Abstract
Pancreatic ductal adenocarcinoma (PDAC) is associated with significant morbidity and mortality and is projected to be the second leading cause of cancer-related deaths by 2030. Mutations in KRAS are found in the vast majority of PDAC cases and plays an important role in the development of the disease. KRAS drives tumor cell proliferation and survival through activating the MAPK pathway to drive cell cycle progression and to lead to MYC-driven cellular programs. Moreover, activated KRAS promotes a pro-tumorigenic microenvironment through forming a desmoplastic stroma and by impairing anti-tumor immunity. Secretion of GM-CSF and recruitment of myeloid-derived suppressor cells and pro-tumorigenic macrophages results in an immunosuppressive environment while secretion of SHH and TGF-beta drive fibroblastic features characteristic of PDAC. Recent development of several small molecules to directly target KRAS mark an important milestone in precision medicine. Many molecules show promise in preclinical models of PDAC and in early phase clinical trials. In this review, we discuss the underlying cell intrinsic and extrinsic roles of KRAS in PDAC tumorigenesis, the pharmacologic development of KRAS inhibition, and therapeutic strategies to target KRAS in PDAC.
Introduction:
The advance of targeted- and immuno-therapy has changed the therapeutic landscape across clinical oncology. However, these interventions have had limited clinical benefit in pancreatic ductal adenocarcinoma (PDAC), which portends a poor prognosis with a 5-year overall survival rate of 12% [1]. Advanced disease at the time of diagnosis and resistance to cytotoxic chemotherapies contribute to poor survival. Additionally, PDAC infrequently possesses druggable genomic alterations, such as mutations in BRCA, PALB2, or fusions with NTRK or NRG1. KRAS mutations, however, are implicated in over 90% of PDACs. Mutations in KRAS drive tumorigenesis while additionally altering the local inflammatory milieu. Though previously regarded as undruggable, the recent clinical breakthrough of active KRASG12C inhibitors has provided new promise against one of the most prevalent oncogenes. Although KRASG12C mutations are uncommon in PDAC, the new ability to target this oncogene is an enormous scientific feat that opens the door to further develop KRAS inhibitors relevant to PDAC. In this review, we discuss the impact of KRAS activation on tumorigenesis and the preclinical and translational efforts to target KRAS in PDAC.
KRAS activation in PDAC tumorigenesis
KRAS (Kirsten rat sarcoma viral oncogene homolog) is a small GTPase that cycles between a GDP-bound “off”-state and a GTP-bound “on”-state through interactions with GTPase-activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs), respectively. Activation of receptor tyrosine kinases (RTKs) promotes recruitment of Src homology, Collagen family adaptor protein (SHC) and growth factor receptor-bound protein 2 (GRB-2). This scaffold recruits the GEF, son of sevenless-1 (SOS1) to help activate KRAS [2]. To serve as a conduit for extracellular signals via receptor tyrosine kinases, KRAS needs to be anchored to the cellular membrane. This is achieved by farnesylation and carboxymethylation of the C185 residue to allow insertion into the lipid bilayer (Figure 1A). Moreover, the presence of a polybasic stretch of amino acids in the highly variable region preceding C185 allows for interaction with both acidic phospholipids and prenylation-binding proteins to ensure proper localization of KRAS to the plasma membrane [3–5]. Mutations frequently impact the G12, G13, and Q61 codons of KRAS, and 90% of mutations occur at G12 in PDAC [6]. These mutations localize to the catalytic GTPase site of KRAS, impairing GTP hydrolysis [7, 8]. Notably, oncogenic KRAS is not permanently locked in an “on”-state but, rather, is in a prolonged activated state that is influenced by growth factors and other modifiers, albeit to differing magnitudes [9, 10]. Moreover, different mutations within KRAS differentially impacts the structure of KRAS and influence its interactions with GAPs [11]. For an in-depth discussion on the impact of mutations and nucleotide binding on the structure of KRAS, the authors direct interested readers to the review by Tatu Pantsar [12].
Figure 1.

KRAS signaling and KRAS-directed therapy
A. Schematic outline of KRAS gene-structure, highlighting hotspot mutations and farnesylated cysteine. B. Protein-interaction network evaluating high-confidence interactions with KRAS based on experimentally determined, curated, and co-expression databases. C. Outline of cell-intrinsic factors governed by KRAS signaling. Therapies undergoing clinical evaluation are denoted. D. Outline of cell-extrinsic factors governed by KRAS signaling. HVR – high variable region. CAFs – cancer-associated fibroblasts. MDSC – Myeloid-derived suppressor cells. TCR – T-cell receptor. GAP – GTPase activating protein. Created with BioRender.com
Upon GTP-binding, the switch I and switch II domains of KRAS undergo conformational change to interact with downstream effectors [13]. Effector proteins are defined as proteins that contain a RAS-binding or RAS-association domain. Classical effector proteins include RAF, PI3K, and RALGDS, but there are many others (Figure 1B). Bioinformatic evaluation predicts over 50 different effectors and biochemical purification or proximity labeling implicates hundreds of effector-complexes [14–16]. These effectors potentiate downstream signaling cascades to alter cellular proliferation, survival, metabolism, and motility that ultimate drive pancreatic tumorigenesis [17, 18].
Of the multiple downstream effectors of RAS, expression of oncogenic BRAF to activate the RAF-MEK-ERK pathway in mice is sufficient to drive the development of PDAC, while expression of oncogenic PIK3CA to activate the PI3K-AKT-mTOR pathway results in a grossly normal mouse pancreas [19]. These findings highlight the importance of the RAF-MEK-ERK pathway, rather than the P13K-AKT-mTOR pathway, in the development of PDAC, a finding that is corroborated by the infrequent (~1%) mutational and copy number changes in the PI3K pathway found in human PDAC [6, 20].
Additionally, other mouse models of PDAC and KRAS have provided biologic insight into the development of PDAC. These in vivo models help to understand the underlying features of tumorigenesis given their similarities in their tumor microenvironment and ability to metastasize [21]. In the setting of PDAC, KRAS mutations are early oncogenic events that lead to the development of precancerous pancreatic intraepithelial neoplasia (“PanIN”) lesions [21–23]. Autopsy studies reveal that the prevalence of PanIN lesions may be as high as 70% in healthy persons, with lesions that arise as early as the second decade of life [24–26]. Molecular evaluation using laser micro-dissection of individual PanINs has identified the presence of KRAS mutations within these precursor lesions and recent studies suggest that hundreds of individual PanIN lesions can be present within a single person, all of which are likely derived from independent KRAS mutations [23, 27].
KRAS signaling results in phosphorylation and activation of ERK. ERK drives cellular growth and PDAC progression through increased MYC stability, ultimately enabling unregulated cellular proliferation. This is mechanistically accomplished by 1) activating MYC via phosphorylation and 2) stabilizing MYC by phosphorylation and deactivation of FBXW7, an E3-ligase that promotes proteasomal degradation of MYC [28, 29]. In preclinical models of PDAC, decreasing MYC stability via pharmacologic or molecular inhibition impairs tumor growth [30, 31]. Conversely, increases in MYC copy number is associated with metastatic disease in preclinical models [32]. Evaluation of metastatic PDAC lesions in patient samples correspondingly shows amplification in 30% of metastatic samples [33]. Although signaling flux impinges on ERK activation to help stabilize MYC, ERK provides negative feedback via phosphorylation of SOS1 to impair SOS1 interaction with EGFR [34, 35]. However, mutated KRAS is presumably less sensitive to this negative regulation, shifting the homeostatic signaling state to allow for continual signal propagation to enforce ongoing cellular growth.
