Peppelenbosch MP and Braat H: mTOR is a promising therapeutical target in a subpopulation of pancreatic adenocarcinoma. Cancer Lett. 346:309–317. 2014. View Article : Google Scholar : PubMed/NCBI

36 

Soares HP, Ni Y, Kisfalvi K, Sinnett-Smith J and Rozengurt E: Different patterns of Akt and ERK feedback activation in response to rapamycin, active-site mTOR inhibitors and metformin in pancreatic cancer cells. PLoS One. 8:e572892013. View Article : Google Scholar : PubMed/NCBI

37 

Dai ZJ, Gao J, Kang HF, Ma YG, Ma XB, Lu WF, Lin S, Ma HB, Wang XJ and Wu WY: Targeted inhibition of mammalian target of rapamycin (mTOR) enhances radiosensitivity in pancreatic carcinoma cells. Drug Des Devel Ther. 7:149–159. 2013. View Article : Google Scholar : PubMed/NCBI

38 

Azzariti A, Porcelli L, Gatti G, Nicolin A and Paradiso A: Synergic antiproliferative and antiangiogenic effects of EGFR and mTor inhibitors on pancreatic cancer cells. Biochem Pharmacol. 75:1035–1044. 2008. View Article : Google Scholar : PubMed/NCBI

39 

Morran DC, Wu J, Jamieson NB, Mrowinska A, Kalna G, Karim SA, Au AYM, Scarlett CJ, Chang DK, Pajak MZ, et al: Targeting mTOR dependency in pancreatic cancer. Gut. 63:1481–1489. 2014. View Article : Google Scholar : PubMed/NCBI

40 

Cifarelli V, Lashinger LM, Devlin KL, Dunlap SM, Huang J, Kaaks R, Pollak MN and Hursting SD: Metformin and rapamycin reduce pancreatic cancer growth in obese prediabetic mice by distinct MicroRNA-regulated mechanisms. Diabetes. 64:1632–1642. 2015. View Article : Google Scholar : PubMed/NCBI

41 

Tuncyurek P, Mayer JM, Klug F, Dillmann S, Henne-Bruns D, Keller F and Stracke S: Everolimus and mycophenolate mofetil sensitize human pancreatic cancer cells to gemcitabine in vitro: A novel adjunct to standard chemotherapy? Eur Surg Res. 39:380–387. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Peng T and Dou QP: Everolimus inhibits growth of gemcitabine-resistant pancreatic cancer cells via induction of caspase-dependent apoptosis and G(2) /M arrest. J Cell Biochem. 118:2722–2730. 2017. View Article : Google Scholar : PubMed/NCBI

43 

Pawaskar DK, Straubinger RM, Fetterly GJ, Ma WW and Jusko WJ: Interactions of everolimus and sorafenib in pancreatic cancer cells. AAPS J. 15:78–84. 2013. View Article : Google Scholar : PubMed/NCBI

44 

Wei F, Zhang Y, Geng L, Zhang P, Wang G and Liu Y: mTOR inhibition induces EGFR feedback activation in association with its resistance to human pancreatic cancer. Int J Mol Sci. 16:3267–3282. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Pawaskar DK, Straubinger RM, Fetterly GJ, Hylander BH, Repasky EA, Ma WW and Jusko WJ: Synergistic interactions between sorafenib and everolimus in pancreatic cancer xenografts in mice. Cancer Chemother Pharmacol. 71:1231–1240. 2013. View Article : Google Scholar : PubMed/NCBI

46 

Witkiewicz AK, Balaji U, Eslinger C, McMillan E, Conway W, Posner B, Mills GB, O'Reilly EM and Knudsen ES: Integrated patient-derived models delineate individualized therapeutic vulnerabilities of pancreatic cancer. Cell Rep. 16:2017–2031. 2016. View Article : Google Scholar : PubMed/NCBI

47 

Asano T, Yao Y, Zhu J, Li D, Abbruzzese JL and Reddy SA: The rapamycin analog CCI-779 is a potent inhibitor of pancreatic cancer cell proliferation. Biochem Biophys Res Commun. 331:295–302. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Ito D, Fujimoto K, Mori T, Kami K, Koizumi M, Toyoda E, Kawaguchi Y and Doi R: In vivo antitumor effect of the mTOR inhibitor CCI-779 and gemcitabine in xenograft models of human pancreatic cancer. Int J Cancer. 118:2337–2343. 2006. View Article : Google Scholar : PubMed/NCBI

