Trending Topic

3D Rendered Medical Illustration of Male Anatomy - Colon Cancer; Ascending Colon.
17 mins

Trending Topic

Developed by Touch
Mark CompleteCompleted
BookmarkBookmarked

Rat sarcoma virus (RAS) proteins are a family of prototypical oncogenes frequently mutated in human cancers. Mutations in the RAS gene account for 19% of all pathogenic alterations and are the subject of extensive research in molecular and clinical oncology.1 The RAS family consists of three major isoforms, namely the Harvey rat sarcoma virus (HRAS), the neuroblastoma RAS […]

Advances in Targeting KRAS G12C in Gastrointestinal Malignancies: Focus on KRAS G12C Inhibitors

Celine Hoyek, Angelo Pirozzi, Jeremy Jones, Lorenza Rimassa, Mohamad B Sonbol, Tanios Bekaii-Saab
Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF
Published Online: Dec 2nd 2024 touchREVIEWS in Oncology & Haematology. 2024;20(2):1–7:Online ahead of journal publication
Select a Section…
1

Abstract

Overview

Kirsten rat sarcoma virus (KRAS) gene mutations are amongst the most prevalent oncogenic mutations in gastrointestinal tumours. Attempts at targeting the KRAS protein have been unsuccessful for decades, mainly due to its challenging architecture that lacks small-molecule-binding sites. Recent breakthrough advances in crystallographic studies and related fields have led to the discovery of an allosteric binding pocket specific to the KRAS G12C protein, which inhibitors can selectively target through its cysteine residue. The development of KRAS G12C inhibitors, such as adagrasib and sotorasib, and their demonstrated clinical activity led to their regulatory approval, first for treating non-small-cell lung cancer, and more recently for pancreatic and colorectal cancers. Since then, several KRAS G12C inhibitors have emerged and are entering the clinic. However, a significant challenge currently faced by KRAS G12C inhibitors is the emergence of resistance. Understanding these mechanisms is essential to guide the development of future combination therapies, which include concurrent vertical inhibition of the receptor tyrosine kinase/rat sarcoma virus (RAS)/mitogen-activated protein kinase (MAPK) pathway, combinations with chemotherapy or immunotherapy and other innovations that target key players of RAS pathway activation, such as Src Homologous Protein 2 and Son of Sevenless 1, to prolong the survival of patients with limited treatment options.

Keywords
2

Article

Rat sarcoma virus (RAS) proteins are a family of prototypical oncogenes frequently mutated in human cancers. Mutations in the RAS gene account for 19% of all pathogenic alterations and are the subject of extensive research in molecular and clinical oncology.1 The RAS family consists of three major isoforms, namely the Harvey rat sarcoma virus (HRAS), the neuroblastoma RAS viral oncogene homologue (NRAS) and the Kirsten rat sarcoma virus (KRAS). Amongst these, KRAS is the most frequently mutated of the RAS isoforms, with a remarkably high mutational prevalence in gastrointestinal (GI) malignancies, including 90% of pancreatic ductal adenocarcinomas (PDAC) and 50% of colorectal cancers (CRC).2 The high prevalence of KRAS aberrations in GI malignancies was crucial in promoting studies that identified KRAS’s central role as an oncogenic driver and a critical factor in resistance against cytotoxic chemotherapy.3 However, despite decades of research into its molecular configuration, efforts in targeting KRAS have been elusive for several reasons: the absence of deep pockets in the RAS protein, making it inaccessible to small-molecule inhibitors, the constant transition between two distinct conformational states with vastly different chemical behaviours and the high affinity for guanosine-5′-triphosphate (GTP), allowing the KRAS protein to function despite low GTP concentrations, have made the design of a targeted KRAS inhibitor particularly difficult.4

Nevertheless, advances in molecular biology and conformational biochemistry have profoundly changed our understanding of an anomalous mutant variant known as the Gly12Cys KRAS (KRAS G12C) mutant.5 The successful development of KRAS G12C inhibitors is attributed to the distinct presence of the switch II pocket that allows covalent inhibition of the cysteine residue, as well as the high GTPase activity of KRAS G12C that likely results in an attenuated duration of rapidly accelerated fibrosarcoma (RAF) kinase activation, which is not fully present in other KRAS-mutant types.6 Despite the ubiquity of KRAS mutations in solid tumours, the global prevalence of KRAS G12C mutations is relatively low in PDAC and CRC, accounting for only 1.3 and 3.1% of the total mutational burden, respectively (Table 1).9–11 Moreover, several studies in CRC and PDAC have found that KRAS mutations, particularly KRAS G12C, are associated with a poor response to cytotoxic chemotherapy.12 Indeed, patients with KRAS G12C-mutated CRC had a median overall survival (mOS) of 16.1 (95% confidence interval [CI], 13.019.0) months compared with 18.3 (95% CI, 17.219.3) months for KRAS non-G12C-mutated tumours and 19.2 (18.519.8) months for the mCRC overall cohort .13,14 In PDAC, mOS was 16.7 months in KRAS G12C-mutated tumours versus 24.9 months in KRAS non-G12C-mutated tumours.15 This review aims to summarize the pleiotropic functions of the KRAS gene, highlight the unique features of the KRAS G12C protein concerning its biological role and targeted treatments, describe the main mechanisms of resistance of currently approved drugs and finally review the results from published clinical trials testing KRAS G12C inhibitors, as well as KRAS G12C downstream and upstream protein inhibitors, with a focus on the most promising ongoing trials.

Table 1: Prevalence and distribution of Kirsten rat sarcoma virus G12 mutations across gastrointestinal cancer types7,8

Colorectal (%)

Pancreas (%)

Gastric/Esophageal (%)

Gallbladder/cholangiocarcinoma (%)

KRAS G12C

7.0

1.3

5.9/7.6

0.5/1.2

KRAS G12D

29.9

41.8

27.0/33.0

33.6

KRAS G12V

20.0

31.6

18.0/13.0

27.2

KRAS G12R

1.1

16.1

3.9/0.8

9.6

KRAS = Kirsten rat sarcoma virus.

Kirsten rat sarcoma virus structure and effects of activating Kirsten rat sarcoma virus mutations

RAS proteins are small, membrane-bound guanine nucleotide-binding proteins. The major RAS isoforms are encoded by three genes, HRAS, NRAS and KRAS, producing four proteins, HRAS, NRAS, KRAS4A and KRAS4B, from a KRAS splice variant. The amino-terminal residues 1–165 of these proteins share 92–98% sequence similarity, and the remaining 23–24 carboxy-terminal residues diverge significantly. The structural domain of the RAS protein consists of the first 166–168 residues, forming the G domain, a structure featuring a mix of six-stranded β-sheet and five-α-helix fold, typical of α,β-nucleotide-binding proteins. Four main regions border the nucleotide-binding pocket: the phosphate-binding loop (P-loop, residues 10–17), switch I (residues 30–38), switch II (residues 60–76) and the base-binding loops (residues 116–120 and 145–147).16 Similar to its RAS family counterparts, NRAS and HRAS, KRAS mediates downstream signalling via the activation of the RAF protein/mitogen-activated protein kinase kinase (MEK)/extracellular signal-regulated kinases (ERK) cascade, the phosphoinositide-3-kinase complex/mammalian target of rapamycin (PI3K/mTOR) axis and ultimately the RAS-related protein/nuclear factor-κB (NF-κB) pathway, driving tumour onset, progression and distant spread.17–19 KRAS constantly transitions between an inactive ‘off’ state when bound to guanosine diphosphate (GDP) and an active ‘on’ state when bound to GTP, a process facilitated by guanine nucleotide exchange factors (GEFs) such as Son of Sevenless (SOS) 1 and 2. In the GTP-bound state, threonine-35 (in the switch I region) and glycine-60 (in the switch II region) stabilize the active conformations of the switch I and switch II regions by anchoring to the γ-phosphate of GTP. Upon phosphate release during GTP hydrolysis, switch I and switch II recoil back into their inactive GDP conformations, such that these regions ultimately regulate all known nucleotide-dependent interactions between RAS and its binding partners.16 The exchange of GDP for GTP results in the activation and dimerization of RAS proteins, ultimately leading to the propagation of signal transduction cascades. This transition is mainly regulated by a variety of upstream receptor tyrosine kinases (RTKs) such as the epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2 (HER2), human epidermal growth factor receptor 3 and fibroblast growth factor receptors (FGFRs).20 Most KRAS-activating mutations trigger the conversion of KRAS-GDP into KRAS-GTP, further enhanced by inhibiting GTPase activity that locks KRAS in its ‘on’ state, leading to the constitutive activation of downstream signalling pathways (Figure 1).