KRAS and the PDAC Tumor Microenvironment
Mutated KRAS not only drives intrinsic cellular proliferation, but also acts to modulate the surrounding tumor microenvironment (TME) to help drive PDAC tumorigenesis. PDAC is characterized by an immunosuppressive milieu with a highly desmoplastic stroma [36]. These microenvironment changes occur as early as the PanIN stage. During the development of PanINs, pancreatic acinar cells harboring mutant-KRAS drive the expression of intercellular adhesion molecule-1 (ICAM-1) and granulocyte-macrophage colony-stimulating factor (GM-CSF) to recruit macrophages and myeloid-derived suppressor cells (MDSCs) to dysplastic lesions (Figure 1D). Extrinsic KRAS signaling leads to polarization of tumor-associated macrophages into the tumorigenic M2 phase, which drives tumorigenesis and correlates with poor prognosis [37, 38]. Together, these components function to impair anti-tumor immunity and promote immune evasion [39–41]. Infiltrating myeloid cells subsequently secrete TNF, promoting tumor cell secretion of chemokines such as CXCL1 and CXCL2 via the NF-kappa-B pathway [42]. These chemokines further potentiate the recruitment and reprogramming of MDSCs to exclude and dampen cytolytic CD8+ T-cell function, resulting in an immunologically cold tumor. Additionally, tumor cell production of CCL5 serves as a chemokine for homing suppressive CD4+ T-regulatory (Treg) cells to tumors [43]. Recently, an evaluation of human PDAC samples with single-cell mass cytometry corroborated this preclinical immunoregulatory circuit and demonstrated that this regulatory network is dependent on mutation of TP53 [44]. Disruption of this immunosuppressive circuit by impairing macrophage function (by neutralizing ICAM-1 antibodies or pharmacologic CSF1R inhibition), depleting CD4+ T-cells (by neutralizing antibodies), or compromising chemokine signaling (by genetic ablation of CXCL2, CXCL1 or CCL5 or pharmacologic inhibition of CCR2 or CCR5) results in reduced tumorigenesis and increased sensitization to chemotherapy in murine models, highlighting the crucial role of immune cells in initiating tumorigenesis and modulating therapeutic responses [41, 44–48]. Additionally, genetically removing KRAS in PDAC cell lines significantly impairs their ability to initiate tumors in immunocompetent mouse models more significantly than in immunocompromised mouse models, underlining the central importance of KRAS in immune evasion and tumor maintenance [49].
The dense desmoplastic stroma that is phenotypically intertwined with PDAC tumors is also driven by mutant-KRAS. Pancreatic stellate cells (PSCs) comprise 4% of all pancreatic cells but play an essential role in regulating the extracellular matrix (ECM) and fibrosis in the setting of inflammation. In their quiescent state, these cells maintain ECM homeostasis and are characterized as lipid rich cells that store vitamin A. When activated, PSCs lose their lipid and vitamin A contents and differentiate into myofibroblastic cancer-associated fibroblasts (myCAFs) or inflammatory cancer-associated fibroblasts (iCAFs). myCAFs express alpha-smooth muscle Actin (α-SMA) and remodel the tumor microenvironment while iCAFs secrete interleukin-6 (IL-6), leukemia inhibitory factor (LIF) and other cytokines to promote a local immunosuppressive environment [50–52]. myCAFs are typically found in the proximity of tumor cells, while iCAFs can be more scattered. KRAS-mutated tumor cells secrete sonic hedgehog (SHH), interleukin-1 (IL-1), and transforming growth factor-beta (TGF-β) to activate PSCs to drive subsequent desmoplasia and a fibroinflammatory stroma [22, 53, 54]. Additionally, antigen-presenting CAFs (apCAFs) were recently discovered through single-cell transcriptomic evaluation of murine and human PDAC. These cells were found to express MHCII without costimulatory molecules and are postulated to modulate CD4+ T-cell function, potentially through deactivating CD4+ T-cells [55]. Interrogation of myCAF function was explored through depleting SHH, which ultimately decreased desmoplasia and increased sensitivity to gemcitabine. However, tumors that lacked SHH or α-SMA+ myCAFs were characterized as more undifferentiated and aggressive, revealing the complex effects that desmoplasia can have on tumor biology and therapeutic intervention [56–58]. In addition to fibrosis and immunosuppression, activated PSCs and CAFs can reciprocate paracrine signaling to engage the incipient KRAS-mutated tumor cells. Secretion of IGF-1 and GAS6 from CAFs activates AKT-mediated signaling in tumor cells, while LIF from CAFs activate the STAT3 pathway [59, 60]. Ongoing research aimed at dissecting these cellular relationships in the context of CAF heterogeneity and plasticity may provide strategies to leverage for therapeutic benefit.
Altogether, KRAS drives both intrinsic and extrinsic features of tumorigenesis. This, in combination with the frequency of KRAS mutations, underscores why inhibiting KRAS will be transformative for clinical oncology, particularly for PDAC patients.
Direct KRAS inhibition in PDAC
The smooth surface of KRAS along with its high affinity for GTP/GDP has made targeting this oncogene very challenging, and indeed, RAS-family proteins were initially deemed to be “undruggable”. Given the initial challenges of direct RAS inhibition, indirect inhibitors provided an alternative approach to targeting the MAPK pathway. These initial methods included using farnesyltransferase inhibitors and MEK inhibitors. Conceptually, farnesyltransferase inhibitors were thought to be a viable strategy and proved to inhibit RAS localization. Unfortunately, the farnesyltransferase inhibitor, tipifarnib, showed no added clinical benefit in PDAC patients when compared to gemcitabine monotherapy [61]. One possible cause of the limited efficacy was thought to be from compensatory geranylgeranylation of KRAS in the setting of farnesyltransferase inhibition [62]. Subsequent evaluation with dual farnesyl/geranylgeranyl transferase inhibitors demonstrated promising preclinical activity [63]. Utilizing MEK inhibitors to impair downstream signaling was also thought to be a viable option. However, when trametinib was combined with chemotherapy in a randomized trial, no clinical benefit was observed compared to chemotherapy alone [64].
Targeting KRAS and MAPK in PDAC remained a challenge. However, in 2013, the Shokat lab discovered an allosteric binding pocket near the switch II region of KRAS [65]. This pocket was revealed through disulfide-tethering of the G12C residue of mutant-KRAS and led to the development of compounds that could covalently bind to the newly mutated cysteine residue while interacting with the switch II pocket to alter affinity for GTP/GDP [65]. This spurred investigations that identified improved molecules (ARS-853, ARS1620, MRTX849, AMG510, JDQ443) that could trap KRASG12C in the inactive state in vitro and in vivo [66–71]. ARS-853 was modified to create ARS1620 by addition of a quinazoline ring to interact with the Switch II pocket [68]. Doing so resulted in a 10-fold increase in potency (IC50 of 120 nM compared to 1,700 nM). AMG510 uses a similar quinazoline core as ARS1620, but an isopropyl substitution allowed for utilization of the H95/Y96/Q99 pocket for stronger ligand-protein interactions via van der Waal forces, resulting in a 10-fold increase in potency [70]. MRTX849 uses a tetrahydropyridopyrimidine backbone and modifications of this backbone enable hydrogen bonding with L16, H95, and G10 [69]. JDQ443 uses a pyrazole-backbone and was developed through a combination of structure-based design, optimization, and structure-activity relationship exploration. JDQ443 differentially interacts with the switch II pocket by forming hydrogen bonds with D69 and interacting with the hydrophobic pocket formed by V103, I100, M72, and Q99 [71]. MRTX849 (adagrasib) and AMG510 (sotorasib) have since shown clinical benefit in patients with KRASG12C-mutated non-small cell lung cancer (NSCLC) with objective response rates of 42.9% and 28.1% and median duration of responses of 8.5 and 8.6 months for adagrasib and sotorasib, respectively [72, 73]. Both inhibitors are now FDA approved for this disease.
In 2012, through NMR coupled fragment-based screening, another pocket near the switch I and switch II regions was discovered [74, 75]. BI-2852 was developed to bind within this pocket, leading to steric hindrance of the guanine-nucleotide exchange factor, SOS1 [76]. This method differs from the Shokat lab’s strategy, as this was not dependent on disulphide tethering of the G12C. This discovery led to the identification of another site near the switch II region, which, in turn, aided in the development of other inhibitors (BI-2483, BI-0474, BI-2865, BI-1823911) with in vivo activity against KRASG12C and pan-KRAS [77–79]. Of note, BI-2865 is a pan-KRAS inhibitor that binds with a greater affinity to the inactive conformation of KRAS regardless of the KRAS allele, but only leads to inhibition of downstream signaling in mutated KRAS [77]. Additionally, BI-18239111 is a KRASG12C-specific inhibitor that favors binding to the GDP-bound state and is currently in early clinical trials with the SOS1 inhibitor, BI-1701963, which shifts KRAS to the GDP-bound state (NCT04973163).