49 

Cao P, Maira SM, García-Echeverría C and Hedley DW: Activity of a novel, dual PI3-kinase/mTor inhibitor NVP-BEZ235 against primary human pancreatic cancers grown as orthotopic xenografts. Br J Cancer. 100:1267–1276. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Awasthi N, Yen PL, Schwarz MA and Schwarz RE: The efficacy of a novel, dual PI3K/mTOR inhibitor NVP-BEZ235 to enhance chemotherapy and antiangiogenic response in pancreatic cancer. J Cell Biochem. 113:784–791. 2012. View Article : Google Scholar : PubMed/NCBI

51 

Venkannagari S, Fiskus W, Peth K, Atadja P, Hidalgo M, Maitra A and Bhalla KN: Superior efficacy of co-treatment with dual PI3K/mTOR inhibitor NVP-BEZ235 and pan-histone deacetylase inhibitor against human pancreatic cancer. Oncotarget. 3:1416–1427. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Lee JE, Woo MG, Jung KH, Shin SM, Son MK, Fang Z, Yan HH, Park JH, Yoon YC, Kim YS and Hong SS: Combination therapy of the active KRAS-targeting antibody inRas37 and a PI3K inhibitor in pancreatic cancer. Biomol Ther (Seoul). 30:274–283. 2022. View Article : Google Scholar : PubMed/NCBI

53 

Soares HP, Ming M, Mellon M, Young SH, Han L, Sinnet-Smith J and Rozengurt E: Dual PI3K/mTOR inhibitors induce rapid overactivation of the MEK/ERK pathway in human pancreatic cancer cells through suppression of mTORC2. Mol Cancer Ther. 14:1014–1023. 2015. View Article : Google Scholar : PubMed/NCBI

54 

Venkatasubbarao K, Choudary A and Freeman JW: Farnesyl transferase inhibitor (R115777)-induced inhibition of STAT3(Tyr705) phosphorylation in human pancreatic cancer cell lines require extracellular signal-regulated kinases. Cancer Res. 65:2861–2871. 2005. View Article : Google Scholar : PubMed/NCBI

55 

Shi B, Yaremko B, Hajian G, Terracina G, Bishop WR, Liu M and Nielsen LL: The farnesyl protein transferase inhibitor SCH66336 synergizes with taxanes in vitro and enhances their antitumor activity in vivo. Cancer Chemother Pharmacol. 46:387–393. 2000. View Article : Google Scholar : PubMed/NCBI

56 

Ulivi P, Arienti C, Amadori D, Fabbri F, Carloni S, Tesei A, Vannini I, Silvestrini R and Zoli W: Role of RAF/MEK/ERK pathway, p-STAT-3 and Mcl-1 in sorafenib activity in human pancreatic cancer cell lines. J Cell Physiol. 220:214–221. 2009. View Article : Google Scholar : PubMed/NCBI

57 

Wei G, Wang M and Carr BI: Sorafenib combined vitamin K induces apoptosis in human pancreatic cancer cell lines through RAF/MEK/ERK and c-Jun NH2-terminal kinase pathways. J Cell Physiol. 224:112–119. 2010. View Article : Google Scholar : PubMed/NCBI

58 

Fang Z, Jung KH, Yan HH, Kim SJ, Rumman M, Park JH, Han B, Lee JE, Kang YW, Lim JH and Hong SS: Melatonin synergizes with sorafenib to suppress pancreatic cancer via melatonin receptor and PDGFR-β/STAT3 pathway. Cell Physiol Biochem. 47:1751–1768. 2018. View Article : Google Scholar : PubMed/NCBI

59 

Awasthi N, Zhang C, Hinz S, Schwarz MA and Schwarz RE: Enhancing sorafenib-mediated sensitization to gemcitabine in experimental pancreatic cancer through EMAP II. J Exp Clin Cancer Res. 32:122013. View Article : Google Scholar : PubMed/NCBI

60 

Nakkina SP, Gitto SB, Beardsley JM, Pandey V, Rohr MW, Parikh JG, Phanstiel O IV and Altomare DA: DFMO improves survival and increases immune cell infiltration in association with MYC downregulation in the pancreatic tumor microenvironment. Int J Mol Sci. 22:131752021. View Article : Google Scholar : PubMed/NCBI

61 

Walters DM, Lindberg JM, Adair SJ, Newhook TE, Cowan CR, Stokes JB, Borgman CA, Stelow EB, Lowrey BT, Chopivsky ME, et al: Inhibition of the growth of patient-derived pancreatic cancer xenografts with the MEK inhibitor trametinib is augmented by combined treatment with the epidermal growth factor receptor/HER2 inhibitor lapatinib. Neoplasia. 15:143–155. 2013. View Article : Google Scholar : PubMed/NCBI