Figure 1: Schematic representation of Kirsten rat sarcoma virus G12C signalling pathways and regulatory mechanisms

Figure 1: Schematic representation of Kirsten rat sarcoma virus G12C signalling pathways and regulatory mechanisms

Simplified illustration of the signalling pathways downstream of KRAS G12C in its active and inactive states. Black arrows indicate upregulation or activation. At the cell membrane, RTKs initiate the activation of KRAS. In the inactive state, KRAS G12C is bound to GDP and undergoes activation through interaction with GEFs, which facilitate the exchange of GDP for GTP. Once bound to GTP, KRAS G12C transitions to its active state and promotes the activation of multiple downstream signalling pathways (RAF/MEK/ERK, PI3K/AKT/mTOR pathway and RAL/NF-κB). The GAPs promote the hydrolysis of GTP to GDP, inactivating KRAS G12C and returning it to the inactive GDP-bound state. These signalling pathways ultimately converge at the nucleus, regulating cellular processes such as proliferation, survival and tumour progression.

AKT = protein kinase B; ERK = extracellular signal-regulated kinase; GAP = GTPase-activating protein; GDP = guanosine diphosphate; GEF = guanine nucleotide exchange factors;
GTP = guanosine-5-triphosphate; KRAS = Kirsten rat sarcoma virus; MEK = m
itogen-activated protein kinase kinase; mTOR = mammalian target of rapamycin; NF-kB = nuclear

factor-κB; PI3K = phosphoinositide3-kinase complex; RAF = rapidly accelerated fibrosarcoma; RAL = RAS-related protein; RTKs = receptor tyrosine kinase.

Evolution of Kirsten rat sarcoma virus-targeting strategies

Targeting KRAS has been an important research challenge during the last 30 years when it was considered ‘undruggable’. This challenge stemmed in part from the structure of KRAS, a small protein with a relatively smooth surface that lacks well-defined hydrophobic pockets (except for its GTP/GDP-binding pocket) suitable for small-molecule inhibitor binding, as well as from KRAS’s ability to bind GTP with picomolar affinity in an environment with high intracellular GTP concentrations.16,21–24 In addition, each KRAS mutation alters the structure of the GTP-binding pocket differently, which further complicates the design of an inhibitor that is effective against multiple KRAS alterations.25–27 Strategies therefore focused on indirectly targeting KRAS by inhibiting downstream signalling effectors such as PI3K and MEK, modifying the epigenetic environment with telomerase inhibitors, and RNA interference or promoting synthetic lethality using cyclin-dependent kinase inhibitors, with limited tolerability and efficacy.28–31 In the last decade, molecular studies of KRAS-mutated proteins highlighted the unique structure of the aberrant variant KRAS G12C, resulting from a missense mutation, causing the substitution of glycine with cysteine at codon 12 in exon 2. The presence of cysteine causes a steric block that prevents the arginine finger of GTPase-activating proteins from binding to the GTPase site of RAS, inhibiting the hydrolysis of GTP to GDP, thus maintaining KRAS in a constitutively active state.32 However, a distinguishing feature of the KRAS G12C protein, discovered by Shokat’s laboratory in 2013, is the presence of an allosteric pocket below the switch II region of the mutated cysteine residue, called the switch II pocket, which is susceptible to inhibition through covalent binding.20 Unlike other KRAS mutations, KRAS G12C does not significantly alter the intrinsic GTPase activity, allowing KRAS to remain in the GDP-bound or inactive state for a longer period.33 These biochemical and functional properties paved the way for the development of the first KRAS G12C-targeted therapies: sotorasib and adagrasib. Both drugs irreversibly bind the cysteine residue in the switch II pocket of KRAS G12C, locking it in its inactive GDP-bound state and inhibiting downstream signalling.16,34 While adagrasib has high selectivity for KRAS G12C, sotorasib inhibits NRAS G12C more potently compared with KRAS G12C or HRAS G12C. According to structural and reciprocal mutagenesis studies, differences in isoform-specific binding are mediated only by histidine-95 in KRAS and leucine-95 in NRAS.35 A single patient with NRAS G12C CRC was reported to have had a marked tumour response after being treated with sotorasib and panitumumab, suggesting that sotorasib can be clinically effective in NRAS G12C-mutated tumours.35 Regardless, the clinical application of single-agent sotorasib was studied in 2021 to treat advanced KRAS G12C-mutated non-small-cell lung cancer (NSCLC) in the CodeBreaK 100 trial (A Phase 1/2, Open-label Study Evaluating the Safety, Tolerability, Pharmacokinetics, Pharmacodynamics, and Efficacy of Sotorasib [AMG 510] Monotherapy in Subjects With Advanced Solid Tumors With KRAS p.G12C Mutation and Sotorasib [AMG 510] Combination Therapy in Subjects With Advanced NSCLC With KRAS p.G12C Mutation; ClinicalTrials.gov identifier: NCT03600883).36 This phase II trial enrolled 126 patients (pts) with previously treated KRAS G12C-mutated NSCLC who received sotorasib. The overall response rate (ORR) was 37.1% (95% CI, 28.6–46.2), disease control rate (DCR) was 80.6% (95% CI, 72.687.2), median progression-free survival (mPFS) was 6.8 months (95% CI, 5.1–8.2) and mOS was 12.5 months.36 The CodeBreaK 100 trial has been considered a landmark trial based on these results, and sotorasib was granted accelerated approval by the US Food and Drug Administration (FDA) in May 2021 for the treatment of locally advanced or metastatic KRAS G12C-mutated NSCLC pts who have failed one or more systemic therapies.37. Comparable findings were observed with adagrasib monotherapy in a similar population, prompting its approval by the US FDA in December 2022.38 Sotorasib and adagrasib have also shown encouraging but varied results in GI cancers. The initial findings from the CodeBreaK 100 trial included a cohort of 12 pts with KRAS G12C-mutated PDAC who received sotorasib monotherapy, amongst whom one achieved a partial response (PR).39 When the trial was expanded to include 38 pts with KRAS G12C-mutated PDAC, mPFS was 4.0 months and mOS was 6.9 months. In addition, 21% of pts achieved a PR, with a DCR of 84%.40 In the phase I–II KRYSTAL-1 trial (ClinicalTrials.gov identifier: NCT03785249), adagrasib monotherapy demonstrated comparable outcomes to sotorasib: for 21 pts with metastatic PDAC receiving adagrasib, mPFS was 5.4 months (95% CI, 3.9–8.2) and mOS was 8.0 months (95% CI, 5.2–11.8). ORR was 33% and DCR was 49%.41 So far, both adagrasib and sotorasib have been adopted as single-agent therapeutic options by the National Comprehensive Cancer Network guidelines for advanced KRAS G12C-mutated PDAC and biliary tract cancers (BTC).41 While single-agent KRAS G12C inhibitors have shown promising results in PDAC, their efficacy appears less pronounced in CRC. On the other hand, improved results were observed testing sotorasib and adagrasib in combination with EGFR inhibitors. The analysis of CodeBreaK 101, a phase Ib study of pts with chemorefractory KRAS G12C-mutated CRC receiving sotorasib alone or in combination with EGFR inhibitors, showed an ORR of 30% with sotorasib–panitumumab compared with 9.7% with sotorasib monotherapy.42,43 Subsequently, the CodeBreaK 300 trial evaluated two doses of sotorasib (960 mg once daily and 240 mg once daily) in combination with panitumumab versus standard-of-care trifluridine/tipiracil in chemorefractory metastatic CRC (mCRC) and showed an mPFS of 5.6 and 3.9 months in the 960 mg sotorasib–panitumumab group and 240 mg in the sotorasib–panitumumab group compared with 2.2 months in the standard-care group. ORR was 26.4, 5.7 and 0% for the 960 mg sotorasib–panitumumab, 240 mg sotorasib–panitumumab and standard-care groups, respectively.42 Of note, the phase I–II KRYSTAL-1 trial assessed adagrasib ± cetuximab in the same setting, showing an ORR of 34% (95% CI, 24.6–44.5) versus 21.4% (95% CI, 10.3–36.8) in favour of the combination treatment with a similar DCR (85.1 versus 86.2%) between the two arms. The combination confirmed its superiority in all the other efficacy outcomes, with an mPFS of 6.9 months (95% CI, 5.7–7.4) versus 4.1 months (95% CI, 2.8–6.5). mOS was 15.0 months (95% CI, 11.8–18.8) versus 12.2 months (95% CI, 8.1–15.2). Interestingly, the combination was more tolerated with 27% of grade 3–4 treatment-related adverse events (TRAEs) versus 34%, respectively.44 Fuelled by these positive results, the KRYSTAL-10 (ClinicalTrials.gov identifier: NCT04793958), a global, open-label, randomized phase III study, evaluates adagrasib and cetuximab against chemotherapy in the same patient cohort.45 On the other hand, the use of KRAS G12C inhibitors remains limited in other GI cancers. The prevalence of KRAS G12C varies across GI malignancies: it is limited in BTC, accounting for only 1% of mutations, and is slightly more prevalent in appendiceal cancers (3–4%).10 A cohort of KRYSTAL-1 focusing on non-CRC GI cancers evaluated 57 pts, amongst whom 12 had BTC and 16 had other GI tumours (nine pts had appendiceal cancer, four had gastro-oesophageal junction/oesophageal cancer and three had small bowel cancer). Updated results published in May 2023 demonstrated an ORR of 35.1%, a DCR of 86.0% and a median duration of response of 5.3 months (95% CI, 2.8–7.3). mPFS was 7.4 months (95% CI, 5.3–8.6) and mOS was 14.0 months (95% CI, 8.5–18.6). Interestingly, in pts with BTC, ORR was 41.7%, DCR was 91.7%, mPFS was 8.6 months (95% CI, 2.7–11.3) and mOS was 15.1 months (95% CI, 8.6–not estimable).41