Although these therapeutic developments marked milestone in targeting KRAS, G12 is infrequently mutated to cysteine in PDAC (~1% of KRAS-mutated PDAC). However, a Phase I/II trial evaluating the efficacy of sotorasib as monotherapy in 38 patients with KRASG12C-mutated PDAC showed tumor shrinkage in 30/38 (79%) patients, though only 8 of those patients (21% of total participants) had a confirmed partial response [80]. The median progression-free survival (mPFS) in this study was 4 months and the median overall survival (mOS) was 6.9 months. Additionally, glecirasib (JAB-21822) was presented at ASCO GI and showed a confirmed ORR of 41.9% and mPFS of 7.0 months without reaching mOS in 31 PDAC patients [81]. Adverse effects were tolerable with both drugs, with the most frequent grade 3 treatment-related adverse effect being diarrhea with sotorasib (16%) and anemia (6%) with glecirasib. Thus, despite the low prevalence of KRASG12C in PDAC, G12C inhibitors can provide benefit to a subset of patients with PDAC.
From the structure of MRTX849, MRTX1133 was developed as a non-covalent inhibitor of KRASG12D. This was accomplished by synthesizing compounds that could interact with the negatively charged aspartate residue at position 12 [82]. Biochemical evaluation of MRTX1133 showed a 700-fold increase in selective binding of KRASG12D over KRASWT and demonstrated the ability of MRTX1133 to bind to the GTP-bound form of KRASG12D, thereby impairing downstream binding of effector proteins [83]. Testing MRTX1133 in immunocompetent mouse models of PDAC revealed that KRASG12D inhibition impacted cell-intrinsic and cell-extrinsic features of the tumor, mimicking genetic ablation of KRAS. Tumors cells exhibited decreased tumor cell proliferation and increased tumor cell death, while the TME showed increased myCAFs, endothelial cells, and T-cell infiltration [84, 85]. Given the efficacy in preclinical models, a phase I/II study is currently underway evaluating MRTX1133 in KRASG12D-mutated advanced solid tumors (NCT05737706). Given that KRASG12D is the most prevalent KRAS mutation in pancreatic adenocarcinoma, allele-specific targeting of KRASG12D has the potential to benefit many patients.
In addition to inhibitors targeting the allosteric pocket near the switch II region, tri-complex KRAS inhibitors are mechanistically distinct molecules with pre-clinical activity [86–88]. These inhibitors aim to target the active form of KRAS by forming a trimeric complex consisting of the active KRAS protein, the inhibitory molecule, and the abundant chaperone protein cyclophilin A (CYPA). Mechanistically, the inhibitor serves as a “molecular glue” by forming a binary complex with CYPA, which subsequently engages with the active form of KRAS to form the trimeric complex. Recent implementation of this strategy has resulted in the development of two molecules (RMC-6291 and RMC-4998) capable of covalently inhibiting KRASG12C to impair effector binding and induce tumor regression in cell-line and patient-derived xenograft (PDX) models bearing KRASG12C-mutated NSCLC or colorectal cancer [89]. RMC-6291 is currently in clinical trials for KRASG12C-mutated solid tumors (NCT05462717). Given that this strategy targets the active form of KRAS, the approach is being adapted to create allele-agnostic inhibitors. The KRASMulti (ON) inhibitor, RMC-6239, was shown to inhibit growth of cell-line and patient derived tumors bearing G12D, G12R, or G12V mutations. Moreover, recent clinical testing revealed activity in two patients, one with NSCLC and one with PDAC [88]. Another KRASMulti (ON) inhibitor, RMC-7977 was shown to impact a wide array of murine and human PDAC samples, harboring various KRAS alleles [86, 90]. Importantly, although RMC-7977 can inhibit KRASWT and thus impact normal tissue, it does not induce measurable cell death in normal tissues. Broadening direct inhibition of KRAS through allele-agnostic strategies will enable targeting of more tumors. Once allele-agnostic inhibitors reach further development, the adverse effects, if any, of targeting KRASWT in normal tissues will be of notable clinical interest.
Indirect KRAS inhibition in PDAC
Despite the limited clinical benefit seen with indirect inhibitors, circuitously targeting KRAS may still represent a viable strategy to augment treatment of KRAS-mutated tumors, particularly through combination therapy. The Src homology region 2 domain-containing phosphatase-2 (SHP2) contains two SH2 domains that recognize the phosphorylated tyrosine at position 32 within the switch I region of KRAS and can activate SOS1 to regulate KRAS-mediated signaling [91]. Phosphorylation of Y32 on KRAS impairs GTP cycling and RAS-effector binding [92]. SHP2 is important in KRAS-mediated tumorigenesis as genetic deletion of SHP2 in a KRAS mutated mouse model of PDAC decreases carcinogenesis [93]. Moreover, combination of SHP2 inhibition with concurrent KRAS inhibition demonstrates improved efficacy in preclinical models and showed TME remodeling [94]. RMC-4550 and INO155 are SHP2 inhibitors that have shown preclinical activity and are currently in early clinical trials (NCT04916236 and NCT03114319, respectively) [95, 96]. Moreover, combined inhibition of SHP2 and KRAS has been shown to overcome resistance from MET-amplification in the setting of KRASG12C inhibition [97].
In addition to SHP2, SOS1 has also garnered interest as a target for KRAS-addicted tumors. SOS1 activates KRAS by promoting nucleotide exchange and is also the target of negative feedback regulation by ERK-1/2-mediated phosphorylation [35]. Not surprisingly, SOS1 is required for tumor growth of implanted human pancreatic cell lines [98]. As such, the SOS1 inhibitor, BI-3406, was discovered through a high-throughput screen and was shown to work synergistically with MEK inhibitors to constrain KRAS-driven tumors, presumably by decreasing feedback reactivation in the setting of MEK inhibition [99]. Based on the structure of BI-3406, MRTX0902 was derived by screening molecules with a similar backbone to BI-3406, followed by subsequent modifications [100]. Moreover, MRTX0902 has shown the ability to limit tumor growth of a KRASG12C-mutated PDAC cell line (MIA PaCa-2) and appears to have synergistic activity with adagrasib. MRTX0209 is being studied in early clinical trials as monotherapy for tumors with activated MAPK pathways or in combination with adagrasib for KRASG12C-mutated tumors (NCT05578092). These SOS1 and SHP2 inhibitor studies suggest that utilizing a second inhibitor within the context of KRAS inhibition can help to potentiate therapeutic benefit and is reminiscent of combined BRAF/MEK inhibition used in melanoma [101].
Additionally, mitogenic signals drive cell cycle progression. However, enhanced oncogenic signaling can trigger cellular senescence through p16INK4A or p15INK4B encoded by CDKN2A or CDKN2B, respectively. Oncogene-induced senescence serves as a barrier to tumorigenesis [102]. However, mutation of CDKN2A impairs cell cycle arrest through accumulation of CDK4/6. The frequent co-mutation of KRAS and CDKN2A in PDAC suggest a strategy for combination of MAPK and CDK4/6 inhibition [6]. As such, preclinical models of MEK inhibition and CDK4/6 inhibition show synergistic effects, resulting in induced tumor cell senescence. These senescent cells secrete an array of cytokines to remodel the vascular microenvironment, leading to immune cell infiltration and drug influx into the tumor [103]. In organoid models, dual inhibition of ERK and CDK4/6 resulted in synergistic effects on apoptosis [104]. In a small case series of PDAC patients, 6 patients with metastatic PDAC were treated with combined MEK inhibition (Trametinib) and CDK4/6 inhibition (Palbociclib) [105]. Notably, this combination showed prolonged activity in two patients, who demonstrated clinical benefit for 9 and 17.5 months. Extrapolation of these data suggest that dual CDK4/6 and KRAS inhibition can lead to a more pronounced effect on tumor cells warrant further clinical investigation.
