Skip to main content
Erschienen in: Molecular Cancer 1/2021

Open Access 01.12.2021 | Review

Oncogenic KRAS blockade therapy: renewed enthusiasm and persistent challenges

verfasst von: Daolin Tang, Guido Kroemer, Rui Kang

Erschienen in: Molecular Cancer | Ausgabe 1/2021

Abstract

Across a broad range of human cancers, gain-of-function mutations in RAS genes (HRAS, NRAS, and KRAS) lead to constitutive activity of oncoproteins responsible for tumorigenesis and cancer progression. The targeting of RAS with drugs is challenging because RAS lacks classic and tractable drug binding sites. Over the past 30 years, this perception has led to the pursuit of indirect routes for targeting RAS expression, processing, upstream regulators, or downstream effectors. After the discovery that the KRAS-G12C variant contains a druggable pocket below the switch-II loop region, it has become possible to design irreversible covalent inhibitors for the variant with improved potency, selectivity and bioavailability. Two such inhibitors, sotorasib (AMG 510) and adagrasib (MRTX849), were recently evaluated in phase I-III trials for the treatment of non-small cell lung cancer with KRAS-G12C mutations, heralding a new era of precision oncology. In this review, we outline the mutations and functions of KRAS in human tumors and then analyze indirect and direct approaches to shut down the oncogenic KRAS network. Specifically, we discuss the mechanistic principles, clinical features, and strategies for overcoming primary or secondary resistance to KRAS-G12C blockade.
Hinweise

Publisher’s Note

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

Key points

  • KRAS is the most frequently mutated oncogene in human cancer and has challenged the development of clinical anticancer therapeutics in the last 30 years.
  • Mutated KRAS oncoprotein disrupts GAP-mediated GTP hydrolysis and thus remains in a continuous GTP binding activation state.
  • Small-molecule inhibitors that directly target KRAS-G12C mutants provide new tools for precision oncology.
  • Clinical trials involving covalent KRAS-G12C inhibitors (adagrasib and sotorasib) have shown promising activity against lung cancers harboring KRAS-G12C mutations.
  • Secondary KRAS mutations, gain-of-function mutations of the MAPK pathway, loss-of-function mutations in tumor suppressor genes, and other gene alterations are conducive to acquired resistance to KRAS-G12C inhibitors.
  • The design and implementation of strategies to minimize or overcome drug resistance is an important goal for the further development of KRAS inhibitors.

Introduction

The RAS gene was initially identified as a virus-encoded gene in 1964 [1], and later was found to be a genome-encoded oncogene that is frequently mutated in human cancers [2]. Thus, activating mutations of RAS are found in 19% of neoplasias, corresponding to approximately 3.4 million new diagnoses of malignant disease worldwide each year [2]. The RAS gene family includes HRAS, NRAS, and KRAS, which are encoding proteins that play partially overlapping but specific roles in signaling transduction [3]. For example, the knockout of Kras is embryonically lethal in mice, while the depletion of Nras or Hras does not affect development [4]. The mutation of human KRAS was first detected in non-small cell lung cancer (NSCLC) [57]. The earliest evidence that RAS is an oncogene was based on the fact that transfecting mouse Kras causes morphological transformation of NIH-3T3 fibroblasts [8]. Subsequent studies involving transgenic mice confirmed that the mutation of Hras, Nras, or Kras mimicked human oncogenesis by triggering the stochastic transformation of cells [912]. Clinical studies revealed the prognostic impact of RAS mutations in certain cancers [2]. However, in spite of decades-long efforts of academia and industry to target RAS protein for cancer therapy [13], the design of direct RAS pharmacological inhibitors has only achieved a major breakthrough during recent years [14]. In 2021, two covalent inhibitors of KRAS-G12C protein (hereafter referred to as G12Ci), sotorasib (AMG 510) [15] and adagrasib (MRTX849) [16], were clinically approved by the U.S. Food and Drug Administration and the European Medicines Agency to treat patients with advanced NSCLC carrying the KRAS-G12C mutation. Monotherapies or combination therapies using G12Ci are being evaluated in clinical trials for advanced or metastatic solid cancer, including NSCLC, colorectal cancer (CRC), and pancreatic ductal adenocarcinoma (PDAC) [17]. This progress is inspiring scientists to continue to design drugs targeting key oncogenic drivers, even those that previously were considered to be “undruggable” like KRAS [18].
Here, we summarize recent therapeutic advances in mutant KRAS blockade that have led to clinical approval or are currently being evaluated in trials (Table 1), while discussing attempts to target upstream regulators, downstream effectors, and mutant KRAS protein itself. We also discuss the challenges associated with G12Ci-based treatments and the future prospects of this evolving topic.
Table 1
Clinical trials targeting KRAS
Target
Agent
Combinations
Study
phase
Tumor type
Recruitment status
Trial number
BRAF, CRAF
LXH-254
None
I
Advanced solid tumors harboring MAPK pathway alterations
Active, not recruiting
NCT02607813
BRAF, CRAF
LXH-254
Rineterkib (RAF/ERK inhibitor); trametinib (MEK inhibitor); ribociclib (CDK4/6 inhibitor); EGF816 (EGFR inhibitor); dabrafenib (BRAF mutant inhibitor)
I, II
Unresectable or metastatic melanoma; EGFR-mutant NSCLC
Recruiting; active, not recruiting
NCT04417621; NCT02974725; NCT03333343; NCT04294160
ERK
LY3214996; GDC-0994; ulixertinib; MK-8353
None
I, II
Acute myeloid leukemia; locally advanced or metastatic solid tumors; metastatic uveal melanoma; acute myelogenous leukemia or myelodysplastic syndromes
Recruiting; completed; terminated
NCT04081259; NCT01875705; NCT04488003; NCT03417739; NCT02296242; NCT01358331
ERK
LY3214996
RMC-4630 (SHP2-inhibitor); abemaciclib (CDK4/6 inhibitor); hydroxychloroquine (autophagy inhibitor)
I, II
Metastatic KRAS mutant cancers; solid tumors harboring pathogenic alterations in BRAF, RAF1, MEK1/2, ERK1/2, and NF1
Not yet recruiting
NCT04916236; NCT04956640; NCT04534283; NCT04616183; NCT04391595; NCT04386057
ERK
GDC-0994
Cobimetinib (MEK inhibitor)
I
Locally advanced or metastatic solid tumors
Completed
NCT02457793
ERK
Ulixertinib
Hydroxychloroquine (autophagy inhibitor); palbociclib (CDK4/6 inhibitor)
I, II
Advanced MAPK-mutated gastrointestinal adenocarcinomas; advanced pancreatic and other solid tumors
Recruiting
NCT041452973; NCT03454035
ERK
MK-8353
Selumetinib (MEK inhibitor); pembrolizumab (anti–PD-1 ab)
I
Advanced malignancies
Completed; active, not recruiting
NCT03745989; NCT02972034
KRAS
AZD4785
None
I
Advanced solid tumors
Completed
NCT03101839
KRAS-G12C
Sotorasib
None
I, II
KRAS-G12C–mutant advanced/metastatic solid tumors
Recruiting; not yet recruiting
NCT04380753; NCT04625647; NCT04667234; NCT04933695
KRAS-G12C
Sotorasib
Docetaxel (microtubule inhibitor); pembrolizumab (anti–PD-1 ab)
I, II, III
KRAS-G12C–mutant advanced/metastatic solid tumors
Active, not recruiting; recruiting
NCT04303780; NCT03600883; NCT04613596
KRAS-G12C
Adagrasib
Docetaxel (microtubule inhibitor); pembrolizumab (anti–PD-1 ab); cetuximab (anti-EGFR ab); afatinib (pan-EGFR inhibitor); TNO155 (SHP2 inhibitor)
I, II, III
KRAS-G12C–mutant advanced/metastatic solid tumors
Recruiting
NCT04685135; NCT03785249; NCT04330664; NCT04793958
KRAS-G12C
GDC-6036
Atezolizumab (anti–PD-L1 ab); cetuximab (anti-EGFR ab); bevacizumab (anti-VEGF ab); erlotinib (EGFR inhibitor)
I
KRAS-G12C–mutant advanced/metastatic solid tumors
Recruiting
NCT04449874
KRAS-G12C
D-1553
Standard treatment
I
KRAS-G12C–mutant advanced/metastatic solid tumors
Recruiting
NCT04585035
KRAS-G12C
JNJ-74699157
Standard treatment
I
KRAS-G12C–mutant advanced/metastatic solid tumors
Completed
NCT04006301
KRAS-G12C
LY3499446
Abemaciclib (CDK4/6 inhibitor); cetuximab (anti-EGFR ab); erlotinib (EGFR inhibitor); docetaxel (microtubule inhibitor)
I, II
KRAS-G12C–mutant advanced/metastatic solid tumors
Terminated
NCT04165031
KRAS-G12D
siG12D-LODER
Gemcitabine + nab-paclitaxel; FOLFIRINOX
II
Advanced pancreatic cancer
Recruiting
NCT01676259
MEK
Cobimetinib
Belvarafenib
(RAF inhibitor)
I
Advanced or metastatic solid tumors
Recruiting
NCT03284502
MEK
Trametinib
LXH254 (RAF inhibitor)
I
NSCLC or melanoma
Recruiting
NCT02974725
MEK
Pimasertib
None
I
N-RAS–mutated locally advanced or metastasis malignant cutaneous melanoma
Recruiting
NCT01693068, NCT00982865
MEK
Pimasertib
SAR405838 (MDM2 antagonist)
I
Advanced solid tumors
Completed
NCT01985191
MEK
Mirdametinib
Lifirfenib
I
Advanced or refractory solid tumors
Recruiting
NCT03905148
p110α
Alpelisib
Capecitabine (nucleoside metabolic inhibitor); paclitaxel (microtubule inhibitor)
I
Patients with PIK3CA mutant metastatic colorectal cancer; PIK3CA-altered metastatic/recurrent gastric cancer
Not yet recruiting
NCT04753203; NCT04526470
p110α
GDC-0077
Entrectinib (pan-TRK inhibitor)
I
Breast cancer and advanced solid tumors
Recruiting
NCT04632992
RAF
PLX8394; TAK-580
None
I, II
Advanced unresectable solid tumors; low-grade glioma
Recruiting
NCT02428712; NCT03429803
RAF
Belvarafenib
None
I
Solid tumors
Completed
NCT02405065
RAF
Belvarafenib
Cobimetinib (MEK inhibitor); cetuximab (anti-EGFR ab); atezolizumab (anti–PD-L1 ab)
I
Advanced or metastatic solid tumors; NRAS-mutant advanced melanoma
Recruiting
NCT03284502; NCT04835805
RAF, EGFR
Lifirfenib
None
I
Solid tumors
Completed
NCT02610361; NCT03641586
RAF, EGFR
Lifirfenib
Mirdametinib (MEK inhibitor)
I
NSCLC with confirmed KRAS mutations
Recruiting
NCT04294160
SHP2
BBP-398; JAB-3068; RMC-4630; RLY-1971; JAB-3312; SH3809
None
I, II
Advanced or metastatic solid tumors
Recruiting
NCT04528836; NCT03565003; NCT03518554; NCT03634982; NCT04252339; NCT04121286; NCT04045496; NCT04843033
SHP2
RMC-4630
LY3214996 (ERK inhibitor); cobimetinib (MEK inhibitor); osimertinib (EGFR inhibitor)
I, II
Advanced or metastatic solid tumors
Not yet recruiting; recruiting
NCT04916236; NCT03989115
SHP2
ERAS-601
Cobimetinib (MEK inhibitor)
I
Advanced or metastatic solid tumors
Recruiting
NCT04670679
SHP2
TNO155
Nazartinib (EGFR inhibitor); spartalizumab (anti–IL-1β antibody); ribociclib (CDK4/6 inhibitor); adagrasib (KRAS-G12C inhibitor); JDQ443 (KRAS-G12C inhibitor)
I, II
Advanced solid tumors
Recruiting
NCT03114319; NCT04000529; NCT04330664; NCT04699188
SOS1
BI 1701963
Trametinib (MEK inhibitor); BI 3011441 (MEK inhibitor); irinotecan (topoisomerase I inhibitor)
I
Advanced or metastatic solid tumors
Recruiting
NCT04111458; NCT04835714; NCT04627142

Type and frequency of KRAS mutation

RAS genes are mutated at different prevalence rates in human cancers (Fig. 1a) [19]. Elucidating similarities and differences among these RAS mutations from a developmental or evolutionary perspective remains a challenge [20, 21]. Some RAS gene mutations are innocuous, but others cause cancer by producing oncoproteins. For example, three amino acid residues (G12, G13, and Q61) in HRAS, KRAS, and NRAS are mutational hot spots, though with distinct frequencies in different human tumor types (Fig. 1b) [14]. A KRAS-G12 mutation is a common event in pancreatic (91%), colorectal (68%), and lung adenocarcinoma (85%; a subtype of NSCLC) [14]. Among these, KRAS-G12D is the leading mutation in pancreatic (45%) and colorectal adenocarcinoma (45%), while KRAS-G12C mainly occurs in lung adenocarcinoma (46%) (Fig. 1c) [14]. In contrast, 85% of human melanomas have KRAS-Q61 mutations, especially Q61R (46%) [14]. Whole-exome sequencing of human PDAC or NSCLC tumors shows that, despite the genetic heterogeneity within the tumor, KRAS is mutated in different regions [22, 23]. Consistent with this, the co-mutational interactions between each KRAS allele and other unrelated genes are highly tissue-specific, emphasizing the complexity of cell type-specific oncogenesis [24]. KRAS mutations usually co-occur with mutations in tumor protein p53 (TP53) and cyclin-dependent kinase inhibitor 2A (CDKN2A) in PDAC, Kelch-like ECH-associated protein 1 (KEAP1) and/or serine/threonine kinase 11 (STK11) in NSCLC, or APC regulator of WNT signaling pathway (APC) and phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) in CRC. While mutations in exon 2 of KRAS are the most common, it is important to note that they affect the differential use of exon 4, giving rise to two splice variants, KRAS4A and KRAS4B (Fig. 1b) [25]. These KRAS variants differ in their C-terminal membrane targeting region, posttranslational modifications, and interactomes, thus exhibiting different signal behaviors in development, metabolism, and proliferation [2628]. Therefore, to target KRAS, investigators must consider changes in the protein structure caused by point mutations, but also isoform-specific properties [29].

Prognostic and predictive value of KRAS mutations

Depending on the clinical settings, the prognostic and predictive values of KRAS mutations are variable and even conflicting. For example, an early study of lung adenocarcinoma patients showed that the KRAS mutation in code 12 is an unfavorable prognostic factor [30]. Similarly, a meta-analysis found that KRAS mutations are associated with poor survival in patients with early resectable NSCLC [31]. In contrast, a pooled analysis of NSCLC patients treated with cisplatin-based chemotherapy revealed that a KRAS mutation was not a prognostic factor [32]. Of note, a KRAS mutation is a negative predictor of response to epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs; e.g., cetuximab, gefitinib, or erlotinib) (Table 2) [3336], but a positive predictor of response to immune checkpoint inhibitors (ICIs; including anti–PD-1 and anti–PD-L1) (Table 3) [3739]. However, response rates are variable and dissenting reports suggest that KRAS mutations cannot guide the therapeutic choice between TKIs and ICIs in NSCLC patients [40, 41].
Table 2
Tyrosine kinase inhibitors
Tyrosine kinase inhibitors (TKIs) are a group of drugs that disrupt the tyrosine kinase (TK) signal transduction pathway through a variety of mechanisms. They can compete with adenosine triphosphate (ATP), phosphorylated entities, substrates, or can act in an allosteric manner, that is, bind to sites outside the active site and affect the sites’ activity through conformational changes. TKs can be divided into receptor tyrosine kinases (RTKs), nonreceptor tyrosine kinases (NRTKs), and dual-specific kinases (DSKs). DSKs phosphorylate serine, threonine, and tyrosine residues. Approximately 20 different transmembrane RTK subfamilies have been identified, such the families for vascular endothelial growth factor receptor (VEGFR), platelet-derived growth factor receptor (PDGFR), insulin receptor (INSR), fibroblast growth factor receptor (FGFR), and epidermal growth factor receptor (EGFR). NRTKs are cytoplasmic proteins and do not have a transmembrane domain. NRTKs are mainly composed of nine families, including those for Abl, Ack, Csk, Fak, Fes/Fer, Jak, Src, Syk/Zap70, and Tec. The most typical example of DSK is mitogen-activated protein kinase kinase (MEK), which is involved in the mitogen-activated protein kinase (MAPK) pathway. More than 50 FDA-approved TKIs (including small-molecule inhibitors and monoclonal antibodies) are used to treat various diseases, including cancer.
Table 3
Immune checkpoint inhibitors
Immune checkpoint inhibitors (ICIs) are a group of drugs that inhibit the activity and function of inhibitory immune checkpoint molecules, such as programmed cell death protein 1 (PD-1), programmed death ligand 1 (PD-L1), cytotoxic T lymphocyte-associated protein 4 (CTLA-4), lymphocyte activation gene 3 (LAG3), and T-cell immunoglobulin and mucin domain-containing protein 3 (TIM3). Under physiological conditions, inhibitory immune checkpoint molecules play an important role in maintaining self-tolerance, preventing autoimmune reactions, and minimizing tissue damage by regulating the duration and intensity of immune responses. However, abnormal expression and excessive activation of immune checkpoint molecules can cause many diseases, including cancer. In particular, inhibitory immune checkpoint molecules are upregulated in various cells within the tumor microenvironment, forming various pairings and limiting the normal antitumor function of immune cells. In contrast, the use of ICIs can restore the function of immune cells hijacked by cancer cells, resulting in an enhanced immunosurveillance with a cytotoxic T lymphocyte (CTL) response. ICIs (e.g., pembrolizumab, nivolumab, cemiplimab, and atezolizumab) have changed the landscape of cancer treatment and become a new hope for cancer patients after the failure of regular chemotherapy or radiotherapy.
In PDAC, patients with KRAS-G12D mutations (but not total KRAS mutations) have a worse prognosis than patients with wild-type KRAS [4244]. Other studies suggest that the prognosis of KRAS-G12V is poorer than that for other mutations [45, 46], commensurate to a KRAS-G12V–associated increase in circulating regulatory T cells that most likely limits antitumor immunity [47]. In advanced PDAC, KRAS mutation status is predictive for the efficacy of erlotinib rather than prognostic [48]. This contradicts other studies reporting that KRAS wild-type patients with PDAC have a significant advantage after treatment with gemcitabine/nimotuzumab [49] or gemcitabine/erlotinib [50] with respect to overall survival. Circulating tumor DNA (ctDNA) has recently become a minimally invasive tool used in precision oncology to evaluate genetic alterations. Mutant KRAS in ctDNA might be a more sensitive predictor of survival than the ELISA-based detection of cancer antigen 19-9 (CA 19-9) [51, 52]. Before a clear conclusion can be drawn regarding the impact of KRAS mutations on overall survival in PDAC [53, 54], additional data from analyses of other genetic alterations are needed.
In localized CRC, KRAS mutations usually suggest a poor prognosis [5557]. Some KRAS mutations, including KRAS-G12V, are more aggressive than others [58]. In contrast to BRAF mutations, KRAS mutations have no major prognostic value in advanced CRC patients [59]. The association between KRAS mutations and poor clinical outcomes from TKI treatment has been confirmed in several independent studies [6063]. Compared with KRAS-G12V mutations or wild-type tumor groups, CRC patients with KRAS-G13D mutations are insensitive to cetuximab therapy [64, 65]. Nevertheless, the impact of KRAS mutations on cetuximab treatment needs to be further evaluated in prospective randomized trials. Reportedly, KRAS-G12D–mediated inhibition of interferon regulatory factor 2 (IRF2) drives CRC resistance to anti–PD-1 therapy in preclinical models [66]. However, the clinical implications of these findings remain elusive.
In conclusion, the prognostic and predictive values of KRAS mutations are affected by many factors, such as tumor type, stage, patient age, sex, the coexistence of mutations affecting other oncogenes or tumor suppressors, and treatment regimens. For this reason, the clinical utility of detecting KRAS mutations may be over- or underestimated. Careful meta-analyses that avoid nonrandom systematic errors due to variations between trials [67] are needed to clarify this issue.