62 

Awasthi N, Monahan S, Stefaniak A, Schwarz MA and Schwarz RE: Inhibition of the MEK/ERK pathway augments nab-paclitaxel-based chemotherapy effects in preclinical models of pancreatic cancer. Oncotarget. 9:5274–5286. 2018. View Article : Google Scholar : PubMed/NCBI

63 

Chao MW, Chang LH, Tu HJ, Chang CD, Lai MJ, Chen YY, Liou JP, Teng CM and Pan SL: Combination treatment strategy for pancreatic cancer involving the novel HDAC inhibitor MPT0E028 with a MEK inhibitor beyond K-Ras status. Clin Epigenetics. 11:852019. View Article : Google Scholar : PubMed/NCBI

64 

Amada K, Hijiya N, Ikarimoto S, Yanagihara K, Hanada T, Hidano S, Kurogi S, Tsukamoto Y, Nakada C, Kinoshita K, et al: Involvement of clusterin expression in the refractory response of pancreatic cancer cells to a MEK inhibitor. Cancer Sci. 114:2189–2202. 2023. View Article : Google Scholar : PubMed/NCBI

65 

Zhou J, Zhao T, Ma L, Liang M, Guo YJ and Zhao LM: Cucurbitacin B and SCH772984 exhibit synergistic anti-pancreatic cancer activities by suppressing EGFR, PI3K/Akt/mTOR, STAT3 and ERK signaling. Oncotarget. 8:103167–103181. 2017. View Article : Google Scholar : PubMed/NCBI

66 

Bryant KL, Stalnecker CA, Zeitouni D, Klomp JE, Peng S, Tikunov AP, Gunda V, Pierobon M, Waters AM, George SD, et al: Combination of ERK and autophagy inhibition as a treatment approach for pancreatic cancer. Nat Med. 25:628–640. 2019. View Article : Google Scholar : PubMed/NCBI

67 

Yan Z, Ohuchida K, Fei S, Zheng B, Guan W, Feng H, Kibe S, Ando Y, Koikawa K, Abe T, et al: Inhibition of ERK1/2 in cancer-associated pancreatic stellate cells suppresses cancer-stromal interaction and metastasis. J Exp Clin Cancer Res. 38:2212019. View Article : Google Scholar : PubMed/NCBI

68 

Goodwin CM, Waters AM, Klomp JE, Javaid S, Bryant KL, Stalnecker CA, Drizyte-Miller K, Papke B, Yang R, Amparo AM, et al: Combination therapies with CDK4/6 inhibitors to treat KRAS-mutant pancreatic cancer. Cancer Res. 83:141–157. 2023. View Article : Google Scholar : PubMed/NCBI

69 

Jiang H, Xu M, Li L, Grierson P, Dodhiawala P, Highkin M, Zhang D, Li Q, Wang-Gillam A and Lim KH: Concurrent HER or PI3K inhibition potentiates the antitumor effect of the erk inhibitor ulixertinib in preclinical pancreatic cancer models. Mol Cancer Ther. 17:2144–2155. 2018. View Article : Google Scholar : PubMed/NCBI

70 

Williams TM, Flecha AR, Keller P, Ram A, Karnak D, Galbán S, Galbán CJ, Ross BD, Lawrence TS, Rehemtulla A and Sebolt-Leopold J: Cotargeting MAPK and PI3K signaling with concurrent radiotherapy as a strategy for the treatment of pancreatic cancer. Mol Cancer Ther. 11:1193–1202. 2012. View Article : Google Scholar : PubMed/NCBI

71 

Dai X, Zhang J, Arfuso F, Chinnathambi Zayed ME, Alharbi SA, Kumar AP, Ahn KS and Sethi G: Targeting TNF-related apoptosis-inducing ligand (TRAIL) receptor by natural products as a potential therapeutic approach for cancer therapy. Exp Biol Med (Maywood). 240:760–773. 2015. View Article : Google Scholar : PubMed/NCBI

72 

Zhang Q, Wang H, Ran L, Zhang Z and Jiang R: The preclinical evaluation of TIC10/ONC201 as an anti-pancreatic cancer agent. Biochem Biophys Res Commun. 476:260–266. 2016. View Article : Google Scholar : PubMed/NCBI

73 

Awasthi N, Kronenberger D, Stefaniak A, Hassan MS, von Holzen U, Schwarz MA and Schwarz RE: Dual inhibition of the PI3K and MAPK pathways enhances nab-paclitaxel/gemcitabine chemotherapy response in preclinical models of pancreatic cancer. Cancer Lett. 459:41–49. 2019. View Article : Google Scholar : PubMed/NCBI

74 

Thomas D and Radhakrishnan P: Tumor-stromal crosstalk in pancreatic cancer and tissue fibrosis. Mol Cancer. 18:142019. View Article : Google Scholar : PubMed/NCBI