Emergence of resistance

Despite the initial clinical responses achieved with KRAS G12C inhibitors, treatment failure eventually occurs due to the emergence of resistance. Resistance mechanisms can be divided into genetic when a mutation is identified in genes involved in RAS-dependent molecular pathways and adaptive when reactivation of upstream proteins occurs. According to paired plasma sample analyses, genetically acquired mutations have been detected in 90% of mCRC pts treated with the new KRAS G12C inhibitor divarasib with or without cetuximab.46 The most common on- and off-target alterations detected include new activating KRAS mutations, upstream RTKs hyperactive alterations (EGFR and FGFRs) and aberrations associated with downstream proteins (PIK3CA, RAF and MEK). Similarly, pts treated with adagrasib with or without cetuximab, for whom genomic-acquired mechanisms of resistance were detectable in more than 70% of pts, the newly acquired mutations affected both downstream signalling proteins, such as mitogen-activated protein kinase (MAPK) and PI3K, and upstream RTKs.47 Adaptive resistance most commonly manifests through the reactivation of upstream RTKs in response to KRAS G12C inhibition, limiting the efficacy of targeted drug therapies.48 Mutant KRAS G12C suppresses the activation of upstream RTKs and other wild-type RAS isoforms through ERK-mediated feedback inhibition. Therefore, using an agent stabilizing KRAS G12C protein in its ‘off’ state relieves this suppression with a consequent upregulation of RTKs, ultimately leading to the activation of wild-type RAS isoforms. This circuit was demonstrated in two studies by Ryan et al., where treatment with KRAS G12C inhibitors resulted in a rapid rebound increase in MAPK signalling as a result of the increased expression of multiple upstream RTKs, including EGFR, HER2 and FGFR.49,50 Uncovering these resistance mechanisms has fuelled the development of novel targeted therapeutic agents, which are under clinical investigation as both single-agent and combined strategies with other agents. Several promising therapies are currently undergoing preclinical or early-phase trials within multicohort studies, with the most relevant highlighted in Table 2.p28,36,51–65

Table 2: Comprehensive list of new Kirsten rat sarcoma virus G12C ‘off’ inhibitors and their treatment-related adverse events28,36,51–65

Agent

Pharmaceutical company

Clinical trial and identifier

Incidence of grade ≥3 TRAEs

Most commonly reported grade ≥3 TRAEs

LY353798251

Eli Lilly and Company

Phase I (NCT04956640)

NA

Diarrhoea, constipation, fatigue, peripheral oedema, nausea and neutropenia

GDC-603628

Genentech

Phase I (NCT04449874)

12%

Increased ALT, increased AST, nausea, vomiting and fatigue

D-155352

InventisBio

Phase I/II (NCT04585035)

22%

Increased AST and ALT, diarrhoea, hypertension, hypokalaemia and nausea

HBI-243853

Huyabio International

Phase I (NCT05485974)

Pending

Pending

JDQ44354

Novartis

Phase Ib/II (NCT04699188)

7.1%

Neutropenia, increased ALT and AST and myalgia

JAB-2182255

Jacobio Pharma

Phase I/II (NCT05009329)

0%

NA

HS-1037036

Jiangsu Hansoh Pharmaceutical Company

Phase I/II (NCT05367778)

27.3%

Increased AST and ALT, anaemia, diarrhoea, weight gain, decreased appetite, hypoproteinaemia, nausea, fatigue and rash

IBI-351 (GFH925)56

Innovent Biologics Inc.