Activation and modulation of KRAS mutations

Wild-type KRAS protein mostly resides on the cytoplasmic side of the plasma membrane, as well as at membranes of intracellular organelles, and is guided by protein localization signals (such as lipid moieties added to the carboxyl terminus) [68]. The RAS family of proteins belongs to a class of enzymes called small GTPases, which play a central role in cell signal transduction [69]. The functions and activities of RAS protein depend on the transition from an inactive guanosine diphosphate (GDP)-bound state to an active guanosine triphosphate (GTP)-bound state (Fig. 2a) [69]. Normally, this conversion process of RAS status is reversible and is maintained in a balanced manner by GTPase activating proteins (GAPs) and guanine nucleotide exchange factors (GEFs). GAPs, such as neurofibromin 1 (NF1), accelerate the GTP hydrolysis of RAS, leading RAS to an inactive state [70, 71]. GEFs, including the most universally expressed SOS Ras/Rac guanine nucleotide exchange factor 1 (SOS1), are responsible for producing active GTP-bound RAS [7274]. The balance between hydrolysis and exchange determines the level of activated KRAS in the cell [75]. Oncogenic mutations of RAS disrupt GAP-mediated GTP hydrolysis, allowing these oncoproteins to accumulate in a continuous GTP-binding active state (Fig. 2b) [13]. Among distinct KRAS mutants, the KRAS-G12C protein exhibits the highest intrinsic GTP hydrolysis rate [76].
The upstream regulators of the RAS pathway involve receptor tyrosine kinases (RTKs), which are cell surface receptors for many growth factors, cytokines, and hormones (Fig. 3). One particular RTK subclass, the EGFR family, is composed of four closely related members, EGFR (also known as ERBB1 or HER1), ERBB2/HER2, ERBB3/HER3, and ERBB4/HER4. EGFR/ERBB1, the best characterized activator of RAS signaling, acts through binding to an adaptor protein, namely growth factor receptor bound protein 2 (GRB2) [77, 78]. GRB2 further mediates the recruitment and activation of SOS1- and SH2-containing protein tyrosine phosphatase 2 (SHP2) to activate GTP-bound RAS [7984]. Activated EGFR mutations are often found in human cancers, especially NSCLC, and lead to the constitutive activation of downstream signals, including RAS [85]. G-protein–coupled receptors (GPCRs), the largest group of membrane receptors, also participate in RAS activation [86, 87]. Thus, the interplay between RTKs and GPCRs may increase the plasticity of RAS activation.
The downstream effectors of the RAS pathway are mainly involved in the activation of the mitogen-activated protein kinase (MAPK) and phosphatidylinositol 3-kinase (PI3K) pathways, which favor anabolic processes including cell growth, protein translation, and proliferation (Fig. 3) [88, 89]. Thus, the constitutive activation of membrane RAS-dependent signal pathways favors oncogenesis [90]. The aggregation of mitochondrial KRAS-G12V protein also favors tumor cell growth through metabolic effects [91]. Another distinctive feature of KRAS-G12D protein is that it can be released by PDAC cells into the tumor microenvironment and then mediates the polarization of pro-tumor macrophages (Fig. 4) [92]. Indeed, PDAC patients with KRAS-G12D–positive macrophages exhibit low survival rates [92]. There is also strong preclinical evidence that mutated KRAS requires additional factor (such as chronic inflammation or a high-fat or high-iron diet) to deploy its full carcinogenic activity [9395]. Further work is needed to elucidate the likely complex cell-autonomous and non-autonomous effects of mutated and unmutated KRAS protein on the tumor microenvironment [21].

Indirect KRAS suppression strategies

The most successful way to inhibit oncogenic kinases is to develop inhibitors that compete with ATP to bind to the kinase domain. However, KRAS uses GTP instead of ATP as a phosphate donor for signal transmission. Attempts to directly enzymatically inhibit KRAS function have been largely frustrated, leading to the development of indirect methods for KRAS inhibition. Below, we highlight some representative drugs that exemplify the main strategies for indirectly targeting mutant KRAS (Table 1 and Fig. 3).

Inhibition of KRAS expression

AZD4785 is a constrained ethyl-containing antisense oligonucleotide that is complementary to the sequence of the 3'UTR of KRAS mRNA, leading to the downregulation of wild-type and mutant KRAS protein [96]. AZD4785 selectively inhibits the proliferation of mutant KRAS-driven tumor cells in vitro or in xenograft models [96, 97]. However, intravenously infused AZD4785 failed to completely reduce KRAS mRNA in patients with NSCLC (NCT03101839) [14], calling for adjustments of the dose and method of administration. Another approach for transcriptionally inhibiting KRAS expression involves the use of a specific small interfering RNA (siRNA) named siG12D-LODER that specifically targets G12D but not wild-type KRAS. This agent showed promise in a phase I study (NCT01188785) in combination with chemotherapy (gemcitabine or FOLFIRINOX, i.e., a combination of 5-fluorouracil, leucovorin, irinotecan, and oxaliplatin) in 12 patients with advanced PDAC [98]. A phase II study (NCT01676259) evaluating this therapeutic strategy is now underway.

Inhibition of KRAS processing

The location of RAS on the cell membrane is the initial step of RAS activation and requires multiple posttranslational processing steps, especially lipid-related prenylation [99]. The two enzymes involved in KRAS prenylation are farnesyltransferase (FT) and geranylgeranyltransferase 1 (GGT1). Although preclinical studies suggest anticancer activity for FT inhibitors (for example, tipifarnib/R115777 and lonafarnib/SCH 66336) against RAS-mutant tumors, clinical studies have been disappointing [100103]. One possible explanation for this failure is functional redundancy among FT and GGT1 [104]. A single molecule with dual inhibitory activity on FT and GGT1, such as FGTI-2734 [105], might have the potential to eliminate RAS-mutant tumors. Alternatively, targeting downstream RAS processing enzymes, such as isoprenylcysteine carboxyl methyltransferase (ICMT), may be attempted. Two ICMT inhibitors (cysmethynil and UCM-1336) impair the membrane localization of RAS (including that of KRAS) [106], but their application in vivo remains to be studied. Once RAS is effectively processed, membrane RAS protein undergoes activating self-association, and this process can be blocked by a synthetic binding protein called NS1 [107]. Since NS1 is an alien protein, its possible recognition by the immune system needs to be evaluated before it is introduced into clinical trials.

Inhibition of upstream signaling molecules

Gefitinib, erlotinib, afatinib (pan-EGFR inhibitors), icotinib, and osimertinib are first-line EGFR TKIs for treating NSCLC patients with EGFR mutations [108]. Lapatinib is the first dual inhibitor of EGFR and ERBB2/HER2 for treating ERBB2-positive breast cancer, whereas brigatinib is a mixed inhibitor of ALK and EGFR used for the treatment of metastatic NSCLC. The clinical benefit of small-molecule EGFR inhibitors on KRAS-mutant cancers is context-dependent. For example, gefitinib alone is not effective against KRAS-mutant NSCLC [109], while the combination of erlotinib and gemcitabine provides transient benefit to patients with KRAS-mutant PDAC [110]. Another approach to inhibit EGFR activity consists of the use of monoclonal antibodies [108]. Cetuximab and panitumumab are approved for metastatic CRC, while necitumumab is used for the treatment of squamous NSCLC. However, some studies suggest that antibodies against EGFR have no effect on KRAS-mutant CRC [60, 62], while others report that CRC patients with KRAS-G13D are sensitive to cetuximab treatment [65]. Regardless, acquired KRAS mutations are a common mechanism of resistance to EGFR inhibitors [111]. Recently, rybrevant, a bispecific antibody against EGFR and MET receptors, has been approved for the treatment of NSCLC patients with EGFR exon 20 insertion mutations. Other EGFR-specific monoclonal antibodies in clinical development are zalutumumab, nimotuzumab, and matuzumab. It will be important to understand how preexisting or acquired KRAS mutations will affect the clinical activity of such drugs.
The proteins SOS1 and SOS2 promote RAS activation by binding to GRB2. The SOS1 inhibitor BI-1701963 is used in combination with MEK inhibitors (trametinib and BI-3011441) or a topoisomerase I inhibitor (irinotecan) in clinical trials enrolling patients with advanced or metastatic solid cancer (Table 1). BI-1701963 has been designed to bind to the catalytic domain of SOS1 to prevent its interaction with KRAS-GDP [79]. SOS1 mutations lead to dysregulated enzymatic activities, which may cause drug resistance [112]. Because there is currently no SOS2-specific inhibitor, it is unclear whether targeting SOS2 would have the same effects as SOS1 inhibitors. It can be speculated that pan-SOS inhibitors might be particularly efficient in blocking the activation of RAS.
SHP2 not only mediates the RTK-stimulated activation of RAS [113] but also acts as a promoter of immune checkpoint pathways [114]. Certain SHP2 inhibitors are in early-phase clinical development for treating advanced or metastatic solid cancer (Table 1). TNO155 is an allosteric inhibitor that maintains SHP2 in a self-inhibited conformation [115]. Preclinical studies have shown promising anticancer activity from TNO155 combined with inhibitors of EGFR, MEK, ERK, CDK4/6, or KRAS-G12C and anti–PD-1 antibodies in xenograft models of NSCLC or CRC cells [116]. The efficacy and toxicity of combination regimens involving TNO155 together with TKIs or ICIs remain to be determined in clinical trials.

Inhibition of downstream signaling molecules

Oncogenic transformation mediated by RAS requires the downstream activation of the RAF/MEK/ERK and the PI3K/AKT/mTOR pathways. In theory, inhibition of any of these effectors should block oncogenic KRAS signaling. In fact, these two pathways intersect with each other and even form a feedforward loop to activate KRAS as an upstream signal [14]. Nonetheless, the success of targeting KRAS-mutant tumors by inhibiting single downstream molecules has been limited [18]. Despite these results, certain MAPK and PI3K pathway inhibitors have been approved or are entering clinical trials for combination therapies (Table 1) [88, 89]. In this section, we highlight some of these drugs and their application for KRAS-mutant cancers.

RAF inhibitors

The RAF family consists of ARAF, BRAF, and CRAF, all sharing RAS as a common upstream activator. Belvarafenib (HM95573) is a pan-RAF dimerization inhibitor that demonstrates selective anticancer activity with either cobimetinib or cetuximab in preclinical models, as well as in cancer patients with RAS or RAF mutations, especially melanoma patients (Table 1). ARAF mutations are conducive to resistance to bevacafenib, indicating that the RAF subtype has a compensatory function, and a secondary mutation of a RAF member may reactivate the MAPK pathway to avoid cell death [117].
LXH-254, an ATP-competitive inhibitor of BRAF and CRAF [118], is used in multiple clinical trials for patients with NSCLC or melanoma (Table 1). The anticancer activity of LXH-254 is demonstrated in tumors carrying BRAF/RAS co-mutations, but it has moderate activity against cancers driven by KRAS mutants [119]. ARAF may also mediate LXH-254 resistance in RAS-mutant cancer cell lines [119], supporting the hypothesis that all RAF isoforms need to be suppressed at the same time in order to achieve tangible antineoplastic effects.
Lifirafenib (BGB-283), a dual inhibitor of RAF and EGFR, is being used in clinical studies enrolling patients with BRAF- or KRAS/NRAS-mutated solid tumors [120]. Preclinical study suggests that lifirafenib enhances the antitumor activity of MEK inhibitors (mirdametinib and selumetinib) in KRAS-mutant tumors [121]. A phase I study on the safety and pharmacokinetics of the combination with lifirfenib and mirdametinib in KRAS-mutant NSCLC is ongoing (NCT03905148).
Other RAF inhibitors, such as PLX8394 and TAK-580 (MLN2480), are being evaluated as single-agent therapeutics in patients with advanced unresectable solid tumors (NCT02428712) or low-grade glioma (NCT03429803), respectively. Vemurafenib, dabrafenib, and encorafenib are RAF inhibitors approved for the treatment of tumors with BRAF-V600E/K, but not RAS, mutations. It appears that monotherapy with RAF inhibitors is not efficient against cancers with KRAS mutations, suggesting that combination with other MAPK pathway inhibitors should be attempted.

MEK inhibitors

Three MEK inhibitors, including trametinib (GSK1120212), cobimetinib (XL518), and binimetinib (MEK162), are approved in combination with BRAF inhibitors for the treatment of patients with advanced melanoma harboring BRAF mutations (V600E or V600K). Although the MEK inhibitor selumetinib (AZD6244) is not efficient against melanoma, it has recently been approved for the treatment of neurofibromatosis type 1 in children. Compared with standard treatments, MEK inhibitors alone are not efficient against solid tumors driven by KRAS mutations [122125]. However, the combination of a MEK inhibitor and RAF inhibitor has shown promising activity against KRAS-mutant (especially KRAS-G13D) cells in vitro [126]. These findings provide the rationale for ongoing clinical trials that combine RAF (belvarafenib or LXH-254) and MEK inhibitors (cobimetinib or trametinib) to treat solid cancer with KRAS mutations (Table 1).
Two MEK inhibitors, pimisertib (MSC1936369B) and mirdametinib (PD0325901), are being evaluated for clinical activity against KRAS-mutant solid cancers (Table 1). Pimasertib alone is better than dacarbazine for improving progression-free survival in patient with BRAF- and NRAS-mutant melanoma [127, 128]. A combination of pimasertib and the MDM2 (a repressor of tumor suppressor TP53) inhibitor SAR405838 has shown preliminary antitumor activity in the treatment of solid cancers with RAS or RAF mutations [129]. Thus, targeting MEK combined with pharmacological TP53 induction may constitute a strategy for combating KRAS-mutant cancers.

ERK inhibitors

Although ERK inhibition is an effective strategy to overcome resistance to upstream MEK or RAF inhibitors [130132], the clinical development of ERK inhibitors has been retarded when compared to that of MEK and RAF inhibitors. In 2020, the FDA granted an expanded access program for the ERK inhibitor ulixertinib (BVD-523) for the treatment of cancer patients with abnormal MAPK pathways, including but not limited to those involving KRAS, NRAS, HRAS, BRAF, MEK, and ERK mutations [133]. Ulixertinib is currently being evaluated in combination with hydroxychloroquine (an autophagy inhibitor) or palbociclib (a CDK4/6 inhibitor) in patients with advanced pancreatic and other solid tumors (NCT041452973 and NCT03454035).
Certain ERK inhibitors, such as LY3214996, GDC-0994, and MK-8353, alone or in combination with other drugs, are in the early stages of clinical development. LY3214996 is being used with abemaciclib, hydroxychloroquine, or RMC-4630, in solid tumors, including KRAS-mutant cancers (Table 1). Despite strong preclinical data [134], patients with advanced solid tumors cannot tolerate combination therapy with GDC-0994 and cobimetinib [135]. However, GDC-0994 alone has acceptable side effects and showed anticancer activity in two patients with BRAF-mutant CRC [136]. The toxicity of GDC-0994 in combination with other MAPK inhibitors needs to be further investigated.
MK-8353 shows a tolerable safety profile and antitumor activity in melanoma patients with a BRAF-V600 mutation, rather than RAS mutation [137]. MK-8353 in combination with selumetinib or pembrolizumab is being investigated in patients with advanced malignancies, including CRC (Table 1). Although preclinical studies have shown that ERK mutations confer resistance to MAPK inhibitors [138], clinical studies have not yet reported the occurrence of acquired resistance to ERK inhibitors.

PI3K pathway inhibitors

Class I phosphatidylinositol-3-kinase (PI3K) is a lipid kinase that phosphorylates the signaling lipid phosphatidylinositol 4,5-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-triphosphate (PIP3), resulting in the recruitment of protein kinase B (PKB, best known as AKT) to the plasma membrane and subsequent activation of mTOR for cell growth and proliferation [139]. In contrast, PTEN, a tumor suppressor, can convert PIP3 to PIP2, thereby diminishing PI3K activity. PI3K consists of a catalytic subunit (p110, including p110α, p110β, p110γ, and p110δ isoforms) and a regulatory subunit (p85α or p85β). Compared with p110γ and p110δ, which are mainly expressed in immune cells, the expression of p110α and p110β is common in various cells or tissues. PIK3CA, which encodes p110α, is frequently mutated in cancer as an important drug target [139]. PIK3CA mutations can coexist with RAS mutations, while RAS mutations and MAPK pathway mutations are usually mutually exclusive [140, 141]. These co-mutation patterns might guide clinical trials to target different signals from the MAPK and PI3K pathways.
In May 2019, the FDA approved the first drug, alpelisib, as a specific p110α inhibitor for treating breast cancer. The combination of alpelisib with other chemotherapy (capecitabine or paclitaxel) is being tested in patients with PIK3CA-mutant CRC or gastric cancer patients (Table 1). The secondary p110α inhibitor GDC-0077 is under clinical development for the treatment of breast cancer (NCT04632992). Additionally, copanlisib (a pan class I PI3K inhibitor), duvelisib (a p110γ/p110δ inhibitor), and idelalisib (a p110δ inhibitor) are approved to treat adult patients with relapsed or refractory follicular lymphoma, but not solid tumors, with RAS mutations. Despite considerable efforts to combine PI3K and MEK inhibitors in preclinical models [142], such a combination can cause significant toxicity in patients with RAS-mutated cancers [143, 144]. Similarly, the combination of AKT or mTOR inhibitors and MAPK inhibitors is generally poorly tolerated by patients, which may limit their applications [145, 146].
In summary, compared with MAPK pathway inhibitors, monotherapy or combination therapy with PI3K pathway inhibitors has limited benefits for patients with KRAS-mutated cancers, although such PI3K inhibitors may reverse resistance to KRAS-G12C inhibitors (discussed later). Current PI3K inhibitors are still challenged by insufficient selectivity, which results from the close structural resemblance among ATP binding sites of different PI3K isoforms [139].