Immune-based strategies to target KRAS in PDAC
Lastly, immune-based therapies are being harnessed to target KRAS in PDAC. Mutant-KRAS-derived peptides are neoepitopes that can be presented on the surface of cells through allele-specific MHC molecules and recognized as tumor antigens by T cells via T-cell receptors (TCRs) [106]. Additionally, dendritic cells (DCs) infiltrate tumors and phagocytose tumor debris, allowing for processing of tumor antigens. DCs then deliver and cross-present tumor antigens to T lymphocytes in lymph nodes and other lymphoid organs to stimulate anti-tumor immunity based on the cytokine-environment [107]. These features initiate anti-tumor immunity but are eventually impaired by an immunosuppressive TME.
Multiple strategies are being employed to improve anti-tumor immunity against the KRAS antigen. Several vaccine-based therapies have shown some benefit in PDAC. A Phase I/II study in 2001, showed that intradermal injection of synthetic KRAS peptides could elicit immune responses in humans and is associated with prolonged survival in a small group of patients [108]. More recently, peptide-based immunization against mutant-KRAS have entered early phase trials and have shown KRAS-specific T-cells post vaccination in 87% of patients (NCT05726864) [109]. Other vaccination strategies, such as using listeria toxin to stimulate T-cell immunity, coupled with expression of KRASG12D, diminishes the progression of early PanINs to PDAC in murine models [110]. This strategy is currently being coupled with immunotherapy in the peri-operative setting of PDAC and colorectal cancer (NCT04117087) [111]. mRNA-based and mature dendritic cell vaccines against mutant-KRAS are in clinical trials (NCT03948763 and NCT03592888). The process of DC vaccination requires collection of mononuclear blood cells and ex-vivo maturation of DCs followed by exposure to mutant-KRAS peptides to produce KRAS-presenting DCs that are subsequently infused into the patient. In addition to using vaccines to stimulate the immune system, gene-therapy techniques to create tumor-targeting T-cells are being developed. A case report demonstrated that adoptive transfer of engineered autologous HLC-C*08:02-restricted TCR T-cells targeting KRASG12D was able to produce tumor regression in a patient with refractory metastatic pancreatic cancer [112]. As expected, there are early trials that are evaluating autologous TCR-engineered T cells for treating specific KRAS mutations in PDAC (NCT03745326, NCT03190941 and NCT06043713).
Future Challenges and Directions
Although KRAS inhibition shows clinical activity in patients, the absolute effect is modest. In CodeBreaK-200, sotorasib was compared to docetaxel in the second-line setting for NSCLC and demonstrated no OS benefit and a modest PFS benefit of 1.1 months [73]. Clinical benefit is curtailed by the development of secondary resistance, as with many targeted-therapy strategies for clinical oncology. Extrapolating from the clinical experience of KRASG12C inhibition in NSCLC and CRC, nearly half of acquired resistance occurs through a second-site mutation or amplification of KRAS, alternative activation of the MAPK or PI3K pathway, or through oncogenic gene-fusions [113, 114]. As such, preclinical studies are evaluating combined therapies that synergize with KRAS inhibition to overcome resistance mechanisms. These observations help drive the rationale for combination strategies in active clinical trials (Table 1). In addition to acquired resistance, 5 out of 38 patients with PDAC progressed at the time of first assessment in the Phase I/II study of sotorasib. Moreover, in Codebreak-200, nearly 15% of patients progressed on sotorasib after two months of therapy. These observations suggest that a small percentage of patients may have intrinsic resistance to KRAS inhibition or develop acquired resistance rapidly [73, 80]. Exploring these underlying mechanisms will help to elucidate better ways of improving therapy.
Table 1.
Active KRAS-directed therapy trials in Pancreas Cancer as of January 2024
| Therapy | Target | Molecular details | Clinical Trial Number | Setting | Phase | Status |
|---|---|---|---|---|---|---|
| BI-1823911 | KRASG12C | Covalently to C12 residue, locks into GDP-bound form IC50: ~10 nM (by viability) |
NCT04973163 | Metastatic; monotherapy or with BI 1701963 (SOS1 inhibitor) | I | Active, not recruiting |
| BPI-421286 | KRASG12C | Covalently to C12 residue IC50: ~2.9 nM (by pERK) |
NCT05315180 | Metastatic; monotherapy | I | Recruiting |
| HBI-2438 | KRASG12C | Undisclosed | NCT05485974 | Metastatic; monotherapy | I | Recruiting |
| JAB-21822 | KRASG12C | Covalently to C12 residue IC50: ~200 nM (by growth inhibition) |
NCT05288205 | Metastatic; given with JAB-3312 (SHP2 inhibitor) | I/II | Recruiting |
| LY3537982 | KRASG12C | Covalent inhibitor IC50: ~3.35 nM (by viability in NSCLC) |
NCT04956640 | Metastatic; monotherapy and multiple combination therapy | I | Recruiting |
| MRTX849 | KRASG12C | Covalently to G12C, affinity for GDP-bound form IC50: ~4.8 nM (by pERK) |
NCT05634525 | Metastatic; monotherapy | Ib | Recruiting |
| MRTX849 | KRASG12C | NCT03785249 | Metastatic; given with either pembrolizumab, Cetuximab, or afatinib | Ib | Recruiting | |
| RMC-6291 | KRASG12C | Inhibitory tri-complex with KRASG12C(ON) and CYPA IC50: ~0.11 nM |
NCT05462717 | Metastatic; monotherapy | I | Recruiting |
| RMC-6291 | KRASG12C | NCT06128551 | Metastatic; given with RMC-6236 (RAS-MULTI(ON) inhibitor) | I | Recruiting | |
| INCB161734 | KRASG12D | Binds both GDP/GTP forms of KRASG12D IC50: ~7 nM (by pERK) |
NCT06179160 | Metastatic; monotherapy or with cetuximab or retifanlimab | I | Recruiting |
| MRTX1133 | KRASG12D | Non-covalent to switch II binding pocket IC50: ~5 nM |
NCT05737706 | Metastatic; monotherapy | I/II | Recruiting |
| RMC-9805 | KRASG12D | Covalent Inhibitory tri-complex with KRASG12D(ON) and CYPA IC50: ~5nM (by viability) |
NCT06040541 | Metastatic; monotherapy | I/IB | Recruiting |
| RMC-6236 | RAS-MULTI(ON) | Inhibitory tri-complex with RAS(ON) and CYPA IC50: ~1–27 nM (by viability) |
NCT05379985 | Metastatic | I | Recruiting |
| YL-17231 | Pan-KRAS | Switch II pocket binding IC50: ~4–10 nM |
NCT06078800 | Metastatic; monotherapy | I | Recruiting |
| BMS-986466 | SHP2 | Allosteric inhibitor IC50: N.R |
NCT06024174 | Metastatic; given with adagrasib +/− cetuximab | I/II | Recruiting |
| HBI-2376 | SHP2 | Allosteric inhibitor IC50: N.R. |
NCT05163028 | Metastatic; monotherapy | I | Recruiting |
| JAB-3312 | SHP2 | Allosteric inhibitor IC50: ~3.62 nM (by pERK in NSCLC) |
NCT04121286 | Metastatic; monotherapy | I | Recruiting |
| RMC-4630 | SHP2 | Allosteric inhibitor IC50: ~32 nM (by proliferation in NSCLC) |
NCT04916236 | Metastatic; given with LY3214996 (ERK Inhibitor) | I | Recruiting |
| KO-2806 | Farnesyl transferase | Inhibitor IC50: ~1 – 10 nM (by viability in HNSCC) |
NCT06026410 | Metastatic; monotherapy | I | Recruiting |
| Vemurafenib | BRAF | BRAFV600E inhibitor IC50: ~31 nM |
NCT05068752 | Metastatic; given with Sorafenib | II | Recruiting |
| Avutometinib | RAF/MEK | Allosteric inhibitor that forms inactive complex with RAF + MEK IC50: ~160 nM (for MEK) |