75 

Krempley BD and Yu KH: Preclinical models of pancreatic ductal adenocarcinoma. Chin Clin Oncol. 6:252017. View Article : Google Scholar : PubMed/NCBI

76 

Yu Y, Yang G, Huang H, Fu Z, Cao Z, Zheng L, You L and Zhang T: Preclinical models of pancreatic ductal adenocarcinoma: Challenges and opportunities in the era of precision medicine. J Exp Clin Cancer Res. 40:82021. View Article : Google Scholar : PubMed/NCBI

77 

Kim RD, Alberts SR, Peña C, Genvresse I, Ajavon-Hartmann A, Xia C, Kelly A and Grilley-Olson JE: Phase I dose-escalation study of copanlisib in combination with gemcitabine or cisplatin plus gemcitabine in patients with advanced cancer. Br J Cancer. 118:462–470. 2018. View Article : Google Scholar : PubMed/NCBI

78 

Soares HP, Al-Toubah TE, Kim RD, Kim J, Lewis NK and Mahipal A: Final report: A phase I trial of BYL719 in combination with gemcitabine and nab-paclitaxel in locally advanced and metastatic pancreatic cancer. J Clin Oncol. 36:398. 2018. View Article : Google Scholar

79 

Bedard PL, Tabernero J, Janku F, Wainberg ZA, Paz-Ares L, Vansteenkiste J, Cutsem EV, Pérez-García J, Stathis A, Britten CD, et al: A phase Ib dose-escalation study of the oral pan-PI3K inhibitor buparlisib (BKM120) in combination with the oral MEK1/2 inhibitor trametinib (GSK1120212) in patients with selected advanced solid tumors. Clin Cancer Res. 21:730–738. 2015. View Article : Google Scholar : PubMed/NCBI

80 

McRee AJ, Sanoff HK, Carlson C, Ivanova A and O'Neil BH: A phase I trial of mFOLFOX6 combined with the oral PI3K inhibitor BKM120 in patients with advanced refractory solid tumors. Invest New Drugs. 33:1225–1231. 2015. View Article : Google Scholar : PubMed/NCBI

81 

Borazanci E, Pishvaian MJ, Nemunaitis J, Weekes C, Huang J and Rajakumaraswamy N: A Phase Ib study of single-agent idelalisib followed by idelalisib in combination with chemotherapy in patients with metastatic pancreatic ductal adenocarcinoma. Oncologist. 25:e1604–e1613. 2020. View Article : Google Scholar : PubMed/NCBI

82 

Marsh RW, Lima CM, Levy DE, Mitchell EP, Rowland KM Jr and Benson AB III: A phase II trial of perifosine in locally advanced, unresectable, or metastatic pancreatic adenocarcinoma. Am J Clin Oncol. 30:26–31. 2007. View Article : Google Scholar : PubMed/NCBI

83 

Hedley D, Moore MJ, Hirte H, Siu L, Vincent M, Jonker H. Mwang D, Nagai J and Dancey J: A phase II trial of perifosine as second line therapy for advanced pancreatic cancer. A study of the princess margaret hospital [PMH] phase II consortium. J Clin Oncol. 23:4166. 2005. View Article : Google Scholar

84 

Chung V, McDonough S, Philip PA, Cardin D, Wang-Gillam A, Hui L, Tejani MA, Seery TE, Dy IA, Al Baghdadi T, et al: Effect of Selumetinib and MK-2206 vs Oxaliplatin and Fluorouracil in patients with metastatic pancreatic cancer after prior therapy: SWOG S1115 study randomized clinical trial. JAMA Oncol. 3:516–522. 2017. View Article : Google Scholar : PubMed/NCBI

85 

Murphy AG, Zahurak M, Shah M, Weekes CD, Hansen A, Siu LL, Spreafico A, LoConte N, Anders NM, Miles T, et al: A phase I study of dinaciclib in combination with MK-2206 in patients with advanced pancreatic cancer. Clin Transl Sci. 13:1178–1188. 2020. View Article : Google Scholar : PubMed/NCBI

86 

Wolpin BM, Hezel AF, Abrams T, Blaszkowsky LS, Meyerhardt JA, Chan JA, Enzinger PC, Allen B, Clark JW, Ryan DP and Fuchs CS: Oral mTOR inhibitor everolimus in patients with gemcitabine-refractory metastatic pancreatic cancer. J Clin Oncol. 27:193–198. 2009. View Article : Google Scholar : PubMed/NCBI