Others

The MAPK and PI3K pathways activate transcription factors to induce or suppress the expression of genes involved in multiple cellular processes. Targeting related downstream processes (for example, autophagy, glycolysis, and immunosuppression) also may help to mitigate the carcinogenic activity of RAS but will not be discussed in this review. Of note, genomic screenings have enabled the discovery of synthetic lethal partners to inhibit tumor growth in KRAS-mutant cancer cells (Table 4).
Table 4
Genes involved in synthetic lethality of mutant KRAS-dependent cancers
Synthetic lethal
genes
Full name
Main function
Tumor type
Reference
ANAPC1
Anaphase-promoting complex subunit 1
Mediates cell cycle progression
KRAS-mutant colon cancer
[147]
ARHGEF2
Rho/Rac guanine nucleotide exchange factor 2
Activates Rho-GTPases
KRAS-mutant pancreatic cancer
[148]
BCL2L1 (BCL-XL)
BCL2-like 1
Inhibits apoptosis
KRAS-mutant solid cancer
[149]
BIRC5 (survivin)
Baculoviral IAP repeat containing 5
Inhibits apoptosis
KRAS-mutant colon cancer
[150]
CDK1
Cyclin-dependent kinase 1
Mediates cell cycle progression
KRAS-mutant colon cancer
[151]
CDK4
Cyclin-dependent kinase 4
Mediates cell cycle progression
KRAS-mutant lung cancer
[152, 153]
DHODH
Dihydroorotate dehydrogenase (quinone)
Inhibits mitochondrial oxidative damage
KRAS-mutant pancreatic cancer
[154]
FGFR1
Fibroblast growth factor receptor 1
Mediates mitogenesis and differentiation
KRAS-mutant lung cancer
[155]
GATA2
GATA binding protein 2
Promotes development and survival
KRAS-mutant lung cancer
[156]
MAP3K7 (TAK1)
Mitogen-activated protein kinase kinase kinase 7
Promotes NF-κB activation
KRAS-mutant colon cancer
[157]
PLK1
Polo-like kinase 1
Promotes centrosome maturation and spindle assembly
KRAS-mutant chronic myelomonocytic leukemia or solid cancer
[147, 158, 159]
PRMT5
Protein arginine methyltransferase 5
Arginine methyltransferase
KRAS-mutant pancreatic cancer
[160]
PSMA5
Proteasome 20S subunit alpha 5
Mediates protein degradation
KRAS-mutant colon cancer
[147]
SHOC2
SHOC2 leucine-rich repeat scaffold protein
Promotes RAS signaling
KRAS-mutant leukemia and solid cancer
[161, 162]
SHP2 (PTPN11)
SH2 containing protein tyrosine phosphatase 2
Promotes RAS signaling
KRAS-mutant solid cancer
[113, 163]
SNAI2
Snail family transcriptional repressor 2
Promotes epithelial-mesenchymal transition
KRAS-mutant colon cancer
[164]
STK33
Serine/threonine kinase 33
Regulates cell cytoskeleton
KRAS-mutant solid cancer
[165]
TBK1
TANK binding kinase 1
Promotes NF-κB activation
KRAS-mutant lung cancer
[166]
WT1
WT1 transcription factor
Promotes development and survival
KRAS-mutant lung cancer
[167]
XPO1
Exportin 1
Mediates nuclear export
KRAS-mutant lung cancer
[168]
YAP1
Yes1-associated transcriptional regulator
Mediates the Hippo signaling pathway
KRAS-mutant pancreatic cancer
[167]

Direct KRAS inhibition

Covalent KRAS-G12C inhibitors

Historically, KRAS was considered "undruggable" because it does not have a classical pocket suitable for binding small inhibitory molecules [18]. Recent structural studies and drug design efforts to produce G12Ci have changed this view (Table 5). The pioneering work of Shokat and colleagues uncovered a hidden pocket (switch-II) in the KRAS-GDP complex that is located next to the mutant cysteine 12 [172]. The proximity of switch-II to cysteine 12 facilitated the development of covalent inhibitors of switch-II, thereby achieving allosteric inhibition of cysteine 12 to prevent the nucleotide exchange catalyzed by GEF and diminish the subsequent interaction between RAS and RAF (Fig. 2c) [170, 172, 173]. Since wild-type KRAS lacks cysteine in the active site, the covalent inhibition of cysteine 12 is expected to be highly specific. ARS-1620 structurally modified from compound 12 [172], 1_AM [169], and ARS-853 [170] turned out to be the first G12Ci to elicit effective tumor suppression in patient-derived xenograft modes [171].
Table 5
Development history and application status of KRAS-G12C inhibitors
Name
Application date
Institutions
Structure
Status
Reference/ trial number
1_AM
August 2017
Dana-Farber Cancer Institute
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Figa_HTML.png
Preclinical
[169]
Adagrasib
October 2019
Mirati
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Figb_HTML.png
Clinical (approved)
[16]
NCT04685135; NCT03785249; NCT04330664; NCT04793958
ARS-853
January 2016
Memorial Sloan Kettering Cancer Center
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Figc_HTML.png
Preclinical
[170]
ARS-1620
January 2018
Wellspring Biosciences
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Figd_HTML.png
Preclinical
[171]
Compound 12
November 2013
University of California
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Fige_HTML.png
Preclinical
[172]
D-1553
October 2020
InventisBio
Structure not disclosed
Clinical (recruiting)
NCT04585035
GDC-6036
June 2020
Genentech
Structure not disclosed
Clinical (recruiting)
NCT04449874
JNJ-74699157
July 2019
Araxes/J&J
Structure not disclosed
Clinical (terminated)
NCT04006301
LY3499446
November 2019
Eli Lilly
Structure not disclosed
Clinical (terminated)
NCT04165031
Sotorasib
October 2019
Amgen
https://static-content.springer.com/image/art%3A10.1186%2Fs12943-021-01422-7/MediaObjects/12943_2021_1422_Figf_HTML.png
Clinical (approved)
[15]
NCT04303780; NCT03600883; NCT04613596
Amgen and Mirati Therapeutics developed two structure-optimized covalent G12Ci formulations, sotorasib [15] and adagrasib [16]. Compared with ARS-1620, sotorasib and adagrasib have larger surface grooves, which enhance the effectiveness of irreversible interactions with the H95 residue in the 3 helix of KRAS-G12C protein [15, 16]. Sotorasib and adagrasib mediate selective tumor suppression activity across a panel of cancer cell lines harboring the KRAS-G12C mutation [14, 16, 174]. Durable responses to sotorasib have been observed in immunocompetent rather than immunodeficient tumor-bearing mice [175]. This may be explained by the fact that sotorasib induces the production of chemokines (CXCL10 and CXCL11) and potential damage-associated molecular patterns (DAMPs), leading to an immune response mediated by cytotoxic lymphocytes (Fig. 5) [175]. Accordingly, the combined use of anti–PD-1 antibodies further enhances sotorasib-induced tumor suppression in mouse models [175]. Whether G12Ci can be used to produce cancer-preventive or therapeutic immune responses is an open question. Interestingly, patient-derived xenograft models indicate that individual genetic alteration (such as in KRAS, TP53, STK11, or CDKN2A) cannot predict the anticancer activity of adagrasib [176]. CRISPR screens have identified negative (MYC, SHP2, mTOR, RPS6, CDK1, CDK2, CDK4/6, and RB1) and positive (KEAP1 and CBL) regulators of adagrasib sensitivity in NSCLC cells [176]. Drug screening further revealed that a pan-EGFR family inhibitor (afatinib), a SHP2 inhibitor (RMC-4550), mTOR inhibitors (vistusertib and everolimus), and a CDK4/6 inhibitor (palbociclib) increase the response rate to adagrasib in cell cultures and mouse models [176], providing potential optimization strategies for translational research.
Clinical studies have shown some promising antitumor activity of sotorasib or adagrasib in patients with KRAS-G12C-mutated NSCLC that previously had been treated with platinum-based chemotherapy and/or PD-1/PD-L1 blockade. In a phase II trial, 37.1% (46/124) of such NSCLC patients responded to single-agent sotorasib (900 mg/kg, once daily) with a median duration of response of 11.1 months across all PD-L1 expression level subgroups (Table 6) [178]. The activity of sotorasib has been observed in patients with mutations in TP53, STK11, and KEAP1 [178], which are associated with a poor prognosis in NSCLC [179]. Another phase I/II trial involved 129 KRAS-G12C–mutant cancer patients, in which 32.2% of NSCLC, 7.1% of CRC, and 14.3% of other tumor patients showed an objective response to sotorasib (Table 6) [177]. In May 2021, the FDA granted accelerated approval to sotorasib for the treatment of KRAS-G12C–mutated NSCLC. In June 2021, the FDA awarded breakthrough therapy designation to adagrasib for KRAS-G12C–mutated NSCLC based on an unpublished phase I/II study showing that 45% (23/51) of participants responded and 51% (26/51) of them were in stable conditions after using adagrasib (600 mg/kg, twice daily). Although the elimination half-lives of sotorasib (6 hours) and adagrasib (25 hours) are different, they have similar treatment-related adverse events (e.g., nausea, diarrhea, and vomiting). A number of clinical trials are underway to evaluate the antitumor activity of sotorasib or adagrasib alone or in combination with target drugs (docetaxel, pembrolizumab, cetuximab, afatinib, or TNO155) in solid cancers carrying KRAS-G12C mutations (Table 1). Specifically, two phase III trials will test the combination of sotorasib or adagrasib with docetaxel or cetuximab to treat KRAS-G12C–mutant NSCLC or CRC, respectively (NCT04685135 and NCT04793958).
Table 6
Clinical results of sotorasib therapy in advanced cancer with KRAS-G12C
 
Hong et al., 2020
(n = 129) [177]
Skoulidis et al., 2021
(n = 126) [178]
Characteristics
Median age (range, year)
62 (33–83)
63.5 (37–80)
NSCLC (n)
59
126
CRC (n)
42
0
Other solid cancer (n)
28
0
Treatment
Sotorasib (orally 180-960 mg/kg, once daily)
Sotorasib (orally 960 mg/kg, once daily)
Efficacy
Objective response (%)
NSCLC: 32.2; CRC: 7.1; Other: 14.3
37.1
Disease control (%)
NSCLC: 88.1; CRC: 73.8; Other: 75.0
80.6
Complete response (%)
NSCLC: 0; CRC: 0; Other: 0
3.2
Partial response (%)
NSCLC: 32.2; CRC: 7.1; Other: 14.3
33.9
Stable disease (%)
NSCLC: 55.9; CRC: 66.7; Other: 60.7
43.5
Progressive disease (%)
NSCLC: 8.5; CRC: 23.8; Other: 14.3
16.1
Could not be evaluated (%)
NSCLC: 1.7; CRC: 2.4; Other: 7.1
1.6
Adverse events
Any grade (%)
96.9
99.2
Serious (%)
45.0
45.3
Resulting in discontinuation of treatment (%)
7.0
7.1
The clinical development of other G12Ci compounds, such as GDC-6036 and D-1553, might provide additional opportunities for selectively targeting advanced solid tumors with KRAS-G12C mutations (Table 1). Notably, the clinical trial of two G12Ci formulations, LY3499446 and JNJ-74699157, has been terminated due to significant toxicity (NCT04165031 and NCT04006301). It remains to be seen whether these toxicities are caused by covalent or noncovalent off-targets.

Pan-KRAS inhibitors

BI-2852 is a pan-KRAS inhibitor that binds between the switch-I and switch-II pockets, thereby blocking the interaction of KRAS protein with GEF, GAP, and its downstream effectors [180]. Early preclinical studies confirmed its activity in blocking the KRAS pathway in NSCLC cells [180]. Using FR054 to inhibit glycosylation reactions further enhances the anticancer activity of BI-2852 against PDAC cells [181], supporting that the hexosamine biosynthesis pathway is a potential target for the treatment of KRAS-mutant cancers [182].
Revolution Medicines utilized a tri-complex technology platform to design a type of RAS(ON) inhibitor. RAS(ON) inhibitors (for example, RM-007 and RM-008) act as molecular glues to mediate protein-protein interactions between different mutant KRAS proteins and an endogenous protein (cyclophilin), thereby inhibiting the binding of mutant KRAS to SOS1 and effector proteins. Thus, the mode of action of RAS(ON) inhibitors is different from that of RAS(OFF) inhibitors, including G12Ci.
More recently, a small-molecule compound called Pen-cRaf-v1 has been identified as a pan-RAS inhibitor capable of targeting G12C and non-G12C RAS mutants to inhibit RAS-effector interaction [183]. Further animal studies are needed to determine the activity, metabolism, and toxicity of pan-KRAS inhibitors before their translational application into clinical medicine.

Others

An interesting trend in recent drug discovery is the selective induction of protein degradation through the proteasome. Proteolysis-targeting chimera (PROTAC) technology can be used to design new drugs that bridge the target protein to E3 ligases, hoping to achieve the target’s degradation to nonfunctional fragments. Whether this approach may be successful for the destruction of oncogenic RAS remains to be explored [184, 185].

Mechanisms of adaptation or resistance to KRAS-G12C inhibitors

Producing new KRAS-G12C protein

The activation of the EGFR-SHP2 pathway maintains newly synthesized KRAS-G12C protein in an active GTP-binding form, thereby leading to the adaptation of KRAS-G12C–mutated cancer cells to ARS-1620 (Fig. 6a) [186]. The cell cycle regulator aurora kinase A (AURKA) binds newly produced KRAS-G12C, which in turn stabilizes the interaction between CRAF and KRAS and mediates subsequent ERK effector signals for cell cycle progression [186]. Consequently, the inhibition of EGFR or AURKA reverses the adaptation of cancer cells to ARS-1620. These preclinical studies provide clues for the development of combined strategies that target EGFR or the cell cycle regulator to delay the development of resistance to KRAS-G12C inhibitors [186]. In addition to AURKA inhibitors (e.g., alisertib), many drugs that are already in clinical use or under development target various cell cycle regulators, especially CDKs. AURKA inhibitors and CDK inhibitors both have shown promise in the treatment of various types of cancer, including KRAS-mutant cancers [187192].

Activating wild-type RAS

Wild-type and mutant RAS subtypes co-exist in the same cell, thus providing a feedback mechanism to reactivate RAS signaling if one of the two RAS pathways is blocked. As such, even if KRAS-G12C is effectively and completely inhibited, residual wild-type RAS (NRAS and HRAS) activity may confer resistance to G12Ci. Multiple RTKs (EGFR, HER2, FGFR, and c-MET), instead of a single RTK, activate wild-type RAS, resulting in acquired resistance to ARS-1620 and sotoracide (Fig. 6b) [193]. This feedback reactivation of wild-type RAS occurs in parallel to the neosynthesis of KRAS-G12C protein, resulting in drug resistance. Since SHP2 and SOS1 are the common nodes of RTK signals, SHP2 inhibitors (e.g., TNO155, SHP099, and RMC-4550) or SOS1 inhibitors (e.g., BAY-293) may either enhance the activity of G12Ci or reverse adaptive resistance. This hypothesis has been confirmed in preclinical models [83, 116, 186, 193197], and such inhibitors are now entering clinical evaluation (NCT04330664).

Inducing epithelial-to-mesenchymal transition

Epithelial-to-mesenchymal transition (EMT), the process whereby epithelial cells are transformed into mesenchymal cells, is one of the acquired resistance mechanisms to antineoplastic therapies, including TKIs [198]. The induction of EMT in sotorasib-sensitive NSCLC cells by adding TGFβ or using transfection with SNAIL leads to acquired resistance to sotorasib through the activation of the PI3K pathway, which is not associated with significant AKT activation [199]. This suggests that the classical KRAS-PI3K-AKT pathway is not essential for acquired resistance to sotorasib, whereas KRAS-independent PI3K activation favors such resistance in lung cancer cells [200]. In the latter, the insulin-like growth factor receptor (IGFR)-IRS1 pathway, as a key upstream signal, mediates PI3K activation in a SHP2-independent manner, leading to acquired resistance to sotorasib or ARS-1620 in NSCLC cells (Fig. 6c) [199]. Therefore, the clinical optimization of G12Ci may profit from patient stratification based on EMT status.
In other cases, the activation of the PI3K-AKT-mTOR pathway clearly limits the efficacy of G12Ci, such as sotoracide or ARS1620, against NSCLC and PDAC cells [176, 201, 202]. Hence, the mechanisms of PI3K pathway-mediated resistance to G12Ci may depend on the tumor type and the degree of cellular (de)differentiation.

Inducing secondary genetic alterations

Specific secondary genetic alterations may provide additional information to predict G12Ci responses (Fig. 6d). Among 38 patients with KRAS-G12C-mutated solid cancers who received adagrasib monotherapy, 45% displayed a putative genetic mechanism for resistance [203]. The reactivation of RAS-MAPK signaling by 10 genetic alterations affecting the RAS-RAF-MEK-ERK pathway has been described in an NSCLC patient with acquired adagrasib resistance [204]. Secondary KRAS mutations, gain-of-function mutations of the MAPK pathway, loss-of-function mutations in tumor suppressor genes, gene fusion, and gene amplification are conducive to acquired resistance to G12Ci (Fig. 6d).
In vitro and in vivo, KRAS mutation studies further confirmed that the expression of clinically observed switch-II pocket mutations conferred resistance to adagrasib in KRAS-G12C–mutant BaF3 cells (a murine interleukin-3–dependent pro-B cell line) [203, 204]. Thus, the mutations Y96C, Y96D, or Y96S led to resistance to both adagrasib and sotorasib [203, 204]. In contrast, H95D, H95Q, and H95R mutants remained sensitive to sotorasib [203]. Interestingly, a RAS(ON) inhibitor retained its therapeutic index against cells harboring dual G12C/Y96D mutations [204], supporting the notion that RAS(ON) inhibitors mediate the inhibition of oncogenic RAS by a completely different mechanism.
Collectively, these studies provide proof of concept and mechanistic support for a combination therapy that suppresses adaptive genetic alterations. In fact, clinical trials evaluating adagrasib or sotorasib in combination with inhibitors of RTKs, MAPK, SOS1, or SHP2 are underway (NCT04330664 and NCT04185883) to explore novel strategies for overcoming cancer drug resistance [79, 205].

Conclusions and future perspectives

With the development of G12Ci, we now have a tool to directly and irreversibly inhibit KRAS-G12C oncoprotein in patients [17]. However, the excitement spurred by this discovery has been dampened by the fact that the vast majority of patients fail to respond to G12Ci treatment due to primary or acquired resistance [177, 203, 204, 206]. Thus, there are still many outstanding problems to be solved.

First, how can we develop next-generation inhibitors?

Different G12Ci compounds exhibit distinct activities and toxicities. For example, adagrasib was found to bind to KRAS-G12C equally in a series of different cell lines, despite the major variability in downstream signals [176]. The main risk of covalent inhibitors is the possibility of nonspecific reactions with off-target proteins containing cysteine residues [173]. Thus, the molecular identification of proteins accounting for off-target effects of G12Ci may improve structure-based drug design [203]. In addition to organ-mediated drug metabolism, tumor-resident microorganisms can directly decompose chemotherapeutic drugs to cause drug resistance [207]. Hence, optimizing the in vivo pharmacokinetic properties of KRAS inhibitors and evaluating the (in)activity of their metabolites is still an important area for examination. Finally, it remains to be seen whether it is possible to develop additional mutation-specific inhibitors or pan-mutant KRAS inhibitors [208].

Second, how can we design combination therapies?

The design and implementation of strategies to minimize or overcome drug resistance is an important goal of clinical oncology in order to achieve complete and durable clinical responses [209]. The observed tumor heterogeneity and the extensive feedback between RAS and other tumor-related signals may promote drug resistance. The combination of several drugs intercepting different signaling pathways (e.g., upstream signaling, downstream signaling, parallel signaling, cell cycle processes, or immune checkpoints) may prevent or delay the development of therapy resistance, but usually at the cost of increased toxicity [14, 17]. A number of clinical trials combining G12Ci with other established agents (including TKIs and ICIs) are being launched. The design of such trials should avoid random combinations and follow a rationale based on the genetic, metabolic, and immune mechanisms of drug resistance.

Third, how can we develop predictive cancer biomarkers?

Predictive biomarkers can guide treatment decisions by indicating the likely impact of a particular therapy on a patient. Some genetic alterations, especially secondary gene mutation, are associated with the development of adagrasib resistance in patients. In addition to using endoscopic ultrasound-guided fine needle aspiration, the analysis of circulating tumor DNA combined with next-generation sequencing technologies provides a way for examining the genetic characteristics of tumors. Despite these obvious technological advances, circulating metabolites or proteins should not be neglected as potential biomarkers, given that their quantitation would be much more convenient.
In summary, the development of ever more efficient and specific drugs blocking oncogenic RAS must be in parallel with major efforts to avoid and overcome therapeutic resistance. Given the continued efforts of industry, academia, and health care providers, as well as the latest breakthroughs in basic, clinical, and translational research on G12Ci, the prospects look bright.

Acknowledgements

We thank Dave Primm (Department of Surgery, University of Texas Southwestern Medical Center) for his critical reading of the manuscript.

Declarations

Not applicable.
All authors agreed to publication.