Phase I/II (NCT05005234)

20%

Anaemia, leukopaenia, increased ALT and pruritus

BI-182391157

Boehringer Ingelheim

Phase I (NCT04973163)

30%

Nausea, diarrhoea, vomiting, fatigue and decreased appetite

JNJ-7469915758

Johnson & Johnson

Phase I (NCT04006301)

55%

Increased blood CPK

GFH92556

GenFleet

Pooled analysis of two phase I studies (NCT05005234 and NCT05497336)

20%

Anaemia, decreased white blood cell count, increased ALT and pruritus

YL-1529359

Shanghai Yingli

Phase I (NCT05173805)

NA

NA

BPI042128660

Belta

Phase I (NCT05315180)

Pending

Pending

GH3561

Suzhou Genhouse Bio

Phase I (NCT05010694)

Pending

Pending

GEC25562

GenEros Biopharma

Phase I

6.7%

Diarrhoea, increased ALT, rash and anaemia

MK-108463

Merck

Phase I (NCT05067283)

Pending

Pending

D3S-00164

D3 Bio

Phase I

14.6%

NA

HBI-243866

Huyabio

Phase I (NCT05485974)

Pending

Pending

SY-593365

Shouyao Holdings

Phase I (NCT06006793)

Pending

Pending

ALT = alanine aminotransferase;AST = aspartate aminotransferase;CPK = blood creatinine phosphokinase;NA = not available;TRAEs = treatment-related adverse events.

New direct inhibitors of Kirsten rat sarcoma virus G12C

Since the FDA approved adagrasib and sotorasib, several KRAS G12C inhibitors with different safety profiles are entering clinical trials. Divarasib is a KRAS G12C inhibitor that binds to the KRAS G12C protein in its ‘off’ state. It is, however, up to 50 times more selective and 5–20 times more potent than sotorasib and adagrasib according to in vitro studies.33 Divarasib monotherapy was evaluated in a phase I trial enrolling 137 pts, 55 of whom had mCRC.32 No dose-limiting toxicity occurred across concentrations ranging from 50 to 400 mg, and grade ≥3 TRAEs were observed in 12% of pts, leading to treatment discontinuation in 3% of cases. Amongst patients in the CRC cohort treated with divarasib 400 mg daily, 35.9% had a confirmed response (complete response [CR] or PR) and an mPFS of 6.9 months. More recently, the combination of divarasib with cetuximab achieved an improved ORR of 62.5% (95% CI, 40.6–81.2%) and an mPFS of 8.1 months (95% CI, 5.5–12.3) in pts with mCRC enrolled in a phase Ib trial (A Phase Ia/Ib Dose-escalation and Dose-expansion Study Evaluating the Safety, Pharmacokinetics, and Activity of GDC-6036 as a Single Agent and in Combination with Other Anti-cancer Therapies in Patients with Advanced or Metastatic Solid Tumors with a KRAS G12C Mutation; ClinicalTrials.gov identifier: NCT04449874).67 In a limited PDAC cohort containing seven pts receiving divarasib, three achieved PR, while four had stable disease (SD). Similarly, amongst seven pts with BTC, one patient had a PR, four had SD and one had progressive disease (PD).46 Therefore, further assessment of divarasib in PDAC and BTC is warranted in more extensive randomized trials. Divarasib is under evaluation in two phase I basket trials (ClinicalTrials.gov identifiers: NCT04449874 and NCT04929223), exploring its efficacy in combination with cetuximab with or without cytotoxic chemotherapy.68,69 Garsorasib is another orally bioavailable small-molecule inhibitor of KRAS G12C shown to be highly potent in vivo using cell line-derived and patient-derived xenograft tumour models, as well as in PDAC and CRC in vitro cell lines. In CRC patient-derived xenografts models, single-agent demonstrated tumour growth inhibition ranging from 60.9 to 105.7%, with three out of nine models showing tumour regression.70 Combining garsorasib with other anti-cancer agents, such as MEK inhibitors, tyrosine phosphatase-2 (Src Homologous Protein 2 [SHP2]) inhibitors and chemotherapy, increased its anti-tumoural activity and enhanced tumour regression.71 The efficacy of garsorasib was evaluated in combination with cetuximab in an ongoing phase II trial (ClinicalTrials.gov identifier: NCT04585035) in pts with refractory mCRC. Preliminary results demonstrated an ORR of 45% and a DCR of 95%. However, data on overall survival are still pending.70–72 Similarly, amongst 10 pts with advanced PDAC who received garsorasib, ORR was 50%, including one CR, DCR was 80% and mPFS was 8.54 months.73 It is essential to mention that in this trial, only 40% of pts received two or more lines of therapy.

Son of Sevenless 1 and Src Homologous Protein 2 inhibitors

Son of Sevenless 1 (SOS1) and SHP2 are signalling intermediates activated by RTKs that act as central nodes in the RAS signalling pathway. SOS1 is a GEF protein that binds inactive KRAS–GDP complex and mediates GTP exchange. SHP2, an adaptive phosphatase, links directly to SOS1, further facilitating GTP exchange. Therefore, inhibiting both SOS1 and SHP2 blocks RTK-positive molecular signalling to KRAS, maintaining it in its inactive GDP state. Published data demonstrated that co-inhibition of SHP2 and KRAS G12C overturns feedback reactivation across different RTKs and that combined KRAS G12C–SHP2 inhibition maintains RAS pathway suppression with improved efficacy both in vitro and in vivo.49,50 Since new activating RTK alterations were also commonly detected in liquid biopsies at the time of radiological treatment progression, approaches that target KRAS activation via SOS1 or SHP2 inhibitors represent a compelling avenue for intervention. The combination of glecirasib with JAB-3312, an SHP2 inhibitor, in pts with NSCLC with no prior exposure to KRAS G12C inhibitors demonstrated an ORR of 50% and a DCR of 100%, albeit with increased toxicity.74 RMC-4630 is another SHP2 inhibitor being examined with sotorasib in the CodeBreaK 101 trial.75 In a cohort of six pts, five (83%) achieved disease control; however, no responses were observed.

Downstream mitogen-activated protein kinase blockade

As mentioned previously, adaptive and acquired resistance mechanisms may involve upregulation of the RTK/RAS/MAPK downstream signalling pathway, hence the rationale behind using RAS downstream inhibitors. A well-described example is the upregulation of EGFR that occurs with B-Raf murine sarcoma viral oncogene homologue B (BRAF) inhibition in BRAF-mutated CRC, necessitating the addition of EGFR inhibitors to counteract drug resistance. Another example is the compensatory activation of the mTOR pathway, which has been documented in KRAS G12C inhibitor-resistant CRC cell lines.76 Currently, mTOR inhibitors such as everolimus are being explored in combination with sotorasib in NSCLC, while relevant phase I basket trials are investigating combinations such as adagrasib with nab-sirolimus (ClinicalTrials.gov identifier: NCT05840510) and divarasib with the PI3K inhibitor inavolisib (ClinicalTrials.gov identifier: NCT04449874) in pts with various solid tumours, including CRC.68,77 Moreover, the addition of MEK inhibitors to G12C inhibitors is being tested in the CodeBreaK 101 study as a new alternative, based on preclinical studies showing that sotorasib and trametinib (MEK inhibitor) combination has a synergistic anti-tumour effect on NSCLC tumour cell lines in vitro compared with either of the single agents alone.78 In this context, sotorasib and trametinib (MEK inhibitor) were evaluated in 36 pts, 18 of whom have KRAS G12C-mutant CRC, achieving a remarkable DCR of 86%.79 It is worth noting that some pts had prior exposure to KRAS G12C inhibitors, and upon receiving the maximum tolerated dose of 2 mg trametinib and 960 mg sotorasib, all pts had radiological SD. These results, albeit reported in a limited number of pts, indicate a clinically significant therapeutic benefit associated with the addition of trametinib to sotorasib. Of note, this trial reported 34% of pts with grade ≥3 toxicity, leading to therapy discontinuation in 24% of cases.