87 

Javle MM, Shroff RT, Xiong H, Varadhachary GA, Fogelman D, Reddy SA, Davis D, Zhang Y, Wolff RA and Abbruzzese JL: Inhibition of the mammalian target of rapamycin (mTOR) in advanced pancreatic cancer: Results of two phase II studies. BMC Cancer. 10:3682010. View Article : Google Scholar : PubMed/NCBI

88 

Kordes S, Klümpen HJ, Weterman MJ, Schellens JH, Richel DJ and Wilmink JW: Phase II study of capecitabine and the oral mTOR inhibitor everolimus in patients with advanced pancreatic cancer. Cancer Chemother Pharmacol. 75:1135–1141. 2015. View Article : Google Scholar : PubMed/NCBI

89 

Karavasilis V, Samantas E, Koliou GA, Kalogera-Fountzila A, Pentheroudakis G, Varthalitis I, Linardou H, Rallis G, Skondra M, Papadopoulos G, et al: Gemcitabine combined with the mTOR inhibitor temsirolimus in patients with locally advanced or metastatic pancreatic cancer. A Hellenic cooperative oncology group phase I/II study. Target Oncol. 13:715–724. 2018. View Article : Google Scholar : PubMed/NCBI

90 

Amin M, Gao F, Terrero G, Picus J, Wang-Gillam A, Suresh R, Ma C, Tan B, Baggstrom M, Naughton MJ, et al: Phase I study of docetaxel and temsirolimus in refractory solid tumors. Am J Clin Oncol. 44:443–448. 2021. View Article : Google Scholar : PubMed/NCBI

91 

Weinberg BA, Wang H, Witkiewicz AK, Marshall JL, He AR, Vail P, Knudsen ES and Pishvaian MJ: A phase I study of ribociclib plus everolimus in patients with metastatic pancreatic adenocarcinoma refractory to chemotherapy. J Pancreat Cancer. 6:45–54. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Carr RM, Duma N, McCleary-Wheeler AL, Almada LL, Marks DL, Graham RP, Smyrk TC, Lowe V, Borad MJ, Kim G, et al: Targeting of the Hedgehog/GLI and mTOR pathways in advanced pancreatic cancer, a phase 1 trial of Vismodegib and Sirolimus combination. Pancreatology. 20:1115–1122. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, Au HJ, Murawa P, Walde D, Wolff RA, et al: Erlotinib plus gemcitabine compared with gemcitabine alone in patients with advanced pancreatic cancer: A phase III trial of the national cancer institute of Canada clinical trials group. J Clin Oncol. 25:1960–1966. 2007. View Article : Google Scholar : PubMed/NCBI

94 

Park H, Williams K, Trikalinos NA, Larson S, Tan B, Waqar S, Suresh R, Morgensztern D, Van Tine BA, Govindan R, et al: A phase I trial of temsirolimus and erlotinib in patients with refractory solid tumors. Cancer Chemother Pharmacol. 87:337–347. 2021. View Article : Google Scholar : PubMed/NCBI

95 

Kordes S, Richel DJ, Klümpen HJ, Weterman MJ, Stevens AJ and Wilmink JW: A phase I/II, non-randomized, feasibility/safety and efficacy study of the combination of everolimus, cetuximab and capecitabine in patients with advanced pancreatic cancer. Invest New Drugs. 31:85–91. 2013. View Article : Google Scholar : PubMed/NCBI

96 

Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, Lewis NL, McLaughlin S, Rogatko A, Perez-Ruixo JJ, et al: Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. J Clin Oncol. 21:1301–1306. 2003. View Article : Google Scholar : PubMed/NCBI

97 

Macdonald JS, McCoy S, Whitehead RP, Iqbal S, Wade JL III, Giguere JK and Abbruzzese JL: A phase II study of farnesyl transferase inhibitor R115777 in pancreatic cancer: A Southwest oncology group (SWOG 9924) study. Invest New Drugs. 23:485–487. 2005. View Article : Google Scholar : PubMed/NCBI

98 

Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, Schoffski P, Post S, Verslype C, Neumann H, et al: Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 22:1430–1438. 2004. View Article : Google Scholar : PubMed/NCBI

99 

Rich TA, Winter K, Safran H, Hoffman JP, Erickson B, Anne PR, Myerson RJ, Cline-Burkhardt VJ, Perez K and Willett C: Weekly paclitaxel, gemcitabine, and external irradiation followed by randomized farnesyl transferase inhibitor R115777 for locally advanced pancreatic cancer. Onco Targets Ther. 5:161–170. 2012. View Article : Google Scholar : PubMed/NCBI

100 

Siu LL, Awada A, Takimoto CH, Piccart M, Schwartz B, Giannaris T, Lathia C, Petrenciuc O and Moore MJ: Phase I trial of sorafenib and gemcitabine in advanced solid tumors with an expanded cohort in advanced pancreatic cancer. Clin Cancer Res. 12:144–151. 2006. View Article : Google Scholar : PubMed/NCBI