Competing interests

The authors declare no competing interests. GK is a co-founder of EverImmune, Samsara Therapeutics, and Therafast Bio.
Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat Harvey JJ. An Unidentified Virus Which Causes the Rapid Production of Tumours in Mice. Nature. 1964;204:1104–5.PubMedCrossRef Harvey JJ. An Unidentified Virus Which Causes the Rapid Production of Tumours in Mice. Nature. 1964;204:1104–5.PubMedCrossRef
3.
Zurück zum Zitat Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3(6):459–65.PubMedCrossRef Malumbres M, Barbacid M. RAS oncogenes: the first 30 years. Nat Rev Cancer. 2003;3(6):459–65.PubMedCrossRef
4.
Zurück zum Zitat Johnson L, Greenbaum D, Cichowski K, Mercer K, Murphy E, Schmitt E, et al. K-ras is an essential gene in the mouse with partial functional overlap with N-ras. Genes Dev. 1997;11(19):2468–81.PubMedPubMedCentralCrossRef Johnson L, Greenbaum D, Cichowski K, Mercer K, Murphy E, Schmitt E, et al. K-ras is an essential gene in the mouse with partial functional overlap with N-ras. Genes Dev. 1997;11(19):2468–81.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Shimizu K, Goldfarb M, Suard Y, Perucho M, Li Y, Kamata T, et al. Three human transforming genes are related to the viral ras oncogenes. Proc Natl Acad Sci U S A. 1983;80(8):2112–6.PubMedPubMedCentralCrossRef Shimizu K, Goldfarb M, Suard Y, Perucho M, Li Y, Kamata T, et al. Three human transforming genes are related to the viral ras oncogenes. Proc Natl Acad Sci U S A. 1983;80(8):2112–6.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Shimizu K, Birnbaum D, Ruley MA, Fasano O, Suard Y, Edlund L, et al. Structure of the Ki-ras gene of the human lung carcinoma cell line Calu-1. Nature. 1983;304(5926):497–500.PubMedCrossRef Shimizu K, Birnbaum D, Ruley MA, Fasano O, Suard Y, Edlund L, et al. Structure of the Ki-ras gene of the human lung carcinoma cell line Calu-1. Nature. 1983;304(5926):497–500.PubMedCrossRef
7.
Zurück zum Zitat Santos E, Martin-Zanca D, Reddy EP, Pierotti MA, Della Porta G, Barbacid M. Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient. Science. 1984;223(4637):661–4.PubMedCrossRef Santos E, Martin-Zanca D, Reddy EP, Pierotti MA, Della Porta G, Barbacid M. Malignant activation of a K-ras oncogene in lung carcinoma but not in normal tissue of the same patient. Science. 1984;223(4637):661–4.PubMedCrossRef
8.
Zurück zum Zitat Parada LF, Weinberg RA. Presence of a Kirsten murine sarcoma virus ras oncogene in cells transformed by 3-methylcholanthrene. Mol Cell Biol. 1983;3(12):2298–301.PubMedPubMedCentral Parada LF, Weinberg RA. Presence of a Kirsten murine sarcoma virus ras oncogene in cells transformed by 3-methylcholanthrene. Mol Cell Biol. 1983;3(12):2298–301.PubMedPubMedCentral
9.
Zurück zum Zitat Johnson L, Mercer K, Greenbaum D, Bronson RT, Crowley D, Tuveson DA, et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature. 2001;410(6832):1111–6.PubMedCrossRef Johnson L, Mercer K, Greenbaum D, Bronson RT, Crowley D, Tuveson DA, et al. Somatic activation of the K-ras oncogene causes early onset lung cancer in mice. Nature. 2001;410(6832):1111–6.PubMedCrossRef
10.
Zurück zum Zitat Sukumar S, Notario V, Martin-Zanca D, Barbacid M. Induction of mammary carcinomas in rats by nitroso-methylurea involves malignant activation of H-ras-1 locus by single point mutations. Nature. 1983;306(5944):658–61.PubMedCrossRef Sukumar S, Notario V, Martin-Zanca D, Barbacid M. Induction of mammary carcinomas in rats by nitroso-methylurea involves malignant activation of H-ras-1 locus by single point mutations. Nature. 1983;306(5944):658–61.PubMedCrossRef
11.
Zurück zum Zitat Chen X, Mitsutake N, LaPerle K, Akeno N, Zanzonico P, Longo VA, et al. Endogenous expression of Hras(G12V) induces developmental defects and neoplasms with copy number imbalances of the oncogene. Proc Natl Acad Sci U S A. 2009;106(19):7979–84.PubMedPubMedCentralCrossRef Chen X, Mitsutake N, LaPerle K, Akeno N, Zanzonico P, Longo VA, et al. Endogenous expression of Hras(G12V) induces developmental defects and neoplasms with copy number imbalances of the oncogene. Proc Natl Acad Sci U S A. 2009;106(19):7979–84.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat Burd CE, Liu W, Huynh MV, Waqas MA, Gillahan JE, Clark KS, et al. Mutation-specific RAS oncogenicity explains NRAS codon 61 selection in melanoma. Cancer Discov. 2014;4(12):1418–29.PubMedPubMedCentralCrossRef Burd CE, Liu W, Huynh MV, Waqas MA, Gillahan JE, Clark KS, et al. Mutation-specific RAS oncogenicity explains NRAS codon 61 selection in melanoma. Cancer Discov. 2014;4(12):1418–29.PubMedPubMedCentralCrossRef
14.
15.
Zurück zum Zitat Lanman BA, Allen JR, Allen JG, Amegadzie AK, Ashton KS, Booker SK, et al. Discovery of a Covalent Inhibitor of KRAS(G12C) (AMG 510) for the Treatment of Solid Tumors. J Med Chem. 2020;63(1):52–65.PubMedCrossRef Lanman BA, Allen JR, Allen JG, Amegadzie AK, Ashton KS, Booker SK, et al. Discovery of a Covalent Inhibitor of KRAS(G12C) (AMG 510) for the Treatment of Solid Tumors. J Med Chem. 2020;63(1):52–65.PubMedCrossRef
16.
Zurück zum Zitat Fell JB, Fischer JP, Baer BR, Blake JF, Bouhana K, Briere DM, et al. Identification of the Clinical Development Candidate MRTX849, a Covalent KRAS(G12C) Inhibitor for the Treatment of Cancer. J Med Chem. 2020;63(13):6679–93.PubMedCrossRef Fell JB, Fischer JP, Baer BR, Blake JF, Bouhana K, Briere DM, et al. Identification of the Clinical Development Candidate MRTX849, a Covalent KRAS(G12C) Inhibitor for the Treatment of Cancer. J Med Chem. 2020;63(13):6679–93.PubMedCrossRef
17.
Zurück zum Zitat Kim D, Xue JY, Lito P. Targeting KRAS(G12C): From Inhibitory Mechanism to Modulation of Antitumor Effects in Patients. Cell. 2020;183(4):850–9.PubMedCrossRefPubMedCentral Kim D, Xue JY, Lito P. Targeting KRAS(G12C): From Inhibitory Mechanism to Modulation of Antitumor Effects in Patients. Cell. 2020;183(4):850–9.PubMedCrossRefPubMedCentral
18.
19.
Zurück zum Zitat Murugan AK, Grieco M, Tsuchida N. RAS mutations in human cancers: Roles in precision medicine. Semin Cancer Biol. 2019;59:23–35.PubMedCrossRef Murugan AK, Grieco M, Tsuchida N. RAS mutations in human cancers: Roles in precision medicine. Semin Cancer Biol. 2019;59:23–35.PubMedCrossRef
21.
Zurück zum Zitat Li S, Balmain A, Counter CM. A model for RAS mutation patterns in cancers: finding the sweet spot. Nat Rev Cancer. 2018;18(12):767–77.PubMedCrossRef Li S, Balmain A, Counter CM. A model for RAS mutation patterns in cancers: finding the sweet spot. Nat Rev Cancer. 2018;18(12):767–77.PubMedCrossRef
22.
Zurück zum Zitat Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7.PubMedPubMedCentralCrossRef Yachida S, Jones S, Bozic I, Antal T, Leary R, Fu B, et al. Distant metastasis occurs late during the genetic evolution of pancreatic cancer. Nature. 2010;467(7319):1114–7.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Zhang J, Fujimoto J, Zhang J, Wedge DC, Song X, Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014;346(6206):256–9.PubMedPubMedCentralCrossRef Zhang J, Fujimoto J, Zhang J, Wedge DC, Song X, Zhang J, et al. Intratumor heterogeneity in localized lung adenocarcinomas delineated by multiregion sequencing. Science. 2014;346(6206):256–9.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Cook JH, Melloni GEM, Gulhan DC, Park PJ, Haigis KM. The origins and genetic interactions of KRAS mutations are allele- and tissue-specific. Nat Commun. 2021;12(1):1808.PubMedPubMedCentralCrossRef Cook JH, Melloni GEM, Gulhan DC, Park PJ, Haigis KM. The origins and genetic interactions of KRAS mutations are allele- and tissue-specific. Nat Commun. 2021;12(1):1808.PubMedPubMedCentralCrossRef
25.
Zurück zum Zitat Zhang X, Cao J, Miller SP, Jing H, Lin H. Comparative Nucleotide-Dependent Interactome Analysis Reveals Shared and Differential Properties of KRas4a and KRas4b. ACS Cent Sci. 2018;4(1):71–80.PubMedCrossRef Zhang X, Cao J, Miller SP, Jing H, Lin H. Comparative Nucleotide-Dependent Interactome Analysis Reveals Shared and Differential Properties of KRas4a and KRas4b. ACS Cent Sci. 2018;4(1):71–80.PubMedCrossRef
26.
Zurück zum Zitat Tsai FD, Lopes MS, Zhou M, Court H, Ponce O, Fiordalisi JJ, et al. K-Ras4A splice variant is widely expressed in cancer and uses a hybrid membrane-targeting motif. Proc Natl Acad Sci U S A. 2015;112(3):779–84.PubMedPubMedCentralCrossRef Tsai FD, Lopes MS, Zhou M, Court H, Ponce O, Fiordalisi JJ, et al. K-Ras4A splice variant is widely expressed in cancer and uses a hybrid membrane-targeting motif. Proc Natl Acad Sci U S A. 2015;112(3):779–84.PubMedPubMedCentralCrossRef
27.
28.
Zurück zum Zitat Plowman SJ, Williamson DJ, O'Sullivan MJ, Doig J, Ritchie AM, Harrison DJ, et al. While K-ras is essential for mouse development, expression of the K-ras 4A splice variant is dispensable. Mol Cell Biol. 2003;23(24):9245–50.PubMedPubMedCentralCrossRef Plowman SJ, Williamson DJ, O'Sullivan MJ, Doig J, Ritchie AM, Harrison DJ, et al. While K-ras is essential for mouse development, expression of the K-ras 4A splice variant is dispensable. Mol Cell Biol. 2003;23(24):9245–50.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Slebos RJ, Kibbelaar RE, Dalesio O, Kooistra A, Stam J, Meijer CJ, et al. K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N Engl J Med. 1990;323(9):561–5.PubMedCrossRef Slebos RJ, Kibbelaar RE, Dalesio O, Kooistra A, Stam J, Meijer CJ, et al. K-ras oncogene activation as a prognostic marker in adenocarcinoma of the lung. N Engl J Med. 1990;323(9):561–5.PubMedCrossRef
31.
Zurück zum Zitat Pan W, Yang Y, Zhu H, Zhang Y, Zhou R, Sun X. KRAS mutation is a weak, but valid predictor for poor prognosis and treatment outcomes in NSCLC: A meta-analysis of 41 studies. Oncotarget. 2016;7(7):8373–88.PubMedPubMedCentralCrossRef Pan W, Yang Y, Zhu H, Zhang Y, Zhou R, Sun X. KRAS mutation is a weak, but valid predictor for poor prognosis and treatment outcomes in NSCLC: A meta-analysis of 41 studies. Oncotarget. 2016;7(7):8373–88.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Shepherd FA, Domerg C, Hainaut P, Janne PA, Pignon JP, Graziano S, et al. Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol. 2013;31(17):2173–81.PubMedPubMedCentralCrossRef Shepherd FA, Domerg C, Hainaut P, Janne PA, Pignon JP, Graziano S, et al. Pooled analysis of the prognostic and predictive effects of KRAS mutation status and KRAS mutation subtype in early-stage resected non-small-cell lung cancer in four trials of adjuvant chemotherapy. J Clin Oncol. 2013;31(17):2173–81.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Douillard JY, Shepherd FA, Hirsh V, Mok T, Socinski MA, Gervais R, et al. Molecular predictors of outcome with gefitinib and docetaxel in previously treated non-small-cell lung cancer: data from the randomized phase III INTEREST trial. J Clin Oncol. 2010;28(5):744–52.PubMedCrossRef Douillard JY, Shepherd FA, Hirsh V, Mok T, Socinski MA, Gervais R, et al. Molecular predictors of outcome with gefitinib and docetaxel in previously treated non-small-cell lung cancer: data from the randomized phase III INTEREST trial. J Clin Oncol. 2010;28(5):744–52.PubMedCrossRef
34.
Zurück zum Zitat Zhu CQ, da Cunha SG, Ding K, Sakurada A, Cutz JC, Liu N, et al. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. 2008;26(26):4268–75.PubMedCrossRef Zhu CQ, da Cunha SG, Ding K, Sakurada A, Cutz JC, Liu N, et al. Role of KRAS and EGFR as biomarkers of response to erlotinib in National Cancer Institute of Canada Clinical Trials Group Study BR.21. J Clin Oncol. 2008;26(26):4268–75.PubMedCrossRef
35.
Zurück zum Zitat Massarelli E, Varella-Garcia M, Tang X, Xavier AC, Ozburn NC, Liu DD, et al. KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res. 2007;13(10):2890–6.PubMedCrossRef Massarelli E, Varella-Garcia M, Tang X, Xavier AC, Ozburn NC, Liu DD, et al. KRAS mutation is an important predictor of resistance to therapy with epidermal growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer. Clin Cancer Res. 2007;13(10):2890–6.PubMedCrossRef
36.
Zurück zum Zitat Hames ML, Chen H, Iams W, Aston J, Lovly CM, Horn L. Correlation between KRAS mutation status and response to chemotherapy in patients with advanced non-small cell lung cancer. Lung Cancer. 2016;92:29–34.PubMedCrossRef Hames ML, Chen H, Iams W, Aston J, Lovly CM, Horn L. Correlation between KRAS mutation status and response to chemotherapy in patients with advanced non-small cell lung cancer. Lung Cancer. 2016;92:29–34.PubMedCrossRef
37.
Zurück zum Zitat Kim JH, Kim HS, Kim BJ. Prognostic value of KRAS mutation in advanced non-small-cell lung cancer treated with immune checkpoint inhibitors: A meta-analysis and review. Oncotarget. 2017;8(29):48248–52.PubMedPubMedCentralCrossRef Kim JH, Kim HS, Kim BJ. Prognostic value of KRAS mutation in advanced non-small-cell lung cancer treated with immune checkpoint inhibitors: A meta-analysis and review. Oncotarget. 2017;8(29):48248–52.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Jeanson A, Tomasini P, Souquet-Bressand M, Brandone N, Boucekine M, Grangeon M, et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). J Thorac Oncol. 2019;14(6):1095–101.PubMedCrossRef Jeanson A, Tomasini P, Souquet-Bressand M, Brandone N, Boucekine M, Grangeon M, et al. Efficacy of Immune Checkpoint Inhibitors in KRAS-Mutant Non-Small Cell Lung Cancer (NSCLC). J Thorac Oncol. 2019;14(6):1095–101.PubMedCrossRef
39.
Zurück zum Zitat Sun L, Hsu M, Cohen RB, Langer CJ, Mamtani R, Aggarwal C. Association Between KRAS Variant Status and Outcomes With First-line Immune Checkpoint Inhibitor-Based Therapy in Patients With Advanced Non-Small-Cell Lung Cancer. JAMA Oncol. 2021;7(6):937–9.PubMedCrossRefPubMedCentral Sun L, Hsu M, Cohen RB, Langer CJ, Mamtani R, Aggarwal C. Association Between KRAS Variant Status and Outcomes With First-line Immune Checkpoint Inhibitor-Based Therapy in Patients With Advanced Non-Small-Cell Lung Cancer. JAMA Oncol. 2021;7(6):937–9.PubMedCrossRefPubMedCentral
40.
Zurück zum Zitat Kartolo A, Feilotter H, Hopman W, Fung AS, Robinson A. A single institution study evaluating outcomes of PD-L1 high KRAS-mutant advanced non-small cell lung cancer (NSCLC) patients treated with first line immune checkpoint inhibitors. Cancer Treat Res Commun. 2021;27:100330.PubMedCrossRef Kartolo A, Feilotter H, Hopman W, Fung AS, Robinson A. A single institution study evaluating outcomes of PD-L1 high KRAS-mutant advanced non-small cell lung cancer (NSCLC) patients treated with first line immune checkpoint inhibitors. Cancer Treat Res Commun. 2021;27:100330.PubMedCrossRef
41.
Zurück zum Zitat Nardo G, Carlet J, Marra L, Bonanno L, Boscolo A, Dal Maso A, et al. Detection of Low-Frequency KRAS Mutations in cfDNA From EGFR-Mutated NSCLC Patients After First-Line EGFR Tyrosine Kinase Inhibitors. Front Oncol. 2020;10:607840.PubMedCrossRef Nardo G, Carlet J, Marra L, Bonanno L, Boscolo A, Dal Maso A, et al. Detection of Low-Frequency KRAS Mutations in cfDNA From EGFR-Mutated NSCLC Patients After First-Line EGFR Tyrosine Kinase Inhibitors. Front Oncol. 2020;10:607840.PubMedCrossRef
42.
Zurück zum Zitat Bournet B, Muscari F, Buscail C, Assenat E, Barthet M, Hammel P, et al. KRAS G12D Mutation Subtype Is A Prognostic Factor for Advanced Pancreatic Adenocarcinoma. Clin Transl Gastroenterol. 2016;7:e157.PubMedPubMedCentralCrossRef Bournet B, Muscari F, Buscail C, Assenat E, Barthet M, Hammel P, et al. KRAS G12D Mutation Subtype Is A Prognostic Factor for Advanced Pancreatic Adenocarcinoma. Clin Transl Gastroenterol. 2016;7:e157.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E, Marten A, Winkelmann C, et al. Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the 'Arbeitsgemeinschaft Internistische Onkologie' (AIO-PK0104). Gut. 2013;62(5):751–9.PubMedCrossRef Heinemann V, Vehling-Kaiser U, Waldschmidt D, Kettner E, Marten A, Winkelmann C, et al. Gemcitabine plus erlotinib followed by capecitabine versus capecitabine plus erlotinib followed by gemcitabine in advanced pancreatic cancer: final results of a randomised phase 3 trial of the 'Arbeitsgemeinschaft Internistische Onkologie' (AIO-PK0104). Gut. 2013;62(5):751–9.PubMedCrossRef
44.
Zurück zum Zitat Qian ZR, Rubinson DA, Nowak JA, Morales-Oyarvide V, Dunne RF, Kozak MM, et al. Association of Alterations in Main Driver Genes With Outcomes of Patients With Resected Pancreatic Ductal Adenocarcinoma. JAMA Oncol. 2018;4(3):e173420.PubMedCrossRef Qian ZR, Rubinson DA, Nowak JA, Morales-Oyarvide V, Dunne RF, Kozak MM, et al. Association of Alterations in Main Driver Genes With Outcomes of Patients With Resected Pancreatic Ductal Adenocarcinoma. JAMA Oncol. 2018;4(3):e173420.PubMedCrossRef
45.
Zurück zum Zitat Ako S, Nouso K, Kinugasa H, Dohi C, Matushita H, Mizukawa S, et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology. 2017;17(2):285–90.PubMedCrossRef Ako S, Nouso K, Kinugasa H, Dohi C, Matushita H, Mizukawa S, et al. Utility of serum DNA as a marker for KRAS mutations in pancreatic cancer tissue. Pancreatology. 2017;17(2):285–90.PubMedCrossRef
46.
Zurück zum Zitat Cheng H, Liu C, Jiang J, Luo G, Lu Y, Jin K, et al. Analysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patients. Int J Cancer. 2017;140(10):2344–50.PubMedCrossRef Cheng H, Liu C, Jiang J, Luo G, Lu Y, Jin K, et al. Analysis of ctDNA to predict prognosis and monitor treatment responses in metastatic pancreatic cancer patients. Int J Cancer. 2017;140(10):2344–50.PubMedCrossRef
47.
Zurück zum Zitat Cheng H, Luo G, Jin K, Fan Z, Huang Q, Gong Y, et al. Kras mutation correlating with circulating regulatory T cells predicts the prognosis of advanced pancreatic cancer patients. Cancer Med. 2020;9(6):2153–9.PubMedPubMedCentralCrossRef Cheng H, Luo G, Jin K, Fan Z, Huang Q, Gong Y, et al. Kras mutation correlating with circulating regulatory T cells predicts the prognosis of advanced pancreatic cancer patients. Cancer Med. 2020;9(6):2153–9.PubMedPubMedCentralCrossRef
48.
Zurück zum Zitat Haas M, Ormanns S, Baechmann S, Remold A, Kruger S, Westphalen CB, et al. Extended RAS analysis and correlation with overall survival in advanced pancreatic cancer. Br J Cancer. 2017;116(11):1462–9.PubMedPubMedCentralCrossRef Haas M, Ormanns S, Baechmann S, Remold A, Kruger S, Westphalen CB, et al. Extended RAS analysis and correlation with overall survival in advanced pancreatic cancer. Br J Cancer. 2017;116(11):1462–9.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Schultheis B, Reuter D, Ebert MP, Siveke J, Kerkhoff A, Berdel WE, et al. Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first-line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: a multicenter, randomized phase IIb study. Ann Oncol. 2017;28(10):2429–35.PubMedCrossRef Schultheis B, Reuter D, Ebert MP, Siveke J, Kerkhoff A, Berdel WE, et al. Gemcitabine combined with the monoclonal antibody nimotuzumab is an active first-line regimen in KRAS wildtype patients with locally advanced or metastatic pancreatic cancer: a multicenter, randomized phase IIb study. Ann Oncol. 2017;28(10):2429–35.PubMedCrossRef
50.
Zurück zum Zitat Boeck S, Jung A, Laubender RP, Neumann J, Egg R, Goritschan C, et al. KRAS mutation status is not predictive for objective response to anti-EGFR treatment with erlotinib in patients with advanced pancreatic cancer. J Gastroenterol. 2013;48(4):544–8.PubMedCrossRef Boeck S, Jung A, Laubender RP, Neumann J, Egg R, Goritschan C, et al. KRAS mutation status is not predictive for objective response to anti-EGFR treatment with erlotinib in patients with advanced pancreatic cancer. J Gastroenterol. 2013;48(4):544–8.PubMedCrossRef
51.
Zurück zum Zitat Perets R, Greenberg O, Shentzer T, Semenisty V, Epelbaum R, Bick T, et al. Mutant KRAS Circulating Tumor DNA Is an Accurate Tool for Pancreatic Cancer Monitoring. Oncologist. 2018;23(5):566–72.PubMedPubMedCentralCrossRef Perets R, Greenberg O, Shentzer T, Semenisty V, Epelbaum R, Bick T, et al. Mutant KRAS Circulating Tumor DNA Is an Accurate Tool for Pancreatic Cancer Monitoring. Oncologist. 2018;23(5):566–72.PubMedPubMedCentralCrossRef
52.