NCT05669482 | Metastatic; given with defactinib (FAK inhibitor) and gemcitabine/nab-paclitaxel | I/II | Recruiting |
| Binimetinib | MEK | Reversible inhibitor IC50: ~12nM |
NCT04132505 | Metastatic; given with hydroxychloroquine | I | Recruiting |
| Binimetinib | MEK | NCT05554367 | Metastatic; given with Palbociclib | II | Recruiting | |
| Cobimetinib | MEK | Reversible inhibitor IC50: ~4.2nM |
NCT04214418 | Metastatic; given with Hydroxychloroquine and atezolizumab | I/II | Active, not recruiting |
| BMF-219 | Menin | Covalent inhibitor to disrupt Menin/Myc IC50: N.R. |
NCT05631574 | Metastatic; monotherapy | I | Recruiting |
| KRAS peptide Vaccine | Mutant KRAS | Mixture of 6 synthetic long Peptide | NCT04117087 | Adjuvant; given with ipilimumab and nivolumab | I | Recruiting |
| KRAS peptide Vaccine (TG01) | Mutant KRAS | Mixture of 7 synthetic peptide 17 a.a. long peptides | NCT06015724 | Metastatic; given with daratumumab and nivolumab | II | Recruiting |
| Protein Vaccine (KISIMA) | KRASG12D; KRASG12V |
Recombinant fusion protein consisting of a cell-penetrating peptide, an antigen domain, and a TLR agonist domain | NCT05846516 | Metastatic; given with ezabenlimab (PD-1 inhibitor) | I | Recruiting |
| KRAS Vaccine (ELI-002 2P) | KRASG12D KRASG12R | Lymph node targeting mKRAS-peptide and CpG DNA | NCT04853017 | Adjuvant, biomarker positive, CT scan negative | I | Active, not recruiting |
| KRAS Vaccine (ELI-002 7P) | KRASG12D, KRASG12R, KRASG12V, KRASG12A, KRASG12C, KRASG12S, KRASG13D | Lymph node targeting mKRAS-peptide and CpG DNA | NCT05726864 | Adjuvant, biomarker positive, CT scan negative | I | Recruiting |
| Dendritic Cell Vaccine | KRASG12D, KRASG12V, KRASG12R, KRASG12C |
Autologous, peptide-pulsed, dendritic cell infusion | NCT03592888 | Adjuvant; HLA-A02, HLA-A03, HLA-A11, HLA-B07 and HLA-C08. | I | Active, not recruiting |
| TCR | KRASG12V | Autologous, KRAS-specific-TCR transduced, T-cell infusion | NCT04146298 | Metastatic; HLA-A*11:01 | I/II | Recruiting |
| TCR (FH-A11KRASG12V-TCR) |
KRASG12V | Autologous, KRAS-specific-TCR transduced, T-cell infusion | NCT06043713 | Metastatic; HLA-A*11:01 | I | Recruiting |
| TCR | KRASG12V, KRASG12D |
Autologous, KRAS-specific-TCR transduced, T-cell infusion | NCT05438667 | Metastatic; HLA -A*11:01 | I | Recruiting |
| Murine TCR | KRASG12V | Murine TCR transduced, PBL infusion | NCT03190941 | Metastatic; HLA-A*11:01 | I/II | Recruiting |
| Murine TCR | KRASG12D | Murine TCR transduced, PBL infusion | NCT03745326 | Metastatic; HLA-A*11:01 | I/II | Recruiting |
TCR – T-cell Receptor. HLA – human leukocyte antigens. IC50 – half-maximal inhibitory concentration. NR – not reported.
In addition to overcoming resistance, better pre-clinical modeling of PDAC can help define important features that predict clinical response. One limitation to pre-clinical mouse models is their isogenic nature compared to the heterogeneity of tumors in PDAC patients. Patient-derived organoids (PDOs) maintain intratumoral heterogeneity similar to their original tumors [115]. A prior study using a wide library of PDOs exemplifies the ability to discover gene-expression signatures to predict response to chemotherapy [116]. Although traditional organoid culturing conditions do not sustain stroma, recent advances to organoid technology has enabled culturing PDOs on a microfluidic chip that can also support co-culture with stromal components, allowing for further dissection of TME and tumorigenesis and therapeutic sensitivity [117]. Thus, utilization of PDOs will be a critical tool for discovering biomarkers of therapeutic response and can serve as a platform to explore new treatment vulnerabilities in the context on KRAS inhibition.
Preclinical studies demonstrate that KRAS helps to drive an immunosuppressive environment and that inhibition of KRAS results in infiltration of T-cells within tumors. Depletion of CD4+ and CD8+ T-cells in the setting of KRAS inhibition results in more rapid relapses and greater tumor growth while the addition of anti-CTLA-4 and anti-PD-1 prolongs survival in mouse in models of PDAC [84, 85]. These two observations highlight the importance of T-cells to potentiate the effect of KRAS inhibition in PDAC and raises the question of whether patients will benefit from combined immune-checkpoint blockade and KRAS inhibition.
Furthermore, the intimate relationship between extrinsic KRAS signaling and macrophage infiltration and function may be leveraged. In the pre-clinical setting, macrophage-directed therapies have been shown to sensitize PDAC tumors to chemotherapy, immunotherapy, and radiation [118, 119]. Despite this, early clinical trials with macrophage-directed therapy (anti-CSF1R antibody) showed limited clinical activity [120]. Whether synergistic effects KRAS inhibition and macrophage-directed therapy exist remains to be explored.
As we look towards these future challenges, it is important to remember that the developmental of these inhibitors would not have been possible without understanding the biochemical structure of KRAS. As such, the structure of many direct inhibitors complexed with a variety of mutant-KRAS is publicly available in the Protein Data Bank (Table 2) and may serve as a resource for the optimizing future inhibitors.
Table 2.
Publicly available co-structures with KRAS and an inhibitory molecule
| PDB ID | KRAS mutant | Nucleotide | Inhibitory molecule |
|---|---|---|---|
| 8AZV | KRASWT | GDP | BI-2865 |
| 8TBF | KRASWT | GNP | RMC-7977 + CypA |
| 5F2E | KRASG12C | GDP | ARS-583 |
| 5V9U | KRASG12C | GDP | ARS-1620 |
| 6B0V | KRASG12C | GDP | ARS-107 |
| 6B0Y | KRASG12C | GDP | ARS-917 |
| 6OIM | KRASG12C | GDP | AMG 510 |
| 6USX | KRASG12C | GDP | MRTX849 |
| 5YXZ | KRASG12C | GDP | JBI484 |
| 5YY1 | KRASG12C | GDP | JBI739 |
| 7R0M | KRASG12C | GDP | JDQ443 |
| 8AZX | KRASG12C | GDP | BI-2865 |
| 8G9P | KRASG12C | GNP | RMC-4998 + CypA |
| 8TBK | KRASG12C | GNP | RMC-7977 + CypA |
| 8JGD | KRASG12C | GDP | YK-8S |
| 8DNI | KRASG12C | GDP | Araxes WO2020/028706A1 compound I-1 |
| 7ACA | KRASG12D | GPPCP | BI-5747 |
| 7T47 | KRASG12D | GPPCP | MRTX1133 |
| 8TBL | KRASG12D | GNP | RMC-7977 + CypA |
| 8TBJ | KRASG12R | GNP | RMC-7977 + CypA |
| 8TBN | KRASG12S | GNP | RMC-7977 + CypA |
| 8AZZ | KRASG12V | GDP | BI-2865 |
| 8TBM | KRASG12V | GNP | RMC-7977 + CypA |
| 8B00 | KRASG13D | GDP | BI-2865 |
| 8ONV | KRASG13D | GDP | BI-2493 |
CypA – Cyclophilin A. GDP – guanosine diphosphate. GNP – phosphoaminophosphonic acid-guanylate ester. GPPCP – Guanosine-5′-[(β,γ)-methyleno]triphosphate.
Conclusion
Pancreatic adenocarcinoma continues to be a fatal disease and its rising incidence is a concerning public health issue. However, decades of research have enabled greater understanding of the molecular underpinnings of this disease and have revolutionized our understanding of the role of mutant-KRAS in tumorigenesis and in the tumor microenvironment. This foundational knowledge is being transformed into therapeutic advancements that have the capacity to change clinical outcomes for patients with PDAC. Although the prognosis of PDAC remains dismal, KRAS-directed therapies represent an approach that may alter the treatment landscape of this disease.
Acknowledgements
MTT is supported by T32 CA009615.