101 

El-Khoueiry AB, Ramanathan RK, Yang DY, Zhang W, Shibata S, Wright JJ, Gandara D and Lenz HJ: A randomized phase II of gemcitabine and sorafenib versus sorafenib alone in patients with metastatic pancreatic cancer. Invest New Drugs. 30:1175–1183. 2012. View Article : Google Scholar : PubMed/NCBI

102 

Gonçalves A, Gilabert M, François E, Dahan L, Perrier H, Lamy R, Re D, Largillier R, Gasmi M, Tchiknavorian X, et al: BAYPAN study: A double-blind phase III randomized trial comparing gemcitabine plus sorafenib and gemcitabine plus placebo in patients with advanced pancreatic cancer. Ann Oncol. 23:2799–2805. 2012. View Article : Google Scholar : PubMed/NCBI

103 

Cascinu S, Berardi R, Sobrero A, Bidoli P, Labianca R, Siena S, Ferrari D, Barni S, Aitini E, Zagonel V, et al: Sorafenib does not improve efficacy of chemotherapy in advanced pancreatic cancer: A GISCAD randomized phase II study. Dig Liver Dis. 46:182–186. 2014. View Article : Google Scholar : PubMed/NCBI

104 

Chiorean EG, Schneider BP, Akisik FM, Perkins SM, Anderson S, Johnson CS, DeWitt J, Helft P, Clark R, Johnston EL, et al: Phase 1 pharmacogenetic and pharmacodynamic study of sorafenib with concurrent radiation therapy and gemcitabine in locally advanced unresectable pancreatic cancer. Int J Radiat Oncol Biol Phys. 89:284–291. 2014. View Article : Google Scholar : PubMed/NCBI

105 

Infante JR, Somer BG, Park JO, Li CP, Scheulen ME, Kasubhai SM, Oh DY, Liu Y, Redhu S, Steplewski K and Le N: A randomised, double-blind, placebo-controlled trial of trametinib, an oral MEK inhibitor, in combination with gemcitabine for patients with untreated metastatic adenocarcinoma of the pancreas. Eur J Cancer. 50:2072–2081. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Huijberts S, van Geel R, van Brummelen EMJ, Opdam FL, Marchetti S, Steeghs N, Pulleman S, Thijssen B, Rosing H, Monkhorst K, et al: Phase I study of lapatinib plus trametinib in patients with KRAS-mutant colorectal, non-small cell lung, and pancreatic cancer. Cancer Chemother Pharmacol. 85:917–930. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Ko AH, Bekaii-Saab T, Van Ziffle J, Mirzoeva OM, Joseph NM, Talasaz A, Kuhn P, Tempero MA, Collisson EA, Kelley RK, et al: A multicenter, open-label phase II clinical trial of combined MEK plus EGFR inhibition for chemotherapy-refractory advanced pancreatic adenocarcinoma. Clin Cancer Res. 22:61–68. 2016. View Article : Google Scholar : PubMed/NCBI

108 

Zhu X, Cao Y, Liu W, Ju X, Zhao X, Jiang L, Ye Y, Jin G and Zhang H: Stereotactic body radiotherapy plus pembrolizumab and trametinib versus stereotactic body radiotherapy plus gemcitabine for locally recurrent pancreatic cancer after surgical resection: an open-label, randomised, controlled, phase 2 trial. Lancet Oncol. 23:e105–e115. 2022. View Article : Google Scholar : PubMed/NCBI

109 

Aung KL, McWhirter E, Welch S, Wang L, Lovell S, Stayner LA, Ali S, Malpage A, Makepeace B, Ramachandran M, et al: A phase II trial of GSK2256098 and trametinib in patients with advanced pancreatic ductal adenocarcinoma. J Gastrointest Oncol. 13:3216–3226. 2022. View Article : Google Scholar : PubMed/NCBI

110 

Bodoky G, Timcheva C, Spigel DR, Stella PJL, Ciuleanu TE, Pover G and Tebbutt NC: A phase II open-label randomized study to assess the efficacy and safety of selumetinib (AZD6244 [ARRY-142886]) versus capecitabine in patients with advanced or metastatic pancreatic cancer who have failed first-line gemcitabine therapy. Invest New Drugs. 30:1216–1223. 2012. View Article : Google Scholar : PubMed/NCBI