Zurück zum Zitat Del Re M, Vivaldi C, Rofi E, Vasile E, Miccoli M, Caparello C, et al. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci Rep. 2017;7(1):7931.PubMedPubMedCentralCrossRef Del Re M, Vivaldi C, Rofi E, Vasile E, Miccoli M, Caparello C, et al. Early changes in plasma DNA levels of mutant KRAS as a sensitive marker of response to chemotherapy in pancreatic cancer. Sci Rep. 2017;7(1):7931.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Watanabe F, Suzuki K, Tamaki S, Abe I, Endo Y, Takayama Y, et al. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PLoS One. 2019;14(12):e0227366.PubMedPubMedCentralCrossRef Watanabe F, Suzuki K, Tamaki S, Abe I, Endo Y, Takayama Y, et al. Longitudinal monitoring of KRAS-mutated circulating tumor DNA enables the prediction of prognosis and therapeutic responses in patients with pancreatic cancer. PLoS One. 2019;14(12):e0227366.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Ogura T, Yamao K, Hara K, Mizuno N, Hijioka S, Imaoka H, et al. Prognostic value of K-ras mutation status and subtypes in endoscopic ultrasound-guided fine-needle aspiration specimens from patients with unresectable pancreatic cancer. J Gastroenterol. 2013;48(5):640–6.PubMedCrossRef Ogura T, Yamao K, Hara K, Mizuno N, Hijioka S, Imaoka H, et al. Prognostic value of K-ras mutation status and subtypes in endoscopic ultrasound-guided fine-needle aspiration specimens from patients with unresectable pancreatic cancer. J Gastroenterol. 2013;48(5):640–6.PubMedCrossRef
55.
Zurück zum Zitat Cerottini JP, Caplin S, Saraga E, Givel JC, Benhattar J. The type of K-ras mutation determines prognosis in colorectal cancer. Am J Surg. 1998;175(3):198–202.PubMedCrossRef Cerottini JP, Caplin S, Saraga E, Givel JC, Benhattar J. The type of K-ras mutation determines prognosis in colorectal cancer. Am J Surg. 1998;175(3):198–202.PubMedCrossRef
56.
Zurück zum Zitat Yoon HH, Tougeron D, Shi Q, Alberts SR, Mahoney MR, Nelson GD, et al. KRAS codon 12 and 13 mutations in relation to disease-free survival in BRAF-wild-type stage III colon cancers from an adjuvant chemotherapy trial (N0147 alliance). Clin Cancer Res. 2014;20(11):3033–43.PubMedPubMedCentralCrossRef Yoon HH, Tougeron D, Shi Q, Alberts SR, Mahoney MR, Nelson GD, et al. KRAS codon 12 and 13 mutations in relation to disease-free survival in BRAF-wild-type stage III colon cancers from an adjuvant chemotherapy trial (N0147 alliance). Clin Cancer Res. 2014;20(11):3033–43.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Taieb J, Zaanan A, Le Malicot K, Julie C, Blons H, Mineur L, et al. Prognostic Effect of BRAF and KRAS Mutations in Patients With Stage III Colon Cancer Treated With Leucovorin, Fluorouracil, and Oxaliplatin With or Without Cetuximab: A Post Hoc Analysis of the PETACC-8 Trial. JAMA Oncol. 2016;2(5):643–53.PubMedCrossRef Taieb J, Zaanan A, Le Malicot K, Julie C, Blons H, Mineur L, et al. Prognostic Effect of BRAF and KRAS Mutations in Patients With Stage III Colon Cancer Treated With Leucovorin, Fluorouracil, and Oxaliplatin With or Without Cetuximab: A Post Hoc Analysis of the PETACC-8 Trial. JAMA Oncol. 2016;2(5):643–53.PubMedCrossRef
58.
Zurück zum Zitat Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter "RASCAL" study. J Natl Cancer Inst. 1998;90(9):675–84.PubMedCrossRef Andreyev HJ, Norman AR, Cunningham D, Oates JR, Clarke PA. Kirsten ras mutations in patients with colorectal cancer: the multicenter "RASCAL" study. J Natl Cancer Inst. 1998;90(9):675–84.PubMedCrossRef
59.
Zurück zum Zitat Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28(3):466–74.PubMedCrossRef Roth AD, Tejpar S, Delorenzi M, Yan P, Fiocca R, Klingbiel D, et al. Prognostic role of KRAS and BRAF in stage II and III resected colon cancer: results of the translational study on the PETACC-3, EORTC 40993, SAKK 60-00 trial. J Clin Oncol. 2010;28(3):466–74.PubMedCrossRef
60.
Zurück zum Zitat Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(10):1626–34.PubMedCrossRef Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, Freeman DJ, et al. Wild-type KRAS is required for panitumumab efficacy in patients with metastatic colorectal cancer. J Clin Oncol. 2008;26(10):1626–34.PubMedCrossRef
61.
Zurück zum Zitat De Roock W, Piessevaux H, De Schutter J, Janssens M, De Hertogh G, Personeni N, et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann Oncol. 2008;19(3):508–15.PubMedCrossRef De Roock W, Piessevaux H, De Schutter J, Janssens M, De Hertogh G, Personeni N, et al. KRAS wild-type state predicts survival and is associated to early radiological response in metastatic colorectal cancer treated with cetuximab. Ann Oncol. 2008;19(3):508–15.PubMedCrossRef
62.
Zurück zum Zitat Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359(17):1757–65.PubMedCrossRef Karapetis CS, Khambata-Ford S, Jonker DJ, O'Callaghan CJ, Tu D, Tebbutt NC, et al. K-ras mutations and benefit from cetuximab in advanced colorectal cancer. N Engl J Med. 2008;359(17):1757–65.PubMedCrossRef
63.
Zurück zum Zitat Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369(11):1023–34.PubMedCrossRef Douillard JY, Oliner KS, Siena S, Tabernero J, Burkes R, Barugel M, et al. Panitumumab-FOLFOX4 treatment and RAS mutations in colorectal cancer. N Engl J Med. 2013;369(11):1023–34.PubMedCrossRef
64.
Zurück zum Zitat Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30(29):3570–7.PubMedCrossRef Tejpar S, Celik I, Schlichting M, Sartorius U, Bokemeyer C, Van Cutsem E. Association of KRAS G13D tumor mutations with outcome in patients with metastatic colorectal cancer treated with first-line chemotherapy with or without cetuximab. J Clin Oncol. 2012;30(29):3570–7.PubMedCrossRef
65.
Zurück zum Zitat De Roock W, Jonker DJ, Di Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, et al. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA. 2010;304(16):1812–20.PubMedCrossRef De Roock W, Jonker DJ, Di Nicolantonio F, Sartore-Bianchi A, Tu D, Siena S, et al. Association of KRAS p.G13D mutation with outcome in patients with chemotherapy-refractory metastatic colorectal cancer treated with cetuximab. JAMA. 2010;304(16):1812–20.PubMedCrossRef
66.
Zurück zum Zitat Liao W, Overman MJ, Boutin AT, Shang X, Zhao D, Dey P, et al. KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer. Cancer Cell. 2019;35(4):559–72 e7.PubMedPubMedCentralCrossRef Liao W, Overman MJ, Boutin AT, Shang X, Zhao D, Dey P, et al. KRAS-IRF2 Axis Drives Immune Suppression and Immune Therapy Resistance in Colorectal Cancer. Cancer Cell. 2019;35(4):559–72 e7.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. 2003;4(5):373–84.PubMedCrossRef Hancock JF. Ras proteins: different signals from different locations. Nat Rev Mol Cell Biol. 2003;4(5):373–84.PubMedCrossRef
69.
Zurück zum Zitat Cherfils J, Zeghouf M. Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev. 2013;93(1):269–309.PubMedCrossRef Cherfils J, Zeghouf M. Regulation of small GTPases by GEFs, GAPs, and GDIs. Physiol Rev. 2013;93(1):269–309.PubMedCrossRef
70.
Zurück zum Zitat Xu GF, O'Connell P, Viskochil D, Cawthon R, Robertson M, Culver M, et al. The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell. 1990;62(3):599–608.PubMedCrossRef Xu GF, O'Connell P, Viskochil D, Cawthon R, Robertson M, Culver M, et al. The neurofibromatosis type 1 gene encodes a protein related to GAP. Cell. 1990;62(3):599–608.PubMedCrossRef
71.
Zurück zum Zitat Trahey M, McCormick F. A cytoplasmic protein stimulates normal N-ras p21 GTPase, but does not affect oncogenic mutants. Science. 1987;238(4826):542–5.PubMedCrossRef Trahey M, McCormick F. A cytoplasmic protein stimulates normal N-ras p21 GTPase, but does not affect oncogenic mutants. Science. 1987;238(4826):542–5.PubMedCrossRef
72.
Zurück zum Zitat Bonfini L, Karlovich CA, Dasgupta C, Banerjee U. The Son of sevenless gene product: a putative activator of Ras. Science. 1992;255(5044):603–6.PubMedCrossRef Bonfini L, Karlovich CA, Dasgupta C, Banerjee U. The Son of sevenless gene product: a putative activator of Ras. Science. 1992;255(5044):603–6.PubMedCrossRef
73.
Zurück zum Zitat Buday L, Downward J. Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor. Cell. 1993;73(3):611–20.PubMedCrossRef Buday L, Downward J. Epidermal growth factor regulates p21ras through the formation of a complex of receptor, Grb2 adapter protein, and Sos nucleotide exchange factor. Cell. 1993;73(3):611–20.PubMedCrossRef
74.
Zurück zum Zitat Ebinu JO, Bottorff DA, Chan EY, Stang SL, Dunn RJ, Stone JC. RasGRP, a Ras guanyl nucleotide- releasing protein with calcium- and diacylglycerol-binding motifs. Science. 1998;280(5366):1082–6.PubMedCrossRef Ebinu JO, Bottorff DA, Chan EY, Stang SL, Dunn RJ, Stone JC. RasGRP, a Ras guanyl nucleotide- releasing protein with calcium- and diacylglycerol-binding motifs. Science. 1998;280(5366):1082–6.PubMedCrossRef
76.
Zurück zum Zitat Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol Cancer Res. 2015;13(9):1325–35.PubMedCrossRef Hunter JC, Manandhar A, Carrasco MA, Gurbani D, Gondi S, Westover KD. Biochemical and Structural Analysis of Common Cancer-Associated KRAS Mutations. Mol Cancer Res. 2015;13(9):1325–35.PubMedCrossRef
77.
Zurück zum Zitat Egan SE, Giddings BW, Brooks MW, Buday L, Sizeland AM, Weinberg RA. Association of Sos Ras exchange protein with Grb2 is implicated in tyrosine kinase signal transduction and transformation. Nature. 1993;363(6424):45–51.PubMedCrossRef Egan SE, Giddings BW, Brooks MW, Buday L, Sizeland AM, Weinberg RA. Association of Sos Ras exchange protein with Grb2 is implicated in tyrosine kinase signal transduction and transformation. Nature. 1993;363(6424):45–51.PubMedCrossRef
78.
Zurück zum Zitat Gale NW, Kaplan S, Lowenstein EJ, Schlessinger J, Bar-Sagi D. Grb2 mediates the EGF-dependent activation of guanine nucleotide exchange on Ras. Nature. 1993;363(6424):88–92.PubMedCrossRef Gale NW, Kaplan S, Lowenstein EJ, Schlessinger J, Bar-Sagi D. Grb2 mediates the EGF-dependent activation of guanine nucleotide exchange on Ras. Nature. 1993;363(6424):88–92.PubMedCrossRef
79.
Zurück zum Zitat Hofmann MH, Gmachl M, Ramharter J, Savarese F, Gerlach D, Marszalek JR, et al. BI-3406, a Potent and Selective SOS1-KRAS Interaction Inhibitor, Is Effective in KRAS-Driven Cancers through Combined MEK Inhibition. Cancer Discov. 2021;11(1):142–57.PubMedCrossRef Hofmann MH, Gmachl M, Ramharter J, Savarese F, Gerlach D, Marszalek JR, et al. BI-3406, a Potent and Selective SOS1-KRAS Interaction Inhibitor, Is Effective in KRAS-Driven Cancers through Combined MEK Inhibition. Cancer Discov. 2021;11(1):142–57.PubMedCrossRef
80.
Zurück zum Zitat Mainardi S, Mulero-Sanchez A, Prahallad A, Germano G, Bosma A, Krimpenfort P, et al. SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat Med. 2018;24(7):961–7.PubMedCrossRef Mainardi S, Mulero-Sanchez A, Prahallad A, Germano G, Bosma A, Krimpenfort P, et al. SHP2 is required for growth of KRAS-mutant non-small-cell lung cancer in vivo. Nat Med. 2018;24(7):961–7.PubMedCrossRef
81.
Zurück zum Zitat Ruess DA, Heynen GJ, Ciecielski KJ, Ai J, Berninger A, Kabacaoglu D, et al. Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nat Med. 2018;24(7):954–60.PubMedCrossRef Ruess DA, Heynen GJ, Ciecielski KJ, Ai J, Berninger A, Kabacaoglu D, et al. Mutant KRAS-driven cancers depend on PTPN11/SHP2 phosphatase. Nat Med. 2018;24(7):954–60.PubMedCrossRef
82.
Zurück zum Zitat Wong GS, Zhou J, Liu JB, Wu Z, Xu X, Li T, et al. Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition. Nat Med. 2018;24(7):968–77.PubMedPubMedCentralCrossRef Wong GS, Zhou J, Liu JB, Wu Z, Xu X, Li T, et al. Targeting wild-type KRAS-amplified gastroesophageal cancer through combined MEK and SHP2 inhibition. Nat Med. 2018;24(7):968–77.PubMedPubMedCentralCrossRef
83.
Zurück zum Zitat Hillig RC, Sautier B, Schroeder J, Moosmayer D, Hilpmann A, Stegmann CM, et al. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci U S A. 2019;116(7):2551–60.PubMedPubMedCentralCrossRef Hillig RC, Sautier B, Schroeder J, Moosmayer D, Hilpmann A, Stegmann CM, et al. Discovery of potent SOS1 inhibitors that block RAS activation via disruption of the RAS-SOS1 interaction. Proc Natl Acad Sci U S A. 2019;116(7):2551–60.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Lin CC, Wieteska L, Suen KM, Kalverda AP, Ahmed Z, Ladbury JE. Grb2 binding induces phosphorylation-independent activation of Shp2. Commun Biol. 2021;4(1):437.PubMedPubMedCentralCrossRef Lin CC, Wieteska L, Suen KM, Kalverda AP, Ahmed Z, Ladbury JE. Grb2 binding induces phosphorylation-independent activation of Shp2. Commun Biol. 2021;4(1):437.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Sigismund S, Avanzato D, Lanzetti L. Emerging functions of the EGFR in cancer. Mol Oncol. 2018;12(1):3–20.PubMedCrossRef Sigismund S, Avanzato D, Lanzetti L. Emerging functions of the EGFR in cancer. Mol Oncol. 2018;12(1):3–20.PubMedCrossRef
86.
Zurück zum Zitat Koch WJ, Hawes BE, Allen LF, Lefkowitz RJ. Direct evidence that Gi-coupled receptor stimulation of mitogen-activated protein kinase is mediated by G beta gamma activation of p21ras. Proc Natl Acad Sci U S A. 1994;91(26):12706–10.PubMedPubMedCentralCrossRef Koch WJ, Hawes BE, Allen LF, Lefkowitz RJ. Direct evidence that Gi-coupled receptor stimulation of mitogen-activated protein kinase is mediated by G beta gamma activation of p21ras. Proc Natl Acad Sci U S A. 1994;91(26):12706–10.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Wan Y, Kurosaki T, Huang XY. Tyrosine kinases in activation of the MAP kinase cascade by G-protein-coupled receptors. Nature. 1996;380(6574):541–4.PubMedCrossRef Wan Y, Kurosaki T, Huang XY. Tyrosine kinases in activation of the MAP kinase cascade by G-protein-coupled receptors. Nature. 1996;380(6574):541–4.PubMedCrossRef
89.
Zurück zum Zitat Braicu C, Buse M, Busuioc C, Drula R, Gulei D, Raduly L, et al. A Comprehensive Review on MAPK: A Promising Therapeutic Target in Cancer. Cancers (Basel). 2019;11(10):1618. Braicu C, Buse M, Busuioc C, Drula R, Gulei D, Raduly L, et al. A Comprehensive Review on MAPK: A Promising Therapeutic Target in Cancer. Cancers (Basel). 2019;11(10):1618.
90.
Zurück zum Zitat Uprety D, Adjei AA. KRAS: From undruggable to a druggable Cancer Target. Cancer Treat Rev. 2020;89:102070. Uprety D, Adjei AA. KRAS: From undruggable to a druggable Cancer Target. Cancer Treat Rev. 2020;89:102070.
91.
Zurück zum Zitat Hu Y, Lu W, Chen G, Wang P, Chen Z, Zhou Y, et al. K-ras(G12V) transformation leads to mitochondrial dysfunction and a metabolic switch from oxidative phosphorylation to glycolysis. Cell Res. 2012;22(2):399–412.PubMedCrossRef Hu Y, Lu W, Chen G, Wang P, Chen Z, Zhou Y, et al. K-ras(G12V) transformation leads to mitochondrial dysfunction and a metabolic switch from oxidative phosphorylation to glycolysis. Cell Res. 2012;22(2):399–412.PubMedCrossRef
92.
Zurück zum Zitat Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-Dependent Ferroptosis Drives Tumor-Associated Macrophage Polarization via Release and Uptake of Oncogenic KRAS Protein. Autophagy. 2020;16(11):2069–83. Dai E, Han L, Liu J, Xie Y, Kroemer G, Klionsky DJ, et al. Autophagy-Dependent Ferroptosis Drives Tumor-Associated Macrophage Polarization via Release and Uptake of Oncogenic KRAS Protein. Autophagy. 2020;16(11):2069–83.
93.
Zurück zum Zitat Carriere C, Young AL, Gunn JR, Longnecker DS, Korc M. Acute pancreatitis markedly accelerates pancreatic cancer progression in mice expressing oncogenic Kras. Biochem Biophys Res Commun. 2009;382(3):561–5.PubMedPubMedCentralCrossRef Carriere C, Young AL, Gunn JR, Longnecker DS, Korc M. Acute pancreatitis markedly accelerates pancreatic cancer progression in mice expressing oncogenic Kras. Biochem Biophys Res Commun. 2009;382(3):561–5.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 2013;145(6):1449–58.PubMedCrossRef Philip B, Roland CL, Daniluk J, Liu Y, Chatterjee D, Gomez SB, et al. A high-fat diet activates oncogenic Kras and COX2 to induce development of pancreatic ductal adenocarcinoma in mice. Gastroenterology. 2013;145(6):1449–58.PubMedCrossRef
95.
Zurück zum Zitat Dai E, Han L, Liu J, Xie Y, Zeh HJ, Kang R, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun. 2020;11(1):6339.PubMedPubMedCentralCrossRef Dai E, Han L, Liu J, Xie Y, Zeh HJ, Kang R, et al. Ferroptotic damage promotes pancreatic tumorigenesis through a TMEM173/STING-dependent DNA sensor pathway. Nat Commun. 2020;11(1):6339.PubMedPubMedCentralCrossRef
96.
Zurück zum Zitat Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, et al. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Sci Transl Med. 2017;9(394):eaal5253. Ross SJ, Revenko AS, Hanson LL, Ellston R, Staniszewska A, Whalley N, et al. Targeting KRAS-dependent tumors with AZD4785, a high-affinity therapeutic antisense oligonucleotide inhibitor of KRAS. Sci Transl Med. 2017;9(394):eaal5253.
97.
98.
Zurück zum Zitat Golan T, Khvalevsky EZ, Hubert A, Gabai RM, Hen N, Segal A, et al. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget. 2015;6(27):24560–70.PubMedPubMedCentralCrossRef Golan T, Khvalevsky EZ, Hubert A, Gabai RM, Hen N, Segal A, et al. RNAi therapy targeting KRAS in combination with chemotherapy for locally advanced pancreatic cancer patients. Oncotarget. 2015;6(27):24560–70.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. J Clin Oncol. 2003;21(7):1301–6.PubMedCrossRef Cohen SJ, Ho L, Ranganathan S, Abbruzzese JL, Alpaugh RK, Beard M, et al. Phase II and pharmacodynamic study of the farnesyltransferase inhibitor R115777 as initial therapy in patients with metastatic pancreatic adenocarcinoma. J Clin Oncol. 2003;21(7):1301–6.PubMedCrossRef
101.
Zurück zum Zitat Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22(8):1430–8.PubMedCrossRef Van Cutsem E, van de Velde H, Karasek P, Oettle H, Vervenne WL, Szawlowski A, et al. Phase III trial of gemcitabine plus tipifarnib compared with gemcitabine plus placebo in advanced pancreatic cancer. J Clin Oncol. 2004;22(8):1430–8.PubMedCrossRef
102.
Zurück zum Zitat Chow LQ, Eckhardt SG, O'Bryant CL, Schultz MK, Morrow M, Grolnic S, et al. A phase I safety, pharmacological, and biological study of the farnesyl protein transferase inhibitor, lonafarnib (SCH 663366), in combination with cisplatin and gemcitabine in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2008;62(4):631–46.PubMedCrossRef Chow LQ, Eckhardt SG, O'Bryant CL, Schultz MK, Morrow M, Grolnic S, et al. A phase I safety, pharmacological, and biological study of the farnesyl protein transferase inhibitor, lonafarnib (SCH 663366), in combination with cisplatin and gemcitabine in patients with advanced solid tumors. Cancer Chemother Pharmacol. 2008;62(4):631–46.PubMedCrossRef
103.
Zurück zum Zitat Sharma S, Kemeny N, Kelsen DP, Ilson D, O'Reilly E, Zaknoen S, et al. A phase II trial of farnesyl protein transferase inhibitor SCH 66336, given by twice-daily oral administration, in patients with metastatic colorectal cancer refractory to 5-fluorouracil and irinotecan. Ann Oncol. 2002;13(7):1067–71.PubMedCrossRef Sharma S, Kemeny N, Kelsen DP, Ilson D, O'Reilly E, Zaknoen S, et al. A phase II trial of farnesyl protein transferase inhibitor SCH 66336, given by twice-daily oral administration, in patients with metastatic colorectal cancer refractory to 5-fluorouracil and irinotecan. Ann Oncol. 2002;13(7):1067–71.PubMedCrossRef
104.
Zurück zum Zitat Whyte DB, Kirschmeier P, Hockenberry TN, Nunez-Oliva I, James L, Catino JJ, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 1997;272(22):14459–64.PubMedCrossRef Whyte DB, Kirschmeier P, Hockenberry TN, Nunez-Oliva I, James L, Catino JJ, et al. K- and N-Ras are geranylgeranylated in cells treated with farnesyl protein transferase inhibitors. J Biol Chem. 1997;272(22):14459–64.PubMedCrossRef
105.
Zurück zum Zitat Kazi A, Xiang S, Yang H, Chen L, Kennedy P, Ayaz M, et al. Dual Farnesyl and Geranylgeranyl Transferase Inhibitor Thwarts Mutant KRAS-Driven Patient-Derived Pancreatic Tumors. Clin Cancer Res. 2019;25(19):5984–96.PubMedPubMedCentralCrossRef Kazi A, Xiang S, Yang H, Chen L, Kennedy P, Ayaz M, et al. Dual Farnesyl and Geranylgeranyl Transferase Inhibitor Thwarts Mutant KRAS-Driven Patient-Derived Pancreatic Tumors. Clin Cancer Res. 2019;25(19):5984–96.PubMedPubMedCentralCrossRef
106.
Zurück zum Zitat Manu KA, Chai TF, Teh JT, Zhu WL, Casey PJ, Wang M. Inhibition of Isoprenylcysteine Carboxylmethyltransferase Induces Cell-Cycle Arrest and Apoptosis through p21 and p21-Regulated BNIP3 Induction in Pancreatic Cancer. Mol Cancer Ther. 2017;16(5):914–23.PubMedCrossRef Manu KA, Chai TF, Teh JT, Zhu WL, Casey PJ, Wang M. Inhibition of Isoprenylcysteine Carboxylmethyltransferase Induces Cell-Cycle Arrest and Apoptosis through p21 and p21-Regulated BNIP3 Induction in Pancreatic Cancer. Mol Cancer Ther. 2017;16(5):914–23.PubMedCrossRef
107.
Zurück zum Zitat Spencer-Smith R, Koide A, Zhou Y, Eguchi RR, Sha F, Gajwani P, et al. Inhibition of RAS function through targeting an allosteric regulatory site. Nat Chem Biol. 2017;13(1):62–8.PubMedCrossRef Spencer-Smith R, Koide A, Zhou Y, Eguchi RR, Sha F, Gajwani P, et al. Inhibition of RAS function through targeting an allosteric regulatory site. Nat Chem Biol. 2017;13(1):62–8.PubMedCrossRef
108.