References:
- 1.Siegel RL, et al. , Cancer statistics, 2022. CA Cancer J Clin, 2022. 72(1): p. 7–33. [DOI] [PubMed] [Google Scholar]
- 2.Sasaoka T, et al. , The signaling pathway coupling epidermal growth factor receptors to activation of p21ras. J Biol Chem, 1994. 269(51): p. 32621–5. [PubMed] [Google Scholar]
- 3.Roy MO, Leventis R, and Silvius JR, Mutational and biochemical analysis of plasma membrane targeting mediated by the farnesylated, polybasic carboxy terminus of K-ras4B. Biochemistry, 2000. 39(28): p. 8298–307. [DOI] [PubMed] [Google Scholar]
- 4.Dharmaiah S, et al. , Structural basis of recognition of farnesylated and methylated KRAS4b by PDEδ. Proc Natl Acad Sci U S A, 2016. 113(44): p. E6766–e6775. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Schmick M, et al. , KRas localizes to the plasma membrane by spatial cycles of solubilization, trapping and vesicular transport. Cell, 2014. 157(2): p. 459–471. [DOI] [PubMed] [Google Scholar]
- 6.Waddell N, et al. , Whole genomes redefine the mutational landscape of pancreatic cancer. Nature, 2015. 518(7540): p. 495–501. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Scheffzek K, et al. , The Ras-RasGAP complex: structural basis for GTPase activation and its loss in oncogenic Ras mutants. Science, 1997. 277(5324): p. 333–8. [DOI] [PubMed] [Google Scholar]
- 8.Hunter JC, et al. , Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol Cancer Res, 2015. 13(9): p. 1325–35. [DOI] [PubMed] [Google Scholar]
- 9.Huang H, et al. , Oncogenic K-Ras requires activation for enhanced activity. Oncogene, 2014. 33(4): p. 532–535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Daniluk J, et al. , An NF-κB pathway-mediated positive feedback loop amplifies Ras activity to pathological levels in mice. J Clin Invest, 2012. 122(4): p. 1519–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Lu S, et al. , The Structural Basis of Oncogenic Mutations G12, G13 and Q61 in Small GTPase K-Ras4B. Scientific Reports, 2016. 6(1): p. 21949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Pantsar T, The current understanding of KRAS protein structure and dynamics. Comput Struct Biotechnol J, 2020. 18: p. 189–198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Mott HR and Owen D, Structures of Ras superfamily effector complexes: What have we learnt in two decades? Crit Rev Biochem Mol Biol, 2015. 50(2): p. 85–133. [DOI] [PubMed] [Google Scholar]
- 14.Rezaei Adariani S, et al. , A comprehensive analysis of RAS-effector interactions reveals interaction hotspots and new binding partners. Journal of Biological Chemistry, 2021. 296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Zhang X, et al. , Comparative Nucleotide-Dependent Interactome Analysis Reveals Shared and Differential Properties of KRas4a and KRas4b. ACS Cent Sci, 2018. 4(1): p. 71–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Kovalski JR, et al. , The Functional Proximal Proteome of Oncogenic Ras Includes mTORC2. Mol Cell, 2019. 73(4): p. 830–844.e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kiel C, Matallanas D, and Kolch W, The Ins and Outs of RAS Effector Complexes. Biomolecules, 2021. 11(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ibáňez Gaspar V, et al. , Analysis of Ras-effector interaction competition in large intestine and colorectal cancer context. Small GTPases, 2021. 12(3): p. 209–225. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Collisson EA, et al. , A central role for RAF→MEK→ERK signaling in the genesis of pancreatic ductal adenocarcinoma. Cancer Discov, 2012. 2(8): p. 685–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Biankin AV, et al. , Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature, 2012. 491(7424): p. 399–405. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hingorani SR, et al. , Preinvasive and invasive ductal pancreatic cancer and its early detection in the mouse. Cancer Cell, 2003. 4(6): p. 437–450. [DOI] [PubMed] [Google Scholar]
- 22.Collins MA, et al. , Oncogenic Kras is required for both the initiation and maintenance of pancreatic cancer in mice. The Journal of Clinical Investigation, 2012. 122(2): p. 639–653. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Kanda M, et al. , Presence of somatic mutations in most early-stage pancreatic intraepithelial neoplasia. Gastroenterology, 2012. 142(4): p. 730–733.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Matsuda Y, et al. , The Prevalence and Clinicopathological Characteristics of High-Grade Pancreatic Intraepithelial Neoplasia: Autopsy Study Evaluating the Entire Pancreatic Parenchyma. Pancreas, 2017. 46(5): p. 658–664. [DOI] [PubMed] [Google Scholar]
- 25.Andea A, Sarkar F, and Adsay VN, Clinicopathological Correlates of Pancreatic Intraepithelial Neoplasia: A Comparative Analysis of 82 Cases With and 152 Cases Without Pancreatic Ductal Adenocarcinoma. Modern Pathology, 2003. 16(10): p. 996–1006. [DOI] [PubMed] [Google Scholar]
- 26.Carpenter ES, et al. , Analysis of Donor Pancreata Defines the Transcriptomic Signature and Microenvironment of Early Neoplastic Lesions. Cancer Discov, 2023. 13(6): p. 1324–1345. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Braxton AM, et al. , Three-dimensional genomic mapping of human pancreatic tissue reveals striking multifocality and genetic heterogeneity in precancerous lesions. bioRxiv, 2023. [Google Scholar]
- 28.Ji S, et al. , ERK kinase phosphorylates and destabilizes the tumor suppressor FBW7 in pancreatic cancer. Cell Research, 2015. 25(5): p. 561–573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Vaseva AV, et al. , KRAS Suppression-Induced Degradation of MYC Is Antagonized by a MEK5-ERK5 Compensatory Mechanism. Cancer Cell, 2018. 34(5): p. 807–822.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Blake DR, et al. , Application of a MYC degradation screen identifies sensitivity to CDK9 inhibitors in KRAS-mutant pancreatic cancer. Science Signaling, 2019. 12(590): p. eaav7259. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Saborowski M, et al. , A modular and flexible ESC-based mouse model of pancreatic cancer. Genes Dev, 2014. 28(1): p. 85–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Maddipati R, et al. , MYC Levels Regulate Metastatic Heterogeneity in Pancreatic Adenocarcinoma. Cancer Discov, 2022. 12(2): p. 542–561. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Schleger C, et al. , c-MYC activation in primary and metastatic ductal adenocarcinoma of the pancreas: incidence, mechanisms, and clinical significance. Mod Pathol, 2002. 15(4): p. 462–9. [DOI] [PubMed] [Google Scholar]
- 34.Rozakis-Adcock M, et al. , MAP kinase phosphorylation of mSos1 promotes dissociation of mSos1-Shc and mSos1-EGF receptor complexes. Oncogene, 1995. 11(7): p. 1417–26. [PubMed] [Google Scholar]
- 35.Corbalan-Garcia S, et al. , Identification of the mitogen-activated protein kinase phosphorylation sites on human Sos1 that regulate interaction with Grb2. Mol Cell Biol, 1996. 16(10): p. 5674–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Whatcott CJ, et al. , Desmoplasia in Primary Tumors and Metastatic Lesions of Pancreatic Cancer. Clin Cancer Res, 2015. 21(15): p. 3561–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Velez-Delgado A, et al. , Extrinsic KRAS Signaling Shapes the Pancreatic Microenvironment Through Fibroblast Reprogramming. Cell Mol Gastroenterol Hepatol, 2022. 13(6): p. 1673–1699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Knudsen ES, et al. , Stratification of Pancreatic Ductal Adenocarcinoma: Combinatorial Genetic, Stromal, and Immunologic Markers. Clin Cancer Res, 2017. 23(15): p. 4429–4440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Bayne LJ, et al. , Tumor-derived granulocyte-macrophage colony-stimulating factor regulates myeloid inflammation and T cell immunity in pancreatic cancer. Cancer Cell, 2012. 21(6): p. 822–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Pylayeva-Gupta Y, et al. , Oncogenic Kras-induced GM-CSF production promotes the development of pancreatic neoplasia. Cancer Cell, 2012. 21(6): p. 836–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Liou GY, et al. , Mutant KRAS-induced expression of ICAM-1 in pancreatic acinar cells causes attraction of macrophages to expedite the formation of precancerous lesions. Cancer Discov, 2015. 