111 

Kenney C, Kunst T, Webb S, Christina D Jr, Arrowood C, Steinberg SM, Mettu NB, Kim EJ and Rudloff U: Phase II study of selumetinib, an orally active inhibitor of MEK1 and MEK2 kinases, in KRAS(G12R)-mutant pancreatic ductal adenocarcinoma. Invest New Drugs. 39:821–828. 2021. View Article : Google Scholar : PubMed/NCBI

112 

Rodon J, Tan DW, Laguna IG, Harb W, Beck JT, Bahary N, Rottey S, Zhu Z, Deng S, Kowalski K, et al: 344 Avelumab + binimetinib in metastatic pancreatic ductal adenocarcinoma (mPDAC): Dose-escalation results from the phase 1b/2 JAVELIN PARP MEKi trial. J Immunother Cancer. 9:A371. 2021. View Article : Google Scholar

113 

Surana R, Lee JJ, Smaglo BG, Zhao D, Lee MS, Wolff RA, Overman MJ, Willis J, Der CJ and Pant S: Phase I study of hydroxychloroquine plus binimetinib in patients with metastatic pancreatic cancer (the HOPE trial). J Clin Oncol. 40:TPS634. 2022. View Article : Google Scholar

114 

Grierson PM, Tan B, Pedersen KS, Park H, Suresh R, Amin MA, Trikalinos NA, Knoerzer D, Kreider B, Reddy A, et al: Phase Ib study of ulixertinib plus gemcitabine and nab-paclitaxel in patients with metastatic pancreatic adenocarcinoma. Oncologist. 28:e115–e123. 2023. View Article : Google Scholar : PubMed/NCBI

115 

Raybould AL, Burgess B, Urban C, Naim R, Lee MS and McRee AJ: A phase Ib trial of ERK inhibition with ulixertinib combined with palbociclib in patients (Pts) with advanced solid tumors. J Clin Oncol. 39:3103. 2021. View Article : Google Scholar

116 

Grilley-Olson JE, Bedard PL, Fasolo A, Cornfeld M, Cartee L, Razak ARA, Stayner LA, Wu Y, Greenwood R, Singh R, et al: A phase Ib dose-escalation study of the MEK inhibitor trametinib in combination with the PI3K/mTOR inhibitor GSK2126458 in patients with advanced solid tumors. Invest New Drugs. 34:740–749. 2016. View Article : Google Scholar : PubMed/NCBI

117 

Bardia A, Gounder M, Rodon J, Janku F, Lolkema MP, Stephenson JJ, Bedard PL, Schuler M, Sessa C, LoRusso P, et al: Phase Ib study of combination therapy with MEK inhibitor Binimetinib and Phosphatidylinositol 3-kinase inhibitor Buparlisib in patients with advanced solid tumors with RAS/RAF alterations. Oncologist. 25:e160–e169. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Ostrem JM and Shokat KM: Direct small-molecule inhibitors of KRAS: From structural insights to mechanism-based design. Nat Rev Drug Discov. 15:771–785. 2016. View Article : Google Scholar : PubMed/NCBI

119 

Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, Gaida K, Holt T, Knutson CG, Koppada N, et al: The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 575:217–223. 2019. View Article : Google Scholar : PubMed/NCBI

120 

Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, Sudhakar N, Bowcut V, Baer BR, Ballard JA, et al: The KRAS(G12C) inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patients. Cancer Discov. 10:54–71. 2020. View Article : Google Scholar : PubMed/NCBI

121 

Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, Italiano A, Schuler M, Borghaei H, Barlesi F, et al: Sotorasib for lung cancers with KRAS p.G12C mutation. N Engl J Med. 384:2371–2381. 2021. View Article : Google Scholar : PubMed/NCBI

122 

Khan S, Wiegand J, Zhang P, Hu W, Thummuri D, Budamagunta V, Hua N, Jin L, Allegra CJ, Kopetz SE, et al: BCL-X(L) PROTAC degrader DT2216 synergizes with sotorasib in preclinical models of KRAS(G12C)-mutated cancers. J Hematol Oncol. 15:232022. View Article : Google Scholar : PubMed/NCBI

123 

Strickler JH, Satake H, George TJ, Yaeger R, Hollebecque A, Garrido-Laguna I, Schuler M, Burns TF, Coveler AL, Falchook GS, et al: Sotorasib in KRAS p.G12C-mutated advanced pancreatic cancer. N Engl J Med. 388:33–43. 2023. View Article : Google Scholar : PubMed/NCBI

124 

Bekaii-Saab TS, Spira AI, Yaeger R, Buchschacher GL, McRee AJ, Sabari JK, Johnson ML, Barve NA, Hafez N, Velastegui K, et al: KRYSTAL-1: Updated activity and safety of adagrasib (MRTX849) in patients (Pts) with unresectable or metastatic pancreatic cancer (PDAC) and other gastrointestinal (GI) tumors harboring a KRASG12C mutation. J Clin Oncol. 40:519. 2022. View Article : Google Scholar