Zurück zum Zitat Ayati A, Moghimi S, Salarinejad S, Safavi M, Pouramiri B, Foroumadi A. A review on progression of epidermal growth factor receptor (EGFR) inhibitors as an efficient approach in cancer targeted therapy. Bioorg Chem. 2020;99:103811.PubMedCrossRef Ayati A, Moghimi S, Salarinejad S, Safavi M, Pouramiri B, Foroumadi A. A review on progression of epidermal growth factor receptor (EGFR) inhibitors as an efficient approach in cancer targeted therapy. Bioorg Chem. 2020;99:103811.PubMedCrossRef
109.
Zurück zum Zitat Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005;352(8):786–92.PubMedCrossRef Kobayashi S, Boggon TJ, Dayaram T, Janne PA, Kocher O, Meyerson M, et al. EGFR mutation and resistance of non-small-cell lung cancer to gefitinib. N Engl J Med. 2005;352(8):786–92.PubMedCrossRef
110.
Zurück zum Zitat Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, 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. 2007;25(15):1960–6.PubMedCrossRef Moore MJ, Goldstein D, Hamm J, Figer A, Hecht JR, Gallinger S, 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. 2007;25(15):1960–6.PubMedCrossRef
111.
Zurück zum Zitat Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532–6.PubMedPubMedCentralCrossRef Misale S, Yaeger R, Hobor S, Scala E, Janakiraman M, Liska D, et al. Emergence of KRAS mutations and acquired resistance to anti-EGFR therapy in colorectal cancer. Nature. 2012;486(7404):532–6.PubMedPubMedCentralCrossRef
112.
Zurück zum Zitat Cai D, Choi PS, Gelbard M, Meyerson M. Identification and Characterization of Oncogenic SOS1 Mutations in Lung Adenocarcinoma. Mol Cancer Res. 2019;17(4):1002–12.PubMedPubMedCentralCrossRef Cai D, Choi PS, Gelbard M, Meyerson M. Identification and Characterization of Oncogenic SOS1 Mutations in Lung Adenocarcinoma. Mol Cancer Res. 2019;17(4):1002–12.PubMedPubMedCentralCrossRef
113.
Zurück zum Zitat Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535(7610):148–52.PubMedCrossRef Chen YN, LaMarche MJ, Chan HM, Fekkes P, Garcia-Fortanet J, Acker MG, et al. Allosteric inhibition of SHP2 phosphatase inhibits cancers driven by receptor tyrosine kinases. Nature. 2016;535(7610):148–52.PubMedCrossRef
114.
Zurück zum Zitat Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–17.PubMedPubMedCentralCrossRef Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med. 2012;209(6):1201–17.PubMedPubMedCentralCrossRef
115.
Zurück zum Zitat LaMarche MJ, Acker M, Argintaru A, Bauer D, Boisclair J, Chan H, et al. Identification of TNO155, an Allosteric SHP2 Inhibitor for the Treatment of Cancer. J Med Chem. 2020;63(22):13578–94.PubMedCrossRef LaMarche MJ, Acker M, Argintaru A, Bauer D, Boisclair J, Chan H, et al. Identification of TNO155, an Allosteric SHP2 Inhibitor for the Treatment of Cancer. J Med Chem. 2020;63(22):13578–94.PubMedCrossRef
116.
Zurück zum Zitat Liu C, Lu H, Wang H, Loo A, Zhang X, Yang G, et al. Combinations with Allosteric SHP2 Inhibitor TNO155 to Block Receptor Tyrosine Kinase Signaling. Clin Cancer Res. 2021;27(1):342–54.PubMedCrossRef Liu C, Lu H, Wang H, Loo A, Zhang X, Yang G, et al. Combinations with Allosteric SHP2 Inhibitor TNO155 to Block Receptor Tyrosine Kinase Signaling. Clin Cancer Res. 2021;27(1):342–54.PubMedCrossRef
117.
Zurück zum Zitat Yen I, Shanahan F, Lee J, Hong YS, Shin SJ, Moore AR, et al. ARAF mutations confer resistance to the RAF inhibitor belvarafenib in melanoma. Nature. 2021;594(7863):418–23.PubMedCrossRef Yen I, Shanahan F, Lee J, Hong YS, Shin SJ, Moore AR, et al. ARAF mutations confer resistance to the RAF inhibitor belvarafenib in melanoma. Nature. 2021;594(7863):418–23.PubMedCrossRef
118.
Zurück zum Zitat Park S, Kim TM, Cho SY, Kim S, Oh Y, Kim M, et al. Combined blockade of polo-like kinase and pan-RAF is effective against NRAS-mutant non-small cell lung cancer cells. Cancer Lett. 2020;495:135–44.PubMedCrossRef Park S, Kim TM, Cho SY, Kim S, Oh Y, Kim M, et al. Combined blockade of polo-like kinase and pan-RAF is effective against NRAS-mutant non-small cell lung cancer cells. Cancer Lett. 2020;495:135–44.PubMedCrossRef
119.
Zurück zum Zitat Monaco KA, Delach S, Yuan J, Mishina Y, Fordjour P, Labrot E, et al. LXH254, a Potent and Selective ARAF-Sparing Inhibitor of BRAF and CRAF for the Treatment of MAPK-Driven Tumors. Clin Cancer Res. 2021;27(7):2061–73.PubMedCrossRef Monaco KA, Delach S, Yuan J, Mishina Y, Fordjour P, Labrot E, et al. LXH254, a Potent and Selective ARAF-Sparing Inhibitor of BRAF and CRAF for the Treatment of MAPK-Driven Tumors. Clin Cancer Res. 2021;27(7):2061–73.PubMedCrossRef
120.
Zurück zum Zitat Desai J, Gan H, Barrow C, Jameson M, Atkinson V, Haydon A, et al. Phase I, Open-Label, Dose-Escalation/Dose-Expansion Study of Lifirafenib (BGB-283), an RAF Family Kinase Inhibitor, in Patients With Solid Tumors. J Clin Oncol. 2020;38(19):2140–50.PubMedPubMedCentralCrossRef Desai J, Gan H, Barrow C, Jameson M, Atkinson V, Haydon A, et al. Phase I, Open-Label, Dose-Escalation/Dose-Expansion Study of Lifirafenib (BGB-283), an RAF Family Kinase Inhibitor, in Patients With Solid Tumors. J Clin Oncol. 2020;38(19):2140–50.PubMedPubMedCentralCrossRef
121.
Zurück zum Zitat Yuan X, Tang Z, Du R, Yao Z, Cheung SH, Zhang X, et al. RAF dimer inhibition enhances the antitumor activity of MEK inhibitors in K-RAS mutant tumors. Mol Oncol. 2020;14(8):1833–49.PubMedPubMedCentralCrossRef Yuan X, Tang Z, Du R, Yao Z, Cheung SH, Zhang X, et al. RAF dimer inhibition enhances the antitumor activity of MEK inhibitors in K-RAS mutant tumors. Mol Oncol. 2020;14(8):1833–49.PubMedPubMedCentralCrossRef
122.
Zurück zum Zitat Janne PA, van den Heuvel MM, Barlesi F, Cobo M, Mazieres J, Crino L, et al. Selumetinib Plus Docetaxel Compared With Docetaxel Alone and Progression-Free Survival in Patients With KRAS-Mutant Advanced Non-Small Cell Lung Cancer: The SELECT-1 Randomized Clinical Trial. JAMA. 2017;317(18):1844–53.PubMedPubMedCentralCrossRef Janne PA, van den Heuvel MM, Barlesi F, Cobo M, Mazieres J, Crino L, et al. Selumetinib Plus Docetaxel Compared With Docetaxel Alone and Progression-Free Survival in Patients With KRAS-Mutant Advanced Non-Small Cell Lung Cancer: The SELECT-1 Randomized Clinical Trial. JAMA. 2017;317(18):1844–53.PubMedPubMedCentralCrossRef
123.
Zurück zum Zitat Blumenschein GR Jr, Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)dagger. Ann Oncol. 2015;26(5):894–901.PubMedPubMedCentralCrossRef Blumenschein GR Jr, Smit EF, Planchard D, Kim DW, Cadranel J, De Pas T, et al. A randomized phase II study of the MEK1/MEK2 inhibitor trametinib (GSK1120212) compared with docetaxel in KRAS-mutant advanced non-small-cell lung cancer (NSCLC)dagger. Ann Oncol. 2015;26(5):894–901.PubMedPubMedCentralCrossRef
124.
Zurück zum Zitat Bodoky G, Timcheva C, Spigel DR, La Stella PJ, Ciuleanu TE, Pover G, et al. 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. Investig New Drugs. 2012;30(3):1216–23.CrossRef Bodoky G, Timcheva C, Spigel DR, La Stella PJ, Ciuleanu TE, Pover G, et al. 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. Investig New Drugs. 2012;30(3):1216–23.CrossRef
125.
Zurück zum Zitat Bennouna J, Lang I, Valladares-Ayerbes M, Boer K, Adenis A, Escudero P, et al. A Phase II, open-label, randomised study to assess the efficacy and safety of the MEK1/2 inhibitor AZD6244 (ARRY-142886) versus capecitabine monotherapy in patients with colorectal cancer who have failed one or two prior chemotherapeutic regimens. Investig New Drugs. 2011;29(5):1021–8.CrossRef Bennouna J, Lang I, Valladares-Ayerbes M, Boer K, Adenis A, Escudero P, et al. A Phase II, open-label, randomised study to assess the efficacy and safety of the MEK1/2 inhibitor AZD6244 (ARRY-142886) versus capecitabine monotherapy in patients with colorectal cancer who have failed one or two prior chemotherapeutic regimens. Investig New Drugs. 2011;29(5):1021–8.CrossRef
126.
Zurück zum Zitat Yen I, Shanahan F, Merchant M, Orr C, Hunsaker T, Durk M, et al. Pharmacological Induction of RAS-GTP Confers RAF Inhibitor Sensitivity in KRAS Mutant Tumors. Cancer Cell. 2018;34(4):611–25 e7.PubMedCrossRef Yen I, Shanahan F, Merchant M, Orr C, Hunsaker T, Durk M, et al. Pharmacological Induction of RAS-GTP Confers RAF Inhibitor Sensitivity in KRAS Mutant Tumors. Cancer Cell. 2018;34(4):611–25 e7.PubMedCrossRef
127.
Zurück zum Zitat Lebbe C, Italiano A, Houede N, Awada A, Aftimos P, Lesimple T, et al. Selective Oral MEK1/2 Inhibitor Pimasertib in Metastatic Melanoma: Antitumor Activity in a Phase I, Dose-Escalation Trial. Target Oncol. 2021;16(1):47–57.PubMedCrossRef Lebbe C, Italiano A, Houede N, Awada A, Aftimos P, Lesimple T, et al. Selective Oral MEK1/2 Inhibitor Pimasertib in Metastatic Melanoma: Antitumor Activity in a Phase I, Dose-Escalation Trial. Target Oncol. 2021;16(1):47–57.PubMedCrossRef
128.
Zurück zum Zitat Lebbe C, Dutriaux C, Lesimple T, Kruit W, Kerger J, Thomas L, et al. Pimasertib Versus Dacarbazine in Patients With Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers (Basel). 2020;12(7):1727. Lebbe C, Dutriaux C, Lesimple T, Kruit W, Kerger J, Thomas L, et al. Pimasertib Versus Dacarbazine in Patients With Unresectable NRAS-Mutated Cutaneous Melanoma: Phase II, Randomized, Controlled Trial with Crossover. Cancers (Basel). 2020;12(7):1727.
129.
Zurück zum Zitat de Weger VA, de Jonge M, Langenberg MHG, Schellens JHM, Lolkema M, Varga A, et al. A phase I study of the HDM2 antagonist SAR405838 combined with the MEK inhibitor pimasertib in patients with advanced solid tumours. Br J Cancer. 2019;120(3):286–93.PubMedCrossRef de Weger VA, de Jonge M, Langenberg MHG, Schellens JHM, Lolkema M, Varga A, et al. A phase I study of the HDM2 antagonist SAR405838 combined with the MEK inhibitor pimasertib in patients with advanced solid tumours. Br J Cancer. 2019;120(3):286–93.PubMedCrossRef
130.
Zurück zum Zitat Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3(7):742–50.PubMedCrossRef Morris EJ, Jha S, Restaino CR, Dayananth P, Zhu H, Cooper A, et al. Discovery of a novel ERK inhibitor with activity in models of acquired resistance to BRAF and MEK inhibitors. Cancer Discov. 2013;3(7):742–50.PubMedCrossRef
131.
Zurück zum Zitat Hatzivassiliou G, Liu B, O'Brien C, Spoerke JM, Hoeflich KP, Haverty PM, et al. ERK inhibition overcomes acquired resistance to MEK inhibitors. Mol Cancer Ther. 2012;11(5):1143–54.PubMedCrossRef Hatzivassiliou G, Liu B, O'Brien C, Spoerke JM, Hoeflich KP, Haverty PM, et al. ERK inhibition overcomes acquired resistance to MEK inhibitors. Mol Cancer Ther. 2012;11(5):1143–54.PubMedCrossRef
132.
Zurück zum Zitat Ma Y, Xu R, Liu X, Zhang Y, Song L, Cai S, et al. LY3214996 relieves acquired resistance to sorafenib in hepatocellular carcinoma cells. Int J Med Sci. 2021;18(6):1456–64.PubMedPubMedCentralCrossRef Ma Y, Xu R, Liu X, Zhang Y, Song L, Cai S, et al. LY3214996 relieves acquired resistance to sorafenib in hepatocellular carcinoma cells. Int J Med Sci. 2021;18(6):1456–64.PubMedPubMedCentralCrossRef
133.
Zurück zum Zitat Sullivan RJ, Infante JR, Janku F, Wong DJL, Sosman JA, Keedy V, et al. First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK Mutant Advanced Solid Tumors: Results of a Phase I Dose-Escalation and Expansion Study. Cancer Discov. 2018;8(2):184–95.PubMedCrossRef Sullivan RJ, Infante JR, Janku F, Wong DJL, Sosman JA, Keedy V, et al. First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK Mutant Advanced Solid Tumors: Results of a Phase I Dose-Escalation and Expansion Study. Cancer Discov. 2018;8(2):184–95.PubMedCrossRef
134.
Zurück zum Zitat Blake JF, Burkard M, Chan J, Chen H, Chou KJ, Diaz D, et al. Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-y l)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development. J Med Chem. 2016;59(12):5650–60.PubMedCrossRef Blake JF, Burkard M, Chan J, Chen H, Chou KJ, Diaz D, et al. Discovery of (S)-1-(1-(4-Chloro-3-fluorophenyl)-2-hydroxyethyl)-4-(2-((1-methyl-1H-pyrazol-5-y l)amino)pyrimidin-4-yl)pyridin-2(1H)-one (GDC-0994), an Extracellular Signal-Regulated Kinase 1/2 (ERK1/2) Inhibitor in Early Clinical Development. J Med Chem. 2016;59(12):5650–60.PubMedCrossRef
135.
Zurück zum Zitat Weekes C, Lockhart A, LoRusso P, Murray E, Park E, Tagen M, et al. A Phase Ib Study to Evaluate the MEK Inhibitor Cobimetinib in Combination with the ERK1/2 Inhibitor GDC-0994 in Patients with Advanced Solid Tumors. Oncologist. 2020;25(10):833–e1438.PubMedPubMedCentralCrossRef Weekes C, Lockhart A, LoRusso P, Murray E, Park E, Tagen M, et al. A Phase Ib Study to Evaluate the MEK Inhibitor Cobimetinib in Combination with the ERK1/2 Inhibitor GDC-0994 in Patients with Advanced Solid Tumors. Oncologist. 2020;25(10):833–e1438.PubMedPubMedCentralCrossRef
136.
Zurück zum Zitat Varga A, Soria JC, Hollebecque A, LoRusso P, Bendell J, Huang SA, et al. A First-in-Human Phase I Study to Evaluate the ERK1/2 Inhibitor GDC-0994 in Patients with Advanced Solid Tumors. Clin Cancer Res. 2020;26(6):1229–36.PubMedCrossRef Varga A, Soria JC, Hollebecque A, LoRusso P, Bendell J, Huang SA, et al. A First-in-Human Phase I Study to Evaluate the ERK1/2 Inhibitor GDC-0994 in Patients with Advanced Solid Tumors. Clin Cancer Res. 2020;26(6):1229–36.PubMedCrossRef
137.
Zurück zum Zitat Moschos SJ, Sullivan RJ, Hwu WJ, Ramanathan RK, Adjei AA, Fong PC, et al. Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight. 2018;3(4):e92352. Moschos SJ, Sullivan RJ, Hwu WJ, Ramanathan RK, Adjei AA, Fong PC, et al. Development of MK-8353, an orally administered ERK1/2 inhibitor, in patients with advanced solid tumors. JCI Insight. 2018;3(4):e92352.
138.
Zurück zum Zitat Goetz EM, Ghandi M, Treacy DJ, Wagle N, Garraway LA. ERK mutations confer resistance to mitogen-activated protein kinase pathway inhibitors. Cancer Res. 2014;74(23):7079–89.PubMedPubMedCentralCrossRef Goetz EM, Ghandi M, Treacy DJ, Wagle N, Garraway LA. ERK mutations confer resistance to mitogen-activated protein kinase pathway inhibitors. Cancer Res. 2014;74(23):7079–89.PubMedPubMedCentralCrossRef
140.
Zurück zum Zitat Janku F, Lee JJ, Tsimberidou AM, Hong DS, Naing A, Falchook GS, et al. PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One. 2011;6(7):e22769.PubMedPubMedCentralCrossRef Janku F, Lee JJ, Tsimberidou AM, Hong DS, Naing A, Falchook GS, et al. PIK3CA mutations frequently coexist with RAS and BRAF mutations in patients with advanced cancers. PLoS One. 2011;6(7):e22769.PubMedPubMedCentralCrossRef
141.
Zurück zum Zitat Cisowski J, Bergo MO. What makes oncogenes mutually exclusive? Small GTPases. 2017;8(3):187–92.PubMedCrossRef Cisowski J, Bergo MO. What makes oncogenes mutually exclusive? Small GTPases. 2017;8(3):187–92.PubMedCrossRef
142.
Zurück zum Zitat Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14(12):1351–6.PubMedPubMedCentralCrossRef Engelman JA, Chen L, Tan X, Crosby K, Guimaraes AR, Upadhyay R, et al. Effective use of PI3K and MEK inhibitors to treat mutant Kras G12D and PIK3CA H1047R murine lung cancers. Nat Med. 2008;14(12):1351–6.PubMedPubMedCentralCrossRef
143.
Zurück zum Zitat Shapiro GI, LoRusso P, Kwak E, Pandya S, Rudin CM, Kurkjian C, et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Investig New Drugs. 2020;38(2):419–32.CrossRef Shapiro GI, LoRusso P, Kwak E, Pandya S, Rudin CM, Kurkjian C, et al. Phase Ib study of the MEK inhibitor cobimetinib (GDC-0973) in combination with the PI3K inhibitor pictilisib (GDC-0941) in patients with advanced solid tumors. Investig New Drugs. 2020;38(2):419–32.CrossRef
144.
Zurück zum Zitat Bedard PL, Tabernero J, Janku F, Wainberg ZA, Paz-Ares L, Vansteenkiste J, 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. 2015;21(4):730–8.PubMedCrossRef Bedard PL, Tabernero J, Janku F, Wainberg ZA, Paz-Ares L, Vansteenkiste J, 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. 2015;21(4):730–8.PubMedCrossRef
145.
Zurück zum Zitat Tolcher AW, Kurzrock R, Valero V, Gonzalez R, Heist RS, Tan AR, et al. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors. Cancer Chemother Pharmacol. 2020;85(4):673–83.PubMedCrossRef Tolcher AW, Kurzrock R, Valero V, Gonzalez R, Heist RS, Tan AR, et al. Phase I dose-escalation trial of the oral AKT inhibitor uprosertib in combination with the oral MEK1/MEK2 inhibitor trametinib in patients with solid tumors. Cancer Chemother Pharmacol. 2020;85(4):673–83.PubMedCrossRef
146.
Zurück zum Zitat Mita M, Fu S, Piha-Paul SA, Janku F, Mita A, Natale R, et al. Phase I trial of MEK 1/2 inhibitor pimasertib combined with mTOR inhibitor temsirolimus in patients with advanced solid tumors. Investig New Drugs. 2017;35(5):616–26.CrossRef Mita M, Fu S, Piha-Paul SA, Janku F, Mita A, Natale R, et al. Phase I trial of MEK 1/2 inhibitor pimasertib combined with mTOR inhibitor temsirolimus in patients with advanced solid tumors. Investig New Drugs. 2017;35(5):616–26.CrossRef
147.
Zurück zum Zitat Luo J, Emanuele MJ, Li D, Creighton CJ, Schlabach MR, Westbrook TF, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137(5):835–48.PubMedPubMedCentralCrossRef Luo J, Emanuele MJ, Li D, Creighton CJ, Schlabach MR, Westbrook TF, et al. A genome-wide RNAi screen identifies multiple synthetic lethal interactions with the Ras oncogene. Cell. 2009;137(5):835–48.PubMedPubMedCentralCrossRef
148.
Zurück zum Zitat Cullis J, Meiri D, Sandi MJ, Radulovich N, Kent OA, Medrano M, et al. The RhoGEF GEF-H1 is required for oncogenic RAS signaling via KSR-1. Cancer Cell. 2014;25(2):181–95.PubMedCrossRef Cullis J, Meiri D, Sandi MJ, Radulovich N, Kent OA, Medrano M, et al. The RhoGEF GEF-H1 is required for oncogenic RAS signaling via KSR-1. Cancer Cell. 2014;25(2):181–95.PubMedCrossRef
149.
Zurück zum Zitat Corcoran RB, Cheng KA, Hata AN, Faber AC, Ebi H, Coffee EM, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell. 2013;23(1):121–8.PubMedCrossRef Corcoran RB, Cheng KA, Hata AN, Faber AC, Ebi H, Coffee EM, et al. Synthetic lethal interaction of combined BCL-XL and MEK inhibition promotes tumor regressions in KRAS mutant cancer models. Cancer Cell. 2013;23(1):121–8.PubMedCrossRef
150.
Zurück zum Zitat Sarthy AV, Morgan-Lappe SE, Zakula D, Vernetti L, Schurdak M, Packer JC, et al. Survivin depletion preferentially reduces the survival of activated K-Ras-transformed cells. Mol Cancer Ther. 2007;6(1):269–76.PubMedCrossRef Sarthy AV, Morgan-Lappe SE, Zakula D, Vernetti L, Schurdak M, Packer JC, et al. Survivin depletion preferentially reduces the survival of activated K-Ras-transformed cells. Mol Cancer Ther. 2007;6(1):269–76.PubMedCrossRef
151.
Zurück zum Zitat Costa-Cabral S, Brough R, Konde A, Aarts M, Campbell J, Marinari E, et al. CDK1 Is a Synthetic Lethal Target for KRAS Mutant Tumours. PLoS One. 2016;11(2):e0149099.PubMedPubMedCentralCrossRef Costa-Cabral S, Brough R, Konde A, Aarts M, Campbell J, Marinari E, et al. CDK1 Is a Synthetic Lethal Target for KRAS Mutant Tumours. PLoS One. 2016;11(2):e0149099.PubMedPubMedCentralCrossRef
152.
Zurück zum Zitat Puyol M, Martin A, Dubus P, Mulero F, Pizcueta P, Khan G, et al. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010;18(1):63–73.PubMedCrossRef Puyol M, Martin A, Dubus P, Mulero F, Pizcueta P, Khan G, et al. A synthetic lethal interaction between K-Ras oncogenes and Cdk4 unveils a therapeutic strategy for non-small cell lung carcinoma. Cancer Cell. 2010;18(1):63–73.PubMedCrossRef
153.
Zurück zum Zitat Mao CQ, Xiong MH, Liu Y, Shen S, Du XJ, Yang XZ, et al. Synthetic lethal therapy for KRAS mutant non-small-cell lung carcinoma with nanoparticle-mediated CDK4 siRNA delivery. Mol Ther. 2014;22(5):964–73.PubMedPubMedCentralCrossRef Mao CQ, Xiong MH, Liu Y, Shen S, Du XJ, Yang XZ, et al. Synthetic lethal therapy for KRAS mutant non-small-cell lung carcinoma with nanoparticle-mediated CDK4 siRNA delivery. Mol Ther. 2014;22(5):964–73.PubMedPubMedCentralCrossRef
154.
Zurück zum Zitat Koundinya M, Sudhalter J, Courjaud A, Lionne B, Touyer G, Bonnet L, et al. Dependence on the Pyrimidine Biosynthetic Enzyme DHODH Is a Synthetic Lethal Vulnerability in Mutant KRAS-Driven Cancers. Cell Chem Biol. 2018;25(6):705–17 e11.PubMedCrossRef Koundinya M, Sudhalter J, Courjaud A, Lionne B, Touyer G, Bonnet L, et al. Dependence on the Pyrimidine Biosynthetic Enzyme DHODH Is a Synthetic Lethal Vulnerability in Mutant KRAS-Driven Cancers. Cell Chem Biol. 2018;25(6):705–17 e11.PubMedCrossRef
155.
Zurück zum Zitat Manchado E, Weissmueller S, Morris JP, Chen CC, Wullenkord R, Lujambio A, et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature. 2016;534(7609):647–51.PubMedPubMedCentralCrossRef Manchado E, Weissmueller S, Morris JP, Chen CC, Wullenkord R, Lujambio A, et al. A combinatorial strategy for treating KRAS-mutant lung cancer. Nature. 2016;534(7609):647–51.PubMedPubMedCentralCrossRef