Combination with immunotherapy

The tumour microenvironment (TME) in PDAC is a complex and dynamic cell network involving immune, stromal and cancer cells, facilitating tumour progression and response to therapy. KRAS mutations have been widely shown to foster anti-inflammatory and pro-inflammatory effects on the TME. Some studies underpinned that KRAS mutations promote an immunosuppressive TME through several mechanisms such as tumour cell expression of inhibitory cytokines interleukin (IL)-10 and transforming growth factor-β, recruitment of myeloid-derived suppressor cells, regulatory T-cell (T-reg cell) activation and suppression of cluster of differentiation 8 (CD8) T-cell activity.21,80 Other studies suggest that KRAS mutations could also interfere with the secretion of pro-inflammatory cytokines, such as intercellular adhesion molecule-1, tumour necrosis factor-α, IL-1β, IL-6 and IL-18, through the induction of NF-κB.81–83 On the other hand, adagrasib and sotorasib were found to induce a pro-inflammatory microenvironment and modulate the TME by recruiting macrophages, dendritic cells and CD8 T-cells, promoting anti-tumour immune response in NSCLC. These findings suggest a potential synergistic interaction between these agents and immune checkpoint inhibitors (ICIs).84,85 However, conflicting data have emerged regarding the efficacy and tolerability of these combined regimens. Early data from the CodeBreaK 101 study, in which pts with NSCLC received sotorasib plus pembrolizumab or atezolizumab, showed a high incidence of liver toxicity, with an ORR of 29% and a DCR of 83%.86 However, results from the KRYSTAL-7 (ClinicalTrials.gov identifier: NCT04613596) study combining adagrasib and pembrolizumab in pts with NSCLC who had programmed death-ligand 1 expression >50% demonstrated an ORR of 63% and a DCR of 84%, with limited high-grade toxicity (10%).87 The combination of KRAS G12C inhibitors with ICIs and other target therapies is currently being assessed in several clinical trials across other KRAS G12C-mutant solid tumours, PDAC and CRC (Table 3).40–42,44–46,51,56–58,67,69,72,74,75,79,88–107

Table 3: Summary of clinical trials testing Kirsten rat sarcoma virus G12C inhibitors as monotherapy or in combination with other agents in gastrointestinal cancers40–42,44–46,51,56–58,67,69,72,74,75,79,88–107

Target

Drugs

Trial name and identifier

Phase

GI malignancy

Number of pts in the trial, if applicable

Clinical efficacy

KRAS G12C ‘off’ inhibitors

Adagrasib41

KRYSTAL-1 (NCT03785249)

I/II

PDAC

21

ORR: 33%

DCR: 49%

mPFS: 5.4 months

mOS: 8.0 months

CRC

43

ORR: 19%

DCR: 86%

mPFS: 5.6 months

mOS: 19.8 months

BTC

12

ORR: 42%

DCR: 92%

Appendiceal adenocarcinoma

7

ORR: 0%

DCR: 86%

GEA

3

ORR: 33%

DCR: 66%

Adagrasib+cetuximab44,45

NCT05634525

I

PDAC

Pending

Pending

KRYSTAL-1 (NCT03785249)

I/II

CRC

28

ORR: 46%

DCR: 100%

mPFS: 6.9 months

mOS: 13.4 months

Adagrasib+cetuximab+irinotecan88

NCT05722327

I

CRC

Pending

Pending

Adagrasib+cetuximab versus chemotherapy89

KRYSTAL-10 (NCT04793958)

III

CRC, second line

Pending

Pending

Adagrasib+TNO15590

KRYSTAL-2 (NCT04330664)

I/II

CRC

Pending

Pending

Adagrasib+BI 170196391

KRYSTAL-14 (NCT04975256)

I

CRC

Pending

Pending

Adagrasib+MRTX090292

NCT05578092

I/II

NA

Pending

Pending

Adagrasib+durvalumab93

NCT05848843

I

CRC

Pending

Pending

Adagrasib+INCB09928094

NCT06039384

I

CRC

Pending

Pending

Sotorasib40,42

CodeBreaK 100 (NCT03600883)

I/II

PDAC

38

ORR: 21%

DCR: 84%

mPFS: 4.0 months

mOS: 6.9 months

CRC

62

ORR = 9.7%

DCR = 82%

Sotorasib+panitumumab95

CodeBreaK 101 (NCT04185883)

I/II

CRC

40

ORR: 30%

DCR: 93%

mPFS: 5.7 months

Sotorasib+panitumumab+FOLFIRI96

CodeBreaK 101 (NCT04185883)

I/II

CRC

31

ORR: 58.1%

Sotorasib+trametinib79

CodeBreaK 101 (NCT04185883)

I/II

CRC

18

ORR: 11.1%

DCR: 83.3%

Sotorasib+panitumumab versus standard of care97

CodeBreaK 300 (NCT05198934)

III

CRC

ORR: 26.4 versus 0% (SOC)

DCR: 71.7 versus 46.3% (SOC)

mPFS: 5.6 versus 2.2 months (SOC)

Sotorasib+BI 170196398

CodeBreaK 101 (NCT04185883)

I/II

NA

Pending

Pending

Sotorasib+RMC-463075

CodeBreaK 101 (NCT04185883)

I/II

CRC

 6

ORR: 0%

DCR: 83.3%

Sotorasib+BBP-39899

NCT05480865

I/II

NA

Pending

Pending

BI1823911 monotherapy+BI 170196357

NCT04973163

I

NA

Pending

Pending

Divarasib46

NCT04449874

I

CRC

55

ORR: 29%

DCR: 85%

mPFS: 5.6 months

PDAC

7

ORR: 43%

DCR: 100%

BTC

5

ORR: 0%

DCR: 80%

Divarasib+cetuximab67

NCT04449874

I

CRC

29

ORR: 62.5%

Divarasib+cetuximab+FOLFOX or FOLFIRI69

INTRINSIC (NCT04929223)

I

CRC

Pending

Pending

Fulzerasib56

Pooled analysis of NCT05005234 and NCT05497336

I

CRC

45

ORR 43.8%

DCR 87.5%

Garsorasib72

NCT04585035

I/II

CRC

24

ORR: 21%

DCR 95.8%

mPFS: 7.6 months

Garsorasib72

NCT04585035

I/II

PDAC

10

ORR: 50%

DCR: 80%

mPFS: 8.5 months

Garsorasib+cetuximab72

NCT04585035

I/II

CRC

40

ORR: 45%

DCR: 95%

mPFS 7.6 months

Glecirasib101

NCT05009329 and NCT05002270

I/II

PDAC, biliary tract, gastric, small bowel, appendiceal, hepatocellular, peritoneal

PDAC = 28

Others = 19

ORR: 46.4%

DCR: 96.4%

mPFS: 5.5 months

Glecirasib+cetuximab102

NCT05002270 and NCT05194995

I/II

CRC, small bowel and appendiceal adenocarcinoma

43

ORR: 62.8%

DCR: 93%

mPFS: Not reached

Glecirasib103

NCT06008288

II

PDAC

Pending

Pending

Glecirasib+JAB-331274

NCT05288205

I/II

CRC and PDAC

Pending

Pending

JDQ443 monotherapy+TNO155 or +tislelizumab104

KontRASt-01 (NCT04699188)