125 

Wang-Gillam A, Hubner RA, Siveke JT, Von Hoff DD, Belanger B, de Jong FA, Mirakhur B and Chen LT: NAPOLI-1 phase 3 study of liposomal irinotecan in metastatic pancreatic cancer: Final overall survival analysis and characteristics of long-term survivors. Eur J Cancer. 108:78–87. 2019. View Article : Google Scholar : PubMed/NCBI

126 

Wang X, Allen S, Blake JF, Bowcut V, Briere DM, Calinisan A, Dahlke JR, Fell JB, Fischer JP, Gunn RJ, et al: Identification of MRTX1133, a Noncovalent, potent, and selective KRAS(G12D) inhibitor. J Med Chem. 65:3123–3133. 2022. View Article : Google Scholar : PubMed/NCBI

127 

Hallin J, Bowcut V, Calinisan A, Briere DM, Hargis L, Engstrom LD, Laguer J, Medwid J, Vanderpool D, Lifset E, et al: Anti-tumor efficacy of a potent and selective non-covalent KRAS(G12D) inhibitor. Nat Med. 28:2171–2182. 2022. View Article : Google Scholar : PubMed/NCBI

128 

Kemp SB, Cheng N, Markosyan N, Sor R, Kim IK, Hallin J, Shoush J, Quinones L, Brown NV, Bassett JB, et al: Efficacy of a small molecule inhibitor of KrasG12D in immunocompetent models of pancreatic cancer. Cancer Discov. 13:298–311. 2022. View Article : Google Scholar : PubMed/NCBI

129 

Bannoura SF, Khan HY and Azmi AS: KRAS G12D targeted therapies for pancreatic cancer: Has the fortress been conquered? Front Oncol. 12:10139022022. View Article : Google Scholar : PubMed/NCBI

130 

Huang L, Guo Z, Wang F and Fu L: KRAS mutation: From undruggable to druggable in cancer. Signal Transduct Target Ther. 6:3862021. View Article : Google Scholar : PubMed/NCBI

131 

Gustafson WC, Wildes D, Rice MA, Lee BJ, Jiang J, Wang Z, Chang S, Flagella M, Mu Y, Dinglasan N, et al: Direct targeting of RAS in pancreatic ductal adenocarcinoma with RMC-6236, a first-in-class, RAS-selective, orally bioavailable, tri-complex RASMULTI(ON) inhibitor. J Clin Oncol. 40:591. 2022. View Article : Google Scholar

132 

Frank KJ, Mulero-Sánchez A, Berninger A, Ruiz-Cañas L, Bosma A, Görgülü K, Wu N, Diakopoulos KN, Kaya-Aksoy E, Ruess DA, et al: Extensive preclinical validation of combined RMC-4550 and LY3214996 supports clinical investigation for KRAS mutant pancreatic cancer. Cell Rep Med. 3:1008152022. View Article : Google Scholar : PubMed/NCBI

133 

Gort E, Johnson ML, Hwang JJ, Pant S, Dünzinger U, Riemann K, Kitzing T and Janne PA: A phase I, open-label, dose-escalation trial of BI 1701963 as monotherapy and in combination with trametinib in patients with KRAS mutated advanced or metastatic solid tumors. J Clin Oncol. 38:TPS3651. 2020. View Article : Google Scholar

Related Articles

Journal Cover

November-2023
Volume 50 Issue 5

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang J, Darman L, Hassan MS, Von Holzen U and Awasthi N: Targeting KRAS for the potential treatment of pancreatic ductal adenocarcinoma: Recent advancements provide hope (Review). Oncol Rep 50: 206, 2023
APA
Zhang, J., Darman, L., Hassan, M.S., Von Holzen, U., & Awasthi, N. (2023). Targeting KRAS for the potential treatment of pancreatic ductal adenocarcinoma: Recent advancements provide hope (Review). Oncology Reports, 50, 206. https://doi.org/10.3892/or.2023.8643
MLA
Zhang, J., Darman, L., Hassan, M. S., Von Holzen, U., Awasthi, N."Targeting KRAS for the potential treatment of pancreatic ductal adenocarcinoma: Recent advancements provide hope (Review)". Oncology Reports 50.5 (2023): 206.
Chicago
Zhang, J., Darman, L., Hassan, M. S., Von Holzen, U., Awasthi, N."Targeting KRAS for the potential treatment of pancreatic ductal adenocarcinoma: Recent advancements provide hope (Review)". Oncology Reports 50, no. 5 (2023): 206. https://doi.org/10.3892/or.2023.8643