156.
Zurück zum Zitat Kumar MS, Hancock DC, Molina-Arcas M, Steckel M, East P, Diefenbacher M, et al. The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer. Cell. 2012;149(3):642–55.PubMedCrossRef Kumar MS, Hancock DC, Molina-Arcas M, Steckel M, East P, Diefenbacher M, et al. The GATA2 transcriptional network is requisite for RAS oncogene-driven non-small cell lung cancer. Cell. 2012;149(3):642–55.PubMedCrossRef
157.
Zurück zum Zitat Singh A, Sweeney MF, Yu M, Burger A, Greninger P, Benes C, et al. TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers. Cell. 2012;148(4):639–50.PubMedPubMedCentralCrossRef Singh A, Sweeney MF, Yu M, Burger A, Greninger P, Benes C, et al. TAK1 inhibition promotes apoptosis in KRAS-dependent colon cancers. Cell. 2012;148(4):639–50.PubMedPubMedCentralCrossRef
158.
Zurück zum Zitat Carr RM, Vorobyev D, Lasho T, Marks DL, Tolosa EJ, Vedder A, et al. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis. Nat Commun. 2021;12(1):2901.PubMedPubMedCentralCrossRef Carr RM, Vorobyev D, Lasho T, Marks DL, Tolosa EJ, Vedder A, et al. RAS mutations drive proliferative chronic myelomonocytic leukemia via a KMT2A-PLK1 axis. Nat Commun. 2021;12(1):2901.PubMedPubMedCentralCrossRef
159.
Zurück zum Zitat Wang J, Hu K, Guo J, Cheng F, Lv J, Jiang W, et al. Suppression of KRas-mutant cancer through the combined inhibition of KRAS with PLK1 and ROCK. Nat Commun. 2016;7:11363.PubMedPubMedCentralCrossRef Wang J, Hu K, Guo J, Cheng F, Lv J, Jiang W, et al. Suppression of KRas-mutant cancer through the combined inhibition of KRAS with PLK1 and ROCK. Nat Commun. 2016;7:11363.PubMedPubMedCentralCrossRef
160.
Zurück zum Zitat Wei X, Yang J, Adair SJ, Ozturk H, Kuscu C, Lee KY, et al. Targeted CRISPR screening identifies PRMT5 as synthetic lethality combinatorial target with gemcitabine in pancreatic cancer cells. Proc Natl Acad Sci U S A. 2020;117(45):28068–79.PubMedPubMedCentralCrossRef Wei X, Yang J, Adair SJ, Ozturk H, Kuscu C, Lee KY, et al. Targeted CRISPR screening identifies PRMT5 as synthetic lethality combinatorial target with gemcitabine in pancreatic cancer cells. Proc Natl Acad Sci U S A. 2020;117(45):28068–79.PubMedPubMedCentralCrossRef
161.
Zurück zum Zitat Wang T, Yu H, Hughes NW, Liu B, Kendirli A, Klein K, et al. Gene Essentiality Profiling Reveals Gene Networks and Synthetic Lethal Interactions with Oncogenic Ras. Cell. 2017;168(5):890–903 e15.PubMedPubMedCentralCrossRef Wang T, Yu H, Hughes NW, Liu B, Kendirli A, Klein K, et al. Gene Essentiality Profiling Reveals Gene Networks and Synthetic Lethal Interactions with Oncogenic Ras. Cell. 2017;168(5):890–903 e15.PubMedPubMedCentralCrossRef
162.
Zurück zum Zitat Sulahian R, Kwon JJ, Walsh KH, Pailler E, Bosse TL, Thaker M, et al. Synthetic Lethal Interaction of SHOC2 Depletion with MEK Inhibition in RAS-Driven Cancers. Cell Rep. 2019;29(1):118–34 e8.PubMedPubMedCentralCrossRef Sulahian R, Kwon JJ, Walsh KH, Pailler E, Bosse TL, Thaker M, et al. Synthetic Lethal Interaction of SHOC2 Depletion with MEK Inhibition in RAS-Driven Cancers. Cell Rep. 2019;29(1):118–34 e8.PubMedPubMedCentralCrossRef
163.
Zurück zum Zitat Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20(9):1064–73.PubMedPubMedCentralCrossRef Nichols RJ, Haderk F, Stahlhut C, Schulze CJ, Hemmati G, Wildes D, et al. RAS nucleotide cycling underlies the SHP2 phosphatase dependence of mutant BRAF-, NF1- and RAS-driven cancers. Nat Cell Biol. 2018;20(9):1064–73.PubMedPubMedCentralCrossRef
164.
Zurück zum Zitat Wang Y, Ngo VN, Marani M, Yang Y, Wright G, Staudt LM, et al. Critical role for transcriptional repressor Snail2 in transformation by oncogenic RAS in colorectal carcinoma cells. Oncogene. 2010;29(33):4658–70.PubMedPubMedCentralCrossRef Wang Y, Ngo VN, Marani M, Yang Y, Wright G, Staudt LM, et al. Critical role for transcriptional repressor Snail2 in transformation by oncogenic RAS in colorectal carcinoma cells. Oncogene. 2010;29(33):4658–70.PubMedPubMedCentralCrossRef
165.
Zurück zum Zitat Scholl C, Frohling S, Dunn IF, Schinzel AC, Barbie DA, Kim SY, et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell. 2009;137(5):821–34.PubMedCrossRef Scholl C, Frohling S, Dunn IF, Schinzel AC, Barbie DA, Kim SY, et al. Synthetic lethal interaction between oncogenic KRAS dependency and STK33 suppression in human cancer cells. Cell. 2009;137(5):821–34.PubMedCrossRef
166.
Zurück zum Zitat Zhu Z, Aref AR, Cohoon TJ, Barbie TU, Imamura Y, Yang S, et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 2014;4(4):452–65.PubMedPubMedCentralCrossRef Zhu Z, Aref AR, Cohoon TJ, Barbie TU, Imamura Y, Yang S, et al. Inhibition of KRAS-driven tumorigenicity by interruption of an autocrine cytokine circuit. Cancer Discov. 2014;4(4):452–65.PubMedPubMedCentralCrossRef
167.
Zurück zum Zitat Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell. 2014;158(1):185–97.PubMedPubMedCentralCrossRef Kapoor A, Yao W, Ying H, Hua S, Liewen A, Wang Q, et al. Yap1 activation enables bypass of oncogenic Kras addiction in pancreatic cancer. Cell. 2014;158(1):185–97.PubMedPubMedCentralCrossRef
168.
Zurück zum Zitat Kim J, McMillan E, Kim HS, Venkateswaran N, Makkar G, Rodriguez-Canales J, et al. XPO1-dependent nuclear export is a druggable vulnerability in KRAS-mutant lung cancer. Nature. 2016;538(7623):114–7.PubMedPubMedCentralCrossRef Kim J, McMillan E, Kim HS, Venkateswaran N, Makkar G, Rodriguez-Canales J, et al. XPO1-dependent nuclear export is a druggable vulnerability in KRAS-mutant lung cancer. Nature. 2016;538(7623):114–7.PubMedPubMedCentralCrossRef
169.
Zurück zum Zitat Zeng M, Lu J, Li L, Feru F, Quan C, Gero TW, et al. Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C. Cell Chem Biol. 2017;24(8):1005–16 e3.PubMedCrossRef Zeng M, Lu J, Li L, Feru F, Quan C, Gero TW, et al. Potent and Selective Covalent Quinazoline Inhibitors of KRAS G12C. Cell Chem Biol. 2017;24(8):1005–16 e3.PubMedCrossRef
170.
Zurück zum Zitat Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351(6273):604–8.PubMedPubMedCentralCrossRef Lito P, Solomon M, Li LS, Hansen R, Rosen N. Allele-specific inhibitors inactivate mutant KRAS G12C by a trapping mechanism. Science. 2016;351(6273):604–8.PubMedPubMedCentralCrossRef
171.
Zurück zum Zitat Janes MR, Zhang J, Li LS, Hansen R, Peters U, Guo X, et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell. 2018;172(3):578–89 e17.PubMedCrossRef Janes MR, Zhang J, Li LS, Hansen R, Peters U, Guo X, et al. Targeting KRAS Mutant Cancers with a Covalent G12C-Specific Inhibitor. Cell. 2018;172(3):578–89 e17.PubMedCrossRef
172.
Zurück zum Zitat Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503(7477):548–51.PubMedPubMedCentralCrossRef Ostrem JM, Peters U, Sos ML, Wells JA, Shokat KM. K-Ras(G12C) inhibitors allosterically control GTP affinity and effector interactions. Nature. 2013;503(7477):548–51.PubMedPubMedCentralCrossRef
173.
Zurück zum Zitat Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, et al. Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. Cancer Discov. 2016;6(3):316–29.PubMedCrossRef Patricelli MP, Janes MR, Li LS, Hansen R, Peters U, Kessler LV, et al. Selective Inhibition of Oncogenic KRAS Output with Small Molecules Targeting the Inactive State. Cancer Discov. 2016;6(3):316–29.PubMedCrossRef
174.
Zurück zum Zitat Saleh K, Kordahi M, Felefly T, Kourie HR, Khalife N. KRAS-targeted therapies in advanced solid cancers: drug the undruggable? Pharmacogenomics. 2021;22(10):587–90.PubMedCrossRef Saleh K, Kordahi M, Felefly T, Kourie HR, Khalife N. KRAS-targeted therapies in advanced solid cancers: drug the undruggable? Pharmacogenomics. 2021;22(10):587–90.PubMedCrossRef
175.
Zurück zum Zitat Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575(7781):217–23.PubMedCrossRef Canon J, Rex K, Saiki AY, Mohr C, Cooke K, Bagal D, et al. The clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunity. Nature. 2019;575(7781):217–23.PubMedCrossRef
176.
Zurück zum Zitat Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov. 2020;10(1):54–71.PubMedCrossRef Hallin J, Engstrom LD, Hargis L, Calinisan A, Aranda R, Briere DM, et al. The KRAS(G12C) Inhibitor MRTX849 Provides Insight toward Therapeutic Susceptibility of KRAS-Mutant Cancers in Mouse Models and Patients. Cancer Discov. 2020;10(1):54–71.PubMedCrossRef
177.
Zurück zum Zitat Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med. 2020;383(13):1207–17.PubMedPubMedCentralCrossRef Hong DS, Fakih MG, Strickler JH, Desai J, Durm GA, Shapiro GI, et al. KRAS(G12C) Inhibition with Sotorasib in Advanced Solid Tumors. N Engl J Med. 2020;383(13):1207–17.PubMedPubMedCentralCrossRef
178.
Zurück zum Zitat Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med. 2021;384(25):2371–81.PubMedCrossRefPubMedCentral Skoulidis F, Li BT, Dy GK, Price TJ, Falchook GS, Wolf J, et al. Sotorasib for Lung Cancers with KRAS p.G12C Mutation. N Engl J Med. 2021;384(25):2371–81.PubMedCrossRefPubMedCentral
179.
Zurück zum Zitat Arbour KC, Jordan E, Kim HR, Dienstag J, Yu HA, Sanchez-Vega F, et al. Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res. 2018;24(2):334–40.PubMedCrossRef Arbour KC, Jordan E, Kim HR, Dienstag J, Yu HA, Sanchez-Vega F, et al. Effects of Co-occurring Genomic Alterations on Outcomes in Patients with KRAS-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res. 2018;24(2):334–40.PubMedCrossRef
180.
Zurück zum Zitat Kessler D, Gmachl M, Mantoulidis A, Martin LJ, Zoephel A, Mayer M, et al. Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci U S A. 2019;116(32):15823–9.PubMedPubMedCentralCrossRef Kessler D, Gmachl M, Mantoulidis A, Martin LJ, Zoephel A, Mayer M, et al. Drugging an undruggable pocket on KRAS. Proc Natl Acad Sci U S A. 2019;116(32):15823–9.PubMedPubMedCentralCrossRef
181.
Zurück zum Zitat Ricciardiello F, Bergamaschi L, De Vitto H, Gang Y, Zhang T, Palorini R, et al. Suppression of the HBP Function Increases Pancreatic Cancer Cell Sensitivity to a Pan-RAS Inhibitor. Cells. 2021;10(2):431. Ricciardiello F, Bergamaschi L, De Vitto H, Gang Y, Zhang T, Palorini R, et al. Suppression of the HBP Function Increases Pancreatic Cancer Cell Sensitivity to a Pan-RAS Inhibitor. Cells. 2021;10(2):431.
182.
Zurück zum Zitat Kim J, Lee HM, Cai F, Ko B, Yang C, Lieu EL, et al. The hexosamine biosynthesis pathway is a targetable liability in KRAS/LKB1 mutant lung cancer. Nat Metab. 2020;2(12):1401–12. Kim J, Lee HM, Cai F, Ko B, Yang C, Lieu EL, et al. The hexosamine biosynthesis pathway is a targetable liability in KRAS/LKB1 mutant lung cancer. Nat Metab. 2020;2(12):1401–12.
184.
Zurück zum Zitat Zeng M, Xiong Y, Safaee N, Nowak RP, Donovan KA, Yuan CJ, et al. Exploring Targeted Degradation Strategy for Oncogenic KRAS(G12C). Cell Chem Biol. 2020;27(1):19–31 e6.PubMedCrossRef Zeng M, Xiong Y, Safaee N, Nowak RP, Donovan KA, Yuan CJ, et al. Exploring Targeted Degradation Strategy for Oncogenic KRAS(G12C). Cell Chem Biol. 2020;27(1):19–31 e6.PubMedCrossRef
185.
Zurück zum Zitat Bond MJ, Chu L, Nalawansha DA, Li K, Crews CM. Targeted Degradation of Oncogenic KRAS(G12C) by VHL-Recruiting PROTACs. ACS Cent Sci. 2020;6(8):1367–75.PubMedPubMedCentralCrossRef Bond MJ, Chu L, Nalawansha DA, Li K, Crews CM. Targeted Degradation of Oncogenic KRAS(G12C) by VHL-Recruiting PROTACs. ACS Cent Sci. 2020;6(8):1367–75.PubMedPubMedCentralCrossRef
186.
Zurück zum Zitat Xue JY, Zhao Y, Aronowitz J, Mai TT, Vides A, Qeriqi B, et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature. 2020;577(7790):421–5.PubMedPubMedCentralCrossRef Xue JY, Zhao Y, Aronowitz J, Mai TT, Vides A, Qeriqi B, et al. Rapid non-uniform adaptation to conformation-specific KRAS(G12C) inhibition. Nature. 2020;577(7790):421–5.PubMedPubMedCentralCrossRef
187.
Zurück zum Zitat Huang J, Chen P, Liu K, Liu J, Zhou B, Wu R, et al. CDK1/2/5 inhibition overcomes IFNG-mediated adaptive immune resistance in pancreatic cancer. Gut. 2021;70(5):890–9. Huang J, Chen P, Liu K, Liu J, Zhou B, Wu R, et al. CDK1/2/5 inhibition overcomes IFNG-mediated adaptive immune resistance in pancreatic cancer. Gut. 2021;70(5):890–9.
188.
Zurück zum Zitat Xie Y, Zhu S, Zhong M, Yang M, Sun X, Liu J, et al. Inhibition of Aurora Kinase A Induces Necroptosis in Pancreatic Carcinoma. Gastroenterology. 2017;153(5):1429–43 e5.PubMedCrossRef Xie Y, Zhu S, Zhong M, Yang M, Sun X, Liu J, et al. Inhibition of Aurora Kinase A Induces Necroptosis in Pancreatic Carcinoma. Gastroenterology. 2017;153(5):1429–43 e5.PubMedCrossRef
189.
Zurück zum Zitat Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018;8(2):216–33.PubMedCrossRef Deng J, Wang ES, Jenkins RW, Li S, Dries R, Yates K, et al. CDK4/6 Inhibition Augments Antitumor Immunity by Enhancing T-cell Activation. Cancer Discov. 2018;8(2):216–33.PubMedCrossRef
190.
Zurück zum Zitat Zhang H, Pandey S, Travers M, Sun H, Morton G, Madzo J, et al. Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell. 2018;175(5):1244–58 e26.PubMedPubMedCentralCrossRef Zhang H, Pandey S, Travers M, Sun H, Morton G, Madzo J, et al. Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer. Cell. 2018;175(5):1244–58 e26.PubMedPubMedCentralCrossRef
191.
Zurück zum Zitat Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–5.PubMedPubMedCentralCrossRef Goel S, DeCristo MJ, Watt AC, BrinJones H, Sceneay J, Li BB, et al. CDK4/6 inhibition triggers anti-tumour immunity. Nature. 2017;548(7668):471–5.PubMedPubMedCentralCrossRef
192.
Zurück zum Zitat Dorand RD, Nthale J, Myers JT, Barkauskas DS, Avril S, Chirieleison SM, et al. Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity. Science. 2016;353(6297):399–403.PubMedPubMedCentralCrossRef Dorand RD, Nthale J, Myers JT, Barkauskas DS, Avril S, Chirieleison SM, et al. Cdk5 disruption attenuates tumor PD-L1 expression and promotes antitumor immunity. Science. 2016;353(6297):399–403.PubMedPubMedCentralCrossRef
193.
Zurück zum Zitat Ryan MB. Fece de la Cruz F, Phat S, Myers DT, Wong E, Shahzade HA et al. Vertical Pathway Inhibition Overcomes Adaptive Feedback Resistance to KRAS(G12C) Inhibition. Clin Cancer Res. 2020;26(7):1633–43.PubMedCrossRef Ryan MB. Fece de la Cruz F, Phat S, Myers DT, Wong E, Shahzade HA et al. Vertical Pathway Inhibition Overcomes Adaptive Feedback Resistance to KRAS(G12C) Inhibition. Clin Cancer Res. 2020;26(7):1633–43.PubMedCrossRef
194.
Zurück zum Zitat Lou K, Steri V, Ge AY, Hwang YC, Yogodzinski CH, Shkedi AR, et al. KRAS(G12C) inhibition produces a driver-limited state revealing collateral dependencies. Sci Signal. 2019;12(583):eaaw9450. Lou K, Steri V, Ge AY, Hwang YC, Yogodzinski CH, Shkedi AR, et al. KRAS(G12C) inhibition produces a driver-limited state revealing collateral dependencies. Sci Signal. 2019;12(583):eaaw9450.
195.
Zurück zum Zitat Santana-Codina N, Chandhoke AS, Yu Q, Malachowska B, Kuljanin M, Gikandi A, et al. Defining and Targeting Adaptations to Oncogenic KRAS(G12C) Inhibition Using Quantitative Temporal Proteomics. Cell Rep. 2020;30(13):4584–99 e4.PubMedCrossRef Santana-Codina N, Chandhoke AS, Yu Q, Malachowska B, Kuljanin M, Gikandi A, et al. Defining and Targeting Adaptations to Oncogenic KRAS(G12C) Inhibition Using Quantitative Temporal Proteomics. Cell Rep. 2020;30(13):4584–99 e4.PubMedCrossRef
196.
Zurück zum Zitat Solanki HS, Welsh EA, Fang B, Izumi V, Darville L, Stone B, et al. Cell Type-specific Adaptive Signaling Responses to KRAS(G12C) Inhibition. Clin Cancer Res. 2021;27(9):2533–48.PubMedCrossRef Solanki HS, Welsh EA, Fang B, Izumi V, Darville L, Stone B, et al. Cell Type-specific Adaptive Signaling Responses to KRAS(G12C) Inhibition. Clin Cancer Res. 2021;27(9):2533–48.PubMedCrossRef
197.
Zurück zum Zitat Fedele C, Li S, Teng KW, Foster CJR, Peng D, Ran H, et al. SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med. 2021;218(1):e20201414. Fedele C, Li S, Teng KW, Foster CJR, Peng D, Ran H, et al. SHP2 inhibition diminishes KRASG12C cycling and promotes tumor microenvironment remodeling. J Exp Med. 2021;218(1):e20201414.
198.
Zurück zum Zitat Suda K, Murakami I, Yu H, Kim J, Tan AC, Mizuuchi H, et al. CD44 Facilitates Epithelial-to-Mesenchymal Transition Phenotypic Change at Acquisition of Resistance to EGFR Kinase Inhibitors in Lung Cancer. Mol Cancer Ther. 2018;17(10):2257–65.PubMedCrossRef Suda K, Murakami I, Yu H, Kim J, Tan AC, Mizuuchi H, et al. CD44 Facilitates Epithelial-to-Mesenchymal Transition Phenotypic Change at Acquisition of Resistance to EGFR Kinase Inhibitors in Lung Cancer. Mol Cancer Ther. 2018;17(10):2257–65.PubMedCrossRef
199.
Zurück zum Zitat Adachi Y, Ito K, Hayashi Y, Kimura R, Tan TZ, Yamaguchi R, et al. Epithelial-to-Mesenchymal Transition is a Cause of Both Intrinsic and Acquired Resistance to KRAS G12C Inhibitor in KRAS G12C-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res. 2020;26(22):5962–73.PubMedCrossRef Adachi Y, Ito K, Hayashi Y, Kimura R, Tan TZ, Yamaguchi R, et al. Epithelial-to-Mesenchymal Transition is a Cause of Both Intrinsic and Acquired Resistance to KRAS G12C Inhibitor in KRAS G12C-Mutant Non-Small Cell Lung Cancer. Clin Cancer Res. 2020;26(22):5962–73.PubMedCrossRef
201.
Zurück zum Zitat Brown WS, McDonald PC, Nemirovsky O, Awrey S, Chafe SC, Schaeffer DF, et al. Overcoming Adaptive Resistance to KRAS and MEK Inhibitors by Co-targeting mTORC1/2 Complexes in Pancreatic Cancer. Cell Rep Med. 2020;1(8):100131.PubMedPubMedCentralCrossRef Brown WS, McDonald PC, Nemirovsky O, Awrey S, Chafe SC, Schaeffer DF, et al. Overcoming Adaptive Resistance to KRAS and MEK Inhibitors by Co-targeting mTORC1/2 Complexes in Pancreatic Cancer. Cell Rep Med. 2020;1(8):100131.PubMedPubMedCentralCrossRef
202.
Zurück zum Zitat Misale S, Fatherree JP, Cortez E, Li C, Bilton S, Timonina D, et al. KRAS G12C NSCLC Models Are Sensitive to Direct Targeting of KRAS in Combination with PI3K Inhibition. Clin Cancer Res. 2019;25(2):796–807.PubMedCrossRef Misale S, Fatherree JP, Cortez E, Li C, Bilton S, Timonina D, et al. KRAS G12C NSCLC Models Are Sensitive to Direct Targeting of KRAS in Combination with PI3K Inhibition. Clin Cancer Res. 2019;25(2):796–807.PubMedCrossRef
203.
Zurück zum Zitat Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N Engl J Med. 2021;384(25):2382–93.PubMedCrossRefPubMedCentral Awad MM, Liu S, Rybkin II, Arbour KC, Dilly J, Zhu VW, et al. Acquired Resistance to KRAS(G12C) Inhibition in Cancer. N Engl J Med. 2021;384(25):2382–93.PubMedCrossRefPubMedCentral
204.
Zurück zum Zitat Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical Acquired Resistance to KRAS(G12C) Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS-MAPK Reactivation. Cancer Discov. 2021;11(8):1913–22. Tanaka N, Lin JJ, Li C, Ryan MB, Zhang J, Kiedrowski LA, et al. Clinical Acquired Resistance to KRAS(G12C) Inhibition through a Novel KRAS Switch-II Pocket Mutation and Polyclonal Alterations Converging on RAS-MAPK Reactivation. Cancer Discov. 2021;11(8):1913–22.
205.
Zurück zum Zitat Koga T, Suda K, Fujino T, Ohara S, Hamada A, Nishino M, et al. KRAS Secondary Mutations That Confer Acquired Resistance to KRAS G12C Inhibitors, Sotorasib and Adagrasib, and Overcoming Strategies: Insights From the In Vitro Experiments. J Thorac Oncol. 2021;16(8):1321–32. Koga T, Suda K, Fujino T, Ohara S, Hamada A, Nishino M, et al. KRAS Secondary Mutations That Confer Acquired Resistance to KRAS G12C Inhibitors, Sotorasib and Adagrasib, and Overcoming Strategies: Insights From the In Vitro Experiments. J Thorac Oncol. 2021;16(8):1321–32.
206.
Zurück zum Zitat Dunnett-Kane V, Nicola P, Blackhall F, Lindsay C. Mechanisms of Resistance to KRAS(G12C) Inhibitors. Cancers (Basel). 2021;13(1):151. Dunnett-Kane V, Nicola P, Blackhall F, Lindsay C. Mechanisms of Resistance to KRAS(G12C) Inhibitors. Cancers (Basel). 2021;13(1):151.
207.
Zurück zum Zitat Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357(6356):1156–60.PubMedPubMedCentralCrossRef Geller LT, Barzily-Rokni M, Danino T, Jonas OH, Shental N, Nejman D, et al. Potential role of intratumor bacteria in mediating tumor resistance to the chemotherapeutic drug gemcitabine. Science. 2017;357(6356):1156–60.PubMedPubMedCentralCrossRef
208.
209.
Zurück zum Zitat Thein KZ, Biter AB, Hong DS. Therapeutics Targeting Mutant KRAS. Annu Rev Med. 2021;72:349–64.PubMedCrossRef Thein KZ, Biter AB, Hong DS. Therapeutics Targeting Mutant KRAS. Annu Rev Med. 2021;72:349–64.PubMedCrossRef
Metadaten
Titel
Oncogenic KRAS blockade therapy: renewed enthusiasm and persistent challenges
verfasst von
Daolin Tang
Guido Kroemer
Rui Kang
Publikationsdatum
01.12.2021
Verlag
BioMed Central
Erschienen in
Molecular Cancer / Ausgabe 1/2021
Elektronische ISSN: 1476-4598
DOI
https://doi.org/10.1186/s12943-021-01422-7