5(1): p. 52–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Burke SJ, et al. , NF-κB and STAT1 control CXCL1 and CXCL2 gene transcription. Am J Physiol Endocrinol Metab, 2014. 306(2): p. E131–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Tan MC, et al. , Disruption of CCR5-dependent homing of regulatory T cells inhibits tumor growth in a murine model of pancreatic cancer. J Immunol, 2009. 182(3): p. 1746–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Bianchi A, et al. , Cell-Autonomous Cxcl1 Sustains Tolerogenic Circuitries and Stromal Inflammation via Neutrophil-Derived TNF in Pancreatic Cancer. Cancer Discov, 2023. 13(6): p. 1428–1453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Nywening TM, et al. , Targeting both tumour-associated CXCR2(+) neutrophils and CCR2(+) macrophages disrupts myeloid recruitment and improves chemotherapeutic responses in pancreatic ductal adenocarcinoma. Gut, 2018. 67(6): p. 1112–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mitchem JB, et al. , Targeting tumor-infiltrating macrophages decreases tumor-initiating cells, relieves immunosuppression, and improves chemotherapeutic responses. Cancer Res, 2013. 73(3): p. 1128–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Zhang Y, et al. , CD4+ T lymphocyte ablation prevents pancreatic carcinogenesis in mice. Cancer Immunol Res, 2014. 2(5): p. 423–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Candido JB, et al. , CSF1R(+) Macrophages Sustain Pancreatic Tumor Growth through T Cell Suppression and Maintenance of Key Gene Programs that Define the Squamous Subtype. Cell Rep, 2018. 23(5): p. 1448–1460. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Ischenko I, et al. , KRAS drives immune evasion in a genetic model of pancreatic cancer. Nature Communications, 2021. 12(1): p. 1482. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Zhou Y, et al. , Pancreatic Stellate Cells: A Rising Translational Physiology Star as a Potential Stem Cell Type for Beta Cell Neogenesis. Front Physiol, 2019. 10: p. 218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Öhlund D, et al. , Distinct populations of inflammatory fibroblasts and myofibroblasts in pancreatic cancer. J Exp Med, 2017. 214(3): p. 579–596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Garg B, et al. , NFκB in Pancreatic Stellate Cells Reduces Infiltration of Tumors by Cytotoxic T Cells and Killing of Cancer Cells, via Up-regulation of CXCL12. Gastroenterology, 2018. 155(3): p. 880–891.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Bailey JM, et al. , Sonic hedgehog promotes desmoplasia in pancreatic cancer. Clin Cancer Res, 2008. 14(19): p. 5995–6004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Ling J, et al. , KrasG12D-induced IKK2/β/NF-κB activation by IL-1α and p62 feedforward loops is required for development of pancreatic ductal adenocarcinoma. Cancer Cell, 2012. 21(1): p. 105–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Elyada E, et al. , Cross-Species Single-Cell Analysis of Pancreatic Ductal Adenocarcinoma Reveals Antigen-Presenting Cancer-Associated Fibroblasts. Cancer Discov, 2019. 9(8): p. 1102–1123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Rhim AD, et al. , Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell, 2014. 25(6): p. 735–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Olive KP, et al. , Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science, 2009. 324(5933): p. 1457–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Özdemir BC, et al. , Depletion of carcinoma-associated fibroblasts and fibrosis induces immunosuppression and accelerates pancreas cancer with reduced survival. Cancer Cell, 2014. 25(6): p. 719–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Tape CJ, et al. , Oncogenic KRAS Regulates Tumor Cell Signaling via Stromal Reciprocation. Cell, 2016. 165(4): p. 910–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Shi Y, et al. , Targeting LIF-mediated paracrine interaction for pancreatic cancer therapy and monitoring. Nature, 2019. 569(7754): p. 131–135. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Van Cutsem E, et al. , Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol, 2004. 22(8): p. 1430–8. [DOI] [PubMed] [Google Scholar]
- 62.Whyte DB, et al. , K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem, 1997. 272(22): p. 14459–64. [DOI] [PubMed] [Google Scholar]
- 63.Kazi A, et al. , Dual Farnesyl and Geranylgeranyl Transferase Inhibitor Thwarts Mutant KRAS-Driven Patient-Derived Pancreatic Tumors. Clinical Cancer Research, 2019. 25(19): p. 5984–5996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Infante JR, et al. , 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. European Journal of Cancer, 2014. 50(12): p. 2072–2081. [DOI] [PubMed] [Google Scholar]
- 65.Ostrem JM, et al. , K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature, 2013. 503(7477): p. 548–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Lito P, et al. , Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science, 2016. 351(6273): p. 604–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Patricelli MP, et al. , Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. Cancer Discov, 2016. 6(3): p. 316–29. [DOI] [PubMed] [Google Scholar]
- 68.Janes MR, et al. , Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell, 2018. 172(3): p. 578–589.e17. [DOI] [PubMed] [Google Scholar]
- 69.Fell JB, et al. , Identification of the Clinical Development Candidate MRTX849, a Covalent KRASG12C Inhibitor for the Treatment of Cancer. Journal of Medicinal Chemistry, 2020. 63(13): p. 6679–6693. [DOI] [PubMed] [Google Scholar]
- 70.Lanman BA, et al. , Discovery of a Covalent Inhibitor of KRASG12C (AMG 510) for the Treatment of Solid Tumors. Journal of Medicinal Chemistry, 2020. 63(1): p. 52–65. [DOI] [PubMed] [Google Scholar]
- 71.Weiss A, et al. , Discovery, Preclinical Characterization, and Early Clinical Activity of JDQ443, a Structurally Novel, Potent, and Selective Covalent Oral Inhibitor of KRASG12C. Cancer Discov, 2022. 12(6): p. 1500–1517. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Jänne PA, et al. , Adagrasib in Non-Small-Cell Lung Cancer Harboring a KRAS(G12C) Mutation. N Engl J Med, 2022. 387(2): p. 120–131. [DOI] [PubMed] [Google Scholar]
- 73.de Langen AJ, et al. , Sotorasib versus docetaxel for previously treated non-small-cell lung cancer with KRAS(G12C) mutation: a randomised, open-label, phase 3 trial. Lancet, 2023. 401(10378): p. 733–746. [DOI] [PubMed] [Google Scholar]
- 74.Maurer T, et al. , Small-molecule ligands bind to a distinct pocket in Ras and inhibit SOS-mediated nucleotide exchange activity. Proc Natl Acad Sci U S A, 2012. 109(14): p. 5299–304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Sun Q, et al. , Discovery of Small Molecules that Bind to K-Ras and Inhibit Sos-Mediated Activation. Angewandte Chemie International Edition, 2012. 51(25): p. 6140–6143. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kessler D, et al. , Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci U S A, 2019. 116(32): p. 15823–15829. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Kim D, et al. , Pan-KRAS inhibitor disables oncogenic signalling and tumour growth. Nature, 2023. 619(7968): p. 160–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Bröker J, et al. , Fragment Optimization of Reversible Binding to the Switch II Pocket on KRAS Leads to a Potent, In Vivo Active KRAS(G12C) Inhibitor. J Med Chem, 2022. 65(21): p. 14614–14629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Savarese F, et al. , Abstract 1271: In vitro and in vivo characterization of BI 1823911 - a novel KRASG12C selective small molecule inhibitor. Cancer Research, 2021. 81(13_Supplement): p. 1271–1271. [Google Scholar]
- 80.Strickler JH, et al. , Sotorasib in KRAS p.G12C-Mutated Advanced Pancreatic Cancer. N Engl J Med, 2023. 388(1): p. 33–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Li J, et al. , Preliminary activity and safety results of KRAS G12C inhibitor glecirasib (JAB-21822) in patients with pancreatic cancer and other solid tumors. Journal of Clinical Oncology, 2024. 42(3_suppl): p. 604–604. [Google Scholar]