I/II

CRC

Pending

Pending

JDQ443+trametinib+ribociclib or + cetuximab105

KontRASt-03 (NCT05358249)

I/II

CRC

Pending

Pending

JNJ-7469915758

NCT04006301

I

CRC

Withdrawn from market

LY353798251

LOXO-RAS-20001 (NCT04956640)

I/II

PDAC

12

ORR: 42%

DCR: 92%

CRC

20

ORR: 10%

DCR: 90%

Others

21

ORR: 52%

DCR: 95%

LY3537982+cetuximab51

LOXO-RAS-20001 (NCT04956640)

I/II

CRC

11

ORR: 45%

DCR: 100%

KRAS G12C ‘on’ inhibitors

BBO-8520106

Preclinical

RMC-6291107

NCT05462717

I

CRC

20

ORR = 40%

DCR = 80%

BTC = biliary tract cancer; CRC = colorectal cancer; DCR = disease control rate; FOLFIRI = leucovorin calcium (folinic acid)–fluorouracil–irinotecan hydrochloride; FOLFOX = leucovorin calcium (folinic acid)–fluorouracil–oxaliplatin; GEA = gastro-oesophageal cancer; GI = gastrointestinal;KRAS = Kirsten rat sarcoma virus;mOS = median overall survival; mPFS = median progression-free survival; NA = not available; ORR = overall response rate; PDAC = pancreatic ductal adenocarcinoma;pts = patients; SOC = standard of care.

Conclusion

Since the first demonstration of sotorasib efficacy with the published results from the CodeBreaK 100 trial in 2019, the current landscape of RAS targeting strategy has shifted from undruggable to druggable. Fuelled by the successful implementation of adagrasib and sotorasib as standard treatment options in PDAC, and recently in BTC and CRC, more than a dozen small-molecule inhibitors targeting KRAS G12C and related upstream and downstream molecules have emerged. They are actively investigating, with many of them already advanced to clinical trials for evaluation in GI cancers. However, despite the crucial development and relative success of KRAS G12C inhibitors in treating GI cancers, clinical research must still prioritize strategies to overcome primary and acquired resistance, primarily through vertical inhibition with RTK/RAS/MAPK pathway inhibitors and combination with immunotherapy or standard chemotherapy. In addition, more data about toxicity management and quality of life are awaited to ensure the safety of new drugs and combinations under evaluation. Other important considerations will be integrating KRAS G12C inhibitors in earlier phases of treatment, such as in the neoadjuvant and adjuvant settings. These strategies may expand the population of pts with GI cancers, benefitting from the inhibition of KRAS G12C. Undoubtedly, we are in a new and promising era of cancer treatment, pushing the boundaries of KRAS target therapy.

3

References

List View
Grid View
1
Copy DOIDOI Copied
Visit DOI Link

 Prior IAHood FEHartley JLThe frequency of Ras mutations in cancerCancer Res2020;80:296974DOI10.1158/0008-5472.CAN-19-3682.

2
Copy DOIDOI Copied
Visit DOI Link

 Cancer Genome Atlas NComprehensive molecular characterization of human colon and rectal cancerNature2012;487:3307. DOI10.1038/nature11252.

3
Copy DOIDOI Copied
Visit DOI Link

 Miyashita HKato SHong DSKRAS G12C inhibitor combination therapies: Current evidence and challengeFront Oncol. 2024;14:1380584. DOI10.3389/fonc.2024.1380584.

4
Copy DOIDOI Copied
Visit DOI Link

 Singhal ALi BTO’Reilly EMTargeting KRAS in cancerNat Med2024;30:96983DOI10.1038/s41591-024-02903-0.

5
Copy DOIDOI Copied
Visit DOI Link

 Ros JVaghi CBaraibar Iet alTargeting KRAS G12C mutation in colorectal cancer, a review: New arrows in the quiverInt J Mol Sci.. 2024;25:3304. DOI10.3390/ijms25063304.

6
Copy DOIDOI Copied
Visit DOI Link

 Hunter JCManandhar ACarrasco MAet alBiochemical and structural analysis of common cancer-associated KRAS mutationsMol Cancer Res2015;13:132535. DOI10.1158/1541-7786.MCR-15-0203.

7
Copy DOIDOI Copied
Visit DOI Link

 Salem MEl-Refai SSha Wet alO-3 characterization of KRAS mutation variants and prevalence of KRAS-G12C in gastrointestinal malignancies. Ann Oncol. 2021;32:S218. DOI10.1016/j.annonc.2021.05.007.

8
Copy DOIDOI Copied
Visit DOI Link

 Lee JKSivakumar SSchrock ABet alComprehensive pan-cancer genomic landscape of KRAS altered cancers and real-world outcomes in solid tumorsNPJ precision oncology2022;6:91DOI10.1038/s41698-022-00334-z.

9
Copy DOIDOI Copied
Visit DOI Link

 Aaltonen LAAbascal FAbeshouse Aet alPan-cancer analysis of whole genomesNature2020;578:8293DOI10.1038/s41586-020-1969-6.

10
Copy DOIDOI Copied
Visit DOI Link

 Salem MEEl-Refai SMSha Wet alLandscape of KRAS(G12C), associated genomic alterations, and interrelation with immuno-oncology biomarkers in KRAS-mutated cancersJCO Precis Oncol2022;6:e2100245DOI10.1200/PO.21.00245.

11
Copy DOIDOI Copied
Visit DOI Link

 Strickler JHYoshino TStevinson Ket alPrevalence of KRAS G12C mutation and co-mutations and associated clinical outcomes in patients with colorectal cancer: A systematic literature reviewOncologist2023;28:e98194DOI10.1093/oncolo/oyad138.

12
Copy DOIDOI Copied
Visit DOI Link

 Jones RPSutton PAEvans JPet alSpecific mutations in KRAS codon 12 are associated with worse overall survival in patients with advanced and recurrent colorectal cancerBr J Cancer2017;116:9239DOI10.1038/bjc.2017.37.

13
Copy DOIDOI Copied
Visit DOI Link

 Fakih MTu HHsu Het alReal-world study of characteristics and treatment outcomes among patients with KRAS p.G12C-mutated or other KRAS mutated metastatic colorectal cancerOncologist2022;27:66374DOI10.1093/oncolo/oyac077.

14
Copy DOIDOI Copied
Visit DOI Link

 van de Haar JMa XOoft SNet alCodon-specific KRAS mutations predict survival benefit of trifluridine/tipiracil in metastatic colorectal cancerNat Med2023;29:60514DOI10.1038/s41591-023-02240-8.

15
Copy DOIDOI Copied
Visit DOI Link

 Boilève ARousseau AHilmi Met alCodon-specific KRAS mutations predict survival in advanced pancreatic cancerESMO Gastrointest Oncol2024;3:100030DOI10.1016/j.esmogo.2023.100030.

16
Copy DOIDOI Copied
Visit DOI Link

 Ostrem JMLShokat KMDirect small-molecule inhibitors of KRAS: From structural insights to mechanism-based designNat Rev Drug Discov2016;15:77185DOI10.1038/nrd.2016.139.

17
Copy DOIDOI Copied
Visit DOI Link

 Hymowitz SGMalek STargeting the MAPK pathway in RAS mutant cancersCold Spring Harb Perspect Med. 2018;8:a031492. DOI10.1101/cshperspect.a031492.

18
Copy DOIDOI Copied
Visit DOI Link

 Erijman AShifman JMRAS/effector interactions from structural and biophysical perspectiveMini Rev Med Chem2016;16:3705. DOI10.2174/1389557515666151001141838.