Weitere Artikel der Ausgabe 1/2021

Molecular Cancer 1/2021 Zur Ausgabe

Mehr Brustkrebs, aber weniger andere gynäkologische Tumoren mit Levonorgestrel-IUS

04.06.2024 Levonorgestrel Nachrichten

Unter Frauen, die ein Levonorgestrel-freisetzendes intrauterines System (IUS) verwenden, ist die Brustkrebsrate um 13% erhöht. Dafür kommt es deutlich seltener zu Endometrium-, Zervix- und Ovarialkarzinomen.

Bei seelischem Stress sind Checkpoint-Hemmer weniger wirksam

03.06.2024 NSCLC Nachrichten

Wie stark Menschen mit fortgeschrittenem NSCLC von einer Therapie mit Immun-Checkpoint-Hemmern profitieren, hängt offenbar auch davon ab, wie sehr die Diagnose ihre psychische Verfassung erschüttert

Antikörper mobilisiert Neutrophile gegen Krebs

03.06.2024 Onkologische Immuntherapie Nachrichten

Ein bispezifischer Antikörper formiert gezielt eine Armee neutrophiler Granulozyten gegen Krebszellen. An den Antikörper gekoppeltes TNF-alpha soll die Zellen zudem tief in solide Tumoren hineinführen.

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Update Onkologie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.