- 82.Wang X, et al. , Identification of MRTX1133, a Noncovalent, Potent, and Selective KRASG12D Inhibitor. Journal of Medicinal Chemistry, 2022. 65(4): p. 3123–3133. [DOI] [PubMed] [Google Scholar]
- 83.Hallin J, et al. , Anti-tumor efficacy of a potent and selective non-covalent KRASG12D inhibitor. Nature Medicine, 2022. 28(10): p. 2171–2182. [DOI] [PubMed] [Google Scholar]
- 84.Kemp SB, et al. , Efficacy of a Small-Molecule Inhibitor of KrasG12D in Immunocompetent Models of Pancreatic Cancer. Cancer Discov, 2023. 13(2): p. 298–311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Mahadevan KK, et al. , KRAS(G12D) inhibition reprograms the microenvironment of early and advanced pancreatic cancer to promote FAS-mediated killing by CD8(+) T cells. Cancer Cell, 2023. 41(9): p. 1606–1620.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Wasko UN, et al. , Tumor-selective activity of RAS-GTP inhibition in pancreatic cancer. Nature, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Holderfield M, et al. , Concurrent inhibition of oncogenic and wild-type RAS-GTP for cancer therapy. Nature, 2024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jiang J, et al. , Translational and Therapeutic Evaluation of RAS-GTP Inhibition by RMC-6236 in RAS-Driven Cancers. Cancer Discovery, 2024: p. OF1–OF24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Schulze CJ, et al. , Chemical remodeling of a cellular chaperone to target the active state of mutant KRAS. Science, 2023. 381(6659): p. 794–799. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Koltun ES, et al. , Abstract 3597: Direct targeting of KRASG12X mutant cancers with RMC-6236, a first-in-class, RAS-selective, orally bioavailable, tri-complex RASMULTI(ON) inhibitor. Cancer Research, 2022. 82(12_Supplement): p. 3597–3597. [Google Scholar]
- 91.Bunda S, et al. , Inhibition of SHP2-mediated dephosphorylation of Ras suppresses oncogenesis. Nature Communications, 2015. 6(1): p. 8859. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Kano Y, et al. , Tyrosyl phosphorylation of KRAS stalls GTPase cycle via alteration of switch I and II conformation. Nat Commun, 2019. 10(1): p. 224. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Ruess DA, et al. , Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nature Medicine, 2018. 24(7): p. 954–960. [DOI] [PubMed] [Google Scholar]
- 94.Fedele C, et al. , SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med, 2021. 218(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Nichols RJ, et al. , RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nature Cell Biology, 2018. 20(9): p. 1064–1073. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.LaMarche MJ, et al. , Identification of TNO155, an Allosteric SHP2 Inhibitor for the Treatment of Cancer. Journal of Medicinal Chemistry, 2020. 63(22): p. 13578–13594. [DOI] [PubMed] [Google Scholar]
- 97.Suzuki S, et al. , KRAS Inhibitor Resistance in MET-Amplified KRAS (G12C) Non-Small Cell Lung Cancer Induced By RAS- and Non-RAS-Mediated Cell Signaling Mechanisms. Clin Cancer Res, 2021. 27(20): p. 5697–5707. [DOI] [PubMed] [Google Scholar]
- 98.Jeng HH, Taylor LJ, and Bar-Sagi D, Sos-mediated cross-activation of wild-type Ras by oncogenic Ras is essential for tumorigenesis. Nat Commun, 2012. 3: p. 1168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Hofmann MH, et al. , BI-3406, a Potent and Selective SOS1–KRAS Interaction Inhibitor, Is Effective in KRAS-Driven Cancers through Combined MEK Inhibition. Cancer Discovery, 2021. 11(1): p. 142–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Ketcham JM, et al. , Design and Discovery of MRTX0902, a Potent, Selective, Brain-Penetrant, and Orally Bioavailable Inhibitor of the SOS1:KRAS Protein–Protein Interaction. Journal of Medicinal Chemistry, 2022. 65(14): p. 9678–9690. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Long GV, et al. , Combined BRAF and MEK Inhibition versus BRAF Inhibition Alone in Melanoma. New England Journal of Medicine, 2014. 371(20): p. 1877–1888. [DOI] [PubMed] [Google Scholar]
- 102.Bartkova J, et al. , Oncogene-induced senescence is part of the tumorigenesis barrier imposed by DNA damage checkpoints. Nature, 2006. 444(7119): p. 633–637. [DOI] [PubMed] [Google Scholar]
- 103.Ruscetti M, et al. , Senescence-Induced Vascular Remodeling Creates Therapeutic Vulnerabilities in Pancreas Cancer. Cell, 2020. 181(2): p. 424–441.e21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Goodwin CM, et al. , Combination Therapies with CDK4/6 Inhibitors to Treat KRAS-Mutant Pancreatic Cancer. Cancer Res, 2023. 83(1): p. 141–157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kato S, et al. , Concomitant MEK and Cyclin Gene Alterations: Implications for Response to Targeted Therapeutics. Clinical Cancer Research, 2021. 27(10): p. 2792–2797. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Abrams SI, et al. , Identification of overlapping epitopes in mutant ras oncogene peptides that activate CD4+ and CD8+ T cell responses. Eur J Immunol, 1996. 26(2): p. 435–43. [DOI] [PubMed] [Google Scholar]
- 107.Xiao Z, et al. , Impaired function of dendritic cells within the tumor microenvironment. Front Immunol, 2023. 14: p. 1213629. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Gjertsen MK, et al. , Intradermal ras peptide vaccination with granulocyte-macrophage colony-stimulating factor as adjuvant: Clinical and immunological responses in patients with pancreatic adenocarcinoma. Int J Cancer, 2001. 92(3): p. 441–50. [DOI] [PubMed] [Google Scholar]
- 109.Perry JR, et al. , 656 ELI-002 immunotherapy induces broad polyfunctional T cell responses in subjects with high relapse risk KRAS mutated pancreatic ductal adenocarcinoma and colorectal cancer. Journal for ImmunoTherapy of Cancer, 2023. 11(Suppl 1): p. A748–A748. [Google Scholar]
- 110.Keenan BP, et al. , A Listeria vaccine and depletion of T-regulatory cells activate immunity against early stage pancreatic intraepithelial neoplasms and prolong survival of mice. Gastroenterology, 2014. 146(7): p. 1784–94.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Zaidi N, et al. , Abstract IA013: Intercepting pancreatic cancer development with mutant KRAS-targeted immunotherapy. Cancer Research, 2022. 82(22_Supplement): p. IA013–IA013. [Google Scholar]
- 112.Leidner R, et al. , Neoantigen T-Cell Receptor Gene Therapy in Pancreatic Cancer. New England Journal of Medicine, 2022. 386(22): p. 2112–2119. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Awad MM, et al. , Acquired Resistance to KRASG12C Inhibition in Cancer. New England Journal of Medicine, 2021. 384(25): p. 2382–2393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Zhao Y, et al. , Diverse alterations associated with resistance to KRAS(G12C) inhibition. Nature, 2021. 599(7886): p. 679–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Boj SF, et al. , Organoid models of human and mouse ductal pancreatic cancer. Cell, 2015. 160(1–2): p. 324–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Tiriac H, et al. , Organoid Profiling Identifies Common Responders to Chemotherapy in Pancreatic Cancer. Cancer Discov, 2018. 8(9): p. 1112–1129. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Haque MR, et al. , Patient-derived pancreatic cancer-on-a-chip recapitulates the tumor microenvironment. Microsystems & Nanoengineering, 2022. 8(1): p. 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Zhu Y, et al. , CSF1/CSF1R blockade reprograms tumor-infiltrating macrophages and improves response to T-cell checkpoint immunotherapy in pancreatic cancer models. Cancer Res, 2014. 74(18): p. 5057–69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Jones KI, et al. , Radiation combined with macrophage depletion promotes adaptive immunity and potentiates checkpoint blockade. EMBO Molecular Medicine, 2018. 10(12): p. e9342. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Papadopoulos KP, et al. , First-in-Human Study of AMG 820, a Monoclonal Anti-Colony-Stimulating Factor 1 Receptor Antibody, in Patients with Advanced Solid Tumors. Clinical Cancer Research, 2017. 23(19): p. 5703–5710. [DOI] [PubMed] [Google Scholar]