19
Copy DOIDOI Copied
Visit DOI Link

 Glaviano AFoo ASLam HYet alPI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancerMol Cancer. 2023;22:138.

20
Copy DOIDOI Copied
Visit DOI Link

 Kim DXue JYLito PTargeting KRAS(G12C): From inhibitory mechanism to modulation of antitumor effects in patients. Cell. 2020;183:8509. DOI10.1016/j.cell.2020.09.044.

21
Copy DOIDOI Copied
Visit DOI Link

 Liu PWang YLi XTargeting the untargetable KRAS in cancer therapyActa Pharmaceutica Sinica B2019;9:8719. DOI10.1016/j.apsb.2019.03.002.

22
Copy DOIDOI Copied
Visit DOI Link

 Ryan MBCorcoran RBTherapeutic strategies to target RAS-mutant cancersNat Rev Clin Oncol2018;15:70920.

23
Copy DOIDOI Copied
Visit DOI Link

 Cox ADFesik SWKimmelman ACet alDrugging the undruggable RAS: Mission possible? Nat Rev Drug Discov2014;13:82851. DOI10.1038/nrd4389.

24
Copy DOIDOI Copied
Visit DOI Link

 Papke BDer CJDrugging RASKnow the enemyScience2017;355:115863.

25
Copy DOIDOI Copied
Visit DOI Link

 Lu SJang HNussinov RZhang JThe structural basis of oncogenic mutations G12, G13 and Q61 in small GTPase K-RAS4bSci Rep. 2016;6:21949. DOI10.1038/srep21949.

26
Copy DOIDOI Copied
Visit DOI Link

 Traut TWPhysiological concentrations of purines and pyrimidinesMol Cell Biochem1994;140:122DOI10.1007/BF00928361.

27
Copy DOIDOI Copied
Visit DOI Link

 John JSohmen RFeuerstein Jet alKinetics of interaction of nucleotides with nucleotide-free H-RAS p21Biochemistry1990;29:605865. DOI10.1021/bi00477a025.

28
Copy DOIDOI Copied
Visit DOI Link

 Schram AMGandhi LMita MMet alA phase Ib dose-escalation and expansion study of the oral MEK inhibitor pimasertib and PI3K/mTOR inhibitor voxtalisib in patients with advanced solid tumoursBr J Cancer2018;119:14716DOI10.1038/s41416-018-0322-4.

29
Copy DOIDOI Copied
Visit DOI Link

 Liu WYin YWang Jet alKRAS mutations increase telomerase activity and targeting telomerase is a promising therapeutic strategy for KRAS-mutant NSCLCOncotarget2017;8:17990DOI10.18632/oncotarget.10162.

30
Copy DOIDOI Copied
Visit DOI Link

 Srikar RSuresh DZambre Aet alTargeted nanoconjugate co-delivering siRNS and tyrosine kinase inhibitor to KRAS mutant NSCLC dissociates GAB1-SHP2 post oncogene knockdownSci Rep2016;6:30245DOI10.1038/srep30245.

31
Copy DOIDOI Copied
Visit DOI Link

 Puyol MMartín ADubus Pet alA synthetic lethal interaction between K-RAS oncogenes and CDK4 unveils a therapeutic strategy for non-small cell lung carcinomaCancer Cell2010;18:6373DOI10.1016/j.ccr.2010.05.025.

32
Copy DOIDOI Copied
Visit DOI Link

 Dearden SStevens JWu Y-Let alMutation incidence and coincidence in non small-cell lung cancer: Meta-analyses by ethnicity and histology (mutMap)Ann Oncol2013;24:23716DOI10.1093/annonc/mdt205.

33
Copy DOIDOI Copied
Visit DOI Link

 Purkey HAbstract ND11: Discovery of GDC-6036, a clinical stage treatment for KRAS G12C-positive cancersCancer Res. 2022;82(Suppl.12):Abstr.ND11. DOI10.1158/1538-7445.AM2022-ND11.

34
Copy DOIDOI Copied
Visit DOI Link

 Hallin JEngstrom LDHargis Let alThe KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of KRAS-mutant cancers in mouse models and patientsCancer Discov2020;10:5471DOI10.1158/2159-8290.CD-19-1167.

35
Copy DOIDOI Copied
Visit DOI Link

 Rubinson DATanaka NFece de la Cruz Fet alSotorasib is a pan-RASG12C inhibitor capable of driving clinical response in NRASG12C cancersCancer Discov2024;14:72736DOI10.1158/2159-8290.CD-23-1138.

36
Copy DOIDOI Copied
Visit DOI Link

 Dong XMeng XZhang Yet alAbstract CT119: Safety and efficacy of HS-10370 in KRAS G12C-mutated solid tumors including non-small cell lung cancer (NSCLC)Cancer Res2024;84(Suppl.):Abstr.CT119DOI10.1158/1538-7445.AM2024-CT119.

37
Copy DOIDOI Copied
Visit DOI Link

 US FDAFDA grants accelerated approval to sotorasib for KRAS G12C mutated NSCLC2021Available atwww.fda.gov/drugs/resources-information-approved-drugs/fda-grants-accelerated-approval-sotorasib-kras-g12c-mutated-nsclc (accessed24 October 2024).

38
Copy DOIDOI Copied
Visit DOI Link

 Janne PARiely GJGadgeel SMet alAdagrasib in non-small-cell lung cancer harboring a KRAS(G12C) mutationN Engl J Med. 2022;387:12031.

39
Copy DOIDOI Copied
Visit DOI Link

 Hong DSFakih MGStrickler JHet alKRAS(G12C) inhibition with sotorasib in advanced solid tumorsN Engl J Med2020;383:120717. DOI10.1056/NEJMoa1917239.

40
Copy DOIDOI Copied
Visit DOI Link

 Strickler JHSatake HGeorge TJet alSotorasib in KRAS p.G12C–mutated advanced pancreatic cancerN Engl J Med2023;388:3343. DOI10.1056/NEJMoa2208470.

41
Copy DOIDOI Copied
Visit DOI Link

 Pant SYaeger RSpira AIet alKRYSTAL-1: activity and safety of adagrasib (MRTX849) in patients with advanced solid tumors harboring a KRAS g12c mutationJ Clin Oncol2023;41(Suppl.):Abstr.425082DOI10.1200/JCO.2023.41.36_suppl.425082.

42
Copy DOIDOI Copied
Visit DOI Link

 Fakih MGKopetz SKuboki Yet alSotorasib for previously treated colorectal cancers with KRASG12C mutation (CodeBreaK100): A prespecified analysis of a single-arm, phase 2 trialLancet Oncol2022;23:11524DOI10.1016/S1470-2045(21)00605-7.

43
Copy DOIDOI Copied
Visit DOI Link

 Skoulidis FLi BTDy GKet alSotorasib for lung cancers with KRAS pG12C mutationN Engl J Med2021;384:237181. DOI10.1056/NEJMoa2103695.

44
Copy DOIDOI Copied
Visit DOI Link

 Yaeger RUboha NVPelster MSet alEfficacy and safety of adagrasib plus cetuximab in patients with KRASG12C-mutated metastatic colorectal cancerCancer Discov2024;14:98293DOI10.1158/2159-8290.CD-24-0217.

45
Copy DOIDOI Copied
Visit DOI Link

 Kopetz SDr Kopetz on the future utility of adagrasib/cetuximab in KRAS G12C–mutant CRC2024Available atwww.onclive.com/view/dr-kopetz-on-the-future-utility-of-adagrasib-cetuximab-in-kras-g12c-mutant-crc (accessed24 October 2024).

46
Copy DOIDOI Copied
Visit DOI Link

 Sacher ALoRusso PPatel MRet alSingle-agent divarasib (GDC-6036) in solid tumors with a KRAS G12C mutationN Engl J Med. 2023;389:71021. DOI10.1056/NEJMoa2303810.

47
Copy DOIDOI Copied
Visit DOI Link

 Awad MMLiu SRybkin IIet alAcquired resistance to KRAS(G12C) inhibition in cancerN Engl J Med2021;384:238293.

48
Copy DOIDOI Copied
Visit DOI Link

 Amodio VYaeger RArcella Pet alEGFR blockade reverts resistance to KRASG12C inhibition in colorectal cancerCancer Discov. 2020;10:112939. DOI10.1158/2159-8290.CD-20-0187.

49
Copy DOIDOI Copied
Visit DOI Link

 Ryan MBFece de la Cruz FPhat Set alVertical pathway inhibition overcomes adaptive feedback resistance to KRASG12C inhibitionClin Cancer Res2020;26:163343DOI10.1158/1078-0432.CCR-19-3523.

50
Copy DOIDOI Copied
Visit DOI Link

 Ryan MBCoker OSorokin Aet alKRASG12C-independent feedback activation of wild-type RAS constrains KRASG12C inhibitor efficacyCell Reports2022;39:110993DOI10.1016/j.celrep.2022.110993.

51
Copy DOIDOI Copied
Visit DOI Link

 Hollebecque AKuboki YMurciano-Goroff YRet alEfficacy and safety of LY3537982, a potent and highly selective KRAS G12C inhibitor in KRAS G12C-mutant GI cancers: Results from a phase 1 studyJ Clin Oncol2024;42(Suppl.3):Abstr.94DOI10.1200/JCO.2024.42.3_suppl.94.

52
Copy DOIDOI Copied
Visit DOI Link

 Ruan DLee MADeng Yet alSafety and efficacy of D-1553 in KRAS G12C-mutated colorectal cancer: Results from a phase I/II studyJ Clin Oncol2023;41(Suppl.16):Abstr.3563DOI10.1200/JCO.2023.41.16_suppl.3563.

53
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Dose Escalation Study of HBI-2438 in Patients With Solid Tumors Harboring KRAS G12C Mutation. ClinicalTrials.gov identifier: NCT05485974. Available atwww.clinicaltrials.gov/study/NCT05485974 (accessed22 November 2024).

54
Copy DOIDOI Copied
Visit DOI Link

 Cassier PADooms CAGazzah Aet alKontRASt-01 update: Safety and efficacy of JDQ443 in KRAS G12C-mutated solid tumors including non-small cell lung cancer (NSCLC)J Clin Oncol2023;41(Suppl.16):Abstr.9007DOI10.1200/JCO.2023.41.16_suppl.9007.

55
Copy DOIDOI Copied
Visit DOI Link

 Li JZhao JCao Bet alA phase I/II study of first-in-human trial of JAB-21822 (KRAS G12C inhibitor) in advanced solid tumorsJ Clin Oncol. 2022;40(Suppl.16):Abstr.3089DOI10.1200/JCO.2022.40.16_suppl.3089.

56
Copy DOIDOI Copied
Visit DOI Link

 Yuan YDeng YJin Yet alEfficacy and safety of IBI351 (GFH925) monotherapy in metastatic colorectal cancer harboring KRAS g12c mutation: preliminary results from a pooled analysis of two phase I studiesJ Clin Oncol. 2023;41(Suppl.16):Abstr.3586. DOI: 10.1200/JCO.2023.41.16_suppl.3586.

57
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study to Test Different Doses of BI 1823911 Alone and Combined With Other Medicines in People With Different Types of Advanced Cancer With KRAS Mutation. ClinicalTrials.Gov Identifier: NCT04973163. Available athttps://clinicaltrials.gov/study/NCT04973163 (accessed22 November 2024).

58
Copy DOIDOI Copied
Visit DOI Link

 Wang JMartin-Romano PCassier Pet alPhase I study of JNJ-74699157 in patients with advanced solid tumors harboring the KRAS G12C mutationOncologist2022;27:536e553DOI10.1093/oncolo/oyab080.

59
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govPhase I Clinical Study of YL-15293 in Patients With Advanced Solid Tumor With KRAS Mutation. ClinicalTrials.gov identifier: NCT05173805. Available atwww.clinicaltrials.gov/study/NCT05173805 (accessed22 November 2024).

60
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Phase 1, Open-label Study of BPI-421286 in Subjects With Advanced Solid Tumors. ClinicalTrials.gov identifier: NCT05315180. Available atwww.clinicaltrials.gov/study/NCT05315180 (accessed22 November 2024).

61
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govGH35 Tablets for Advanced Solid Tumors: A Study on Safety and Early Results. ClinicalTrials.gov identifier: NCT05010694. Available athttps://clinicaltrials.gov/study/NCT05010694 (accessed22 November 2024).

62
Copy DOIDOI Copied
Visit DOI Link

 Zhang YZhou LGong Yet alPhase 1 study evaluating the safety, tolerability, pharmacokinetics (PK), and efficacy of GEC255, a novel KRAS g12c inhibitor, in advanced solid tumorsJ Clin Oncol2023;41(Suppl.16):Abstr.9112DOI10.1200/JCO.2023.41.16_suppl.9112.

63
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study of MK-1084 in KRAS Mutant Advanced Solid Tumors (MK-1084-001). ClinicalTrials.gov identifier: NCT05067283. Available athttps://clinicaltrials.gov/study/NCT05067283 (accessed22 November 2024).

64
Copy DOIDOI Copied
Visit DOI Link

 Cho BCLu SLee MAet alAbstract CT117: A phase 1 study of D3S-001, A second-generation GDP-bound KRAS G12C inhibitor, as monotherapy in patients with KRAS G12C-mutated solid tumorsCancer Res2024;84(Suppl.7):Abstr.CT117DOI10.1158/1538-7445.AM2024-CT117.

65
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study of SY-5933 in Patients With Advanced Solid Tumors Harboring the KRAS p.G12C Mutation. ClinicalTrials.gov identifier: NCT06006793. Available athttps://clinicaltrials.gov/study/NCT06006793 (accessed22 November 2024).

66
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Dose Escalation Study of HBI-2438 in Patients With Solid Tumors Harboring KRAS G12C Mutation. ClinicalTrials.gov identifier: NCT05485974. Available athttps://clinicaltrials.gov/study/NCT05485974 (accessed1 August 2022).

67
Copy DOIDOI Copied
Visit DOI Link

 Desai JAlonso GKim SHet alDivarasib plus cetuximab in KRAS G12C-positive colorectal cancer: A phase 1b trialNat Med2024;30:2718. DOI10.1038/s41591-023-02696-8.

68
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study to Evaluate the Safety, Pharmacokinetics, and Activity of GDC-6036 Alone or in Combination in Participants With Advanced or Metastatic Solid Tumors With A KRAS G12C Mutation. ClinicalTrials.gov identifier: NCT04449874. Available athttps://clinicaltrials.gov/study/NCT04449874 (accessed22 November 2024).

69
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study Evaluating the Safety and Efficacy of Targeted Therapies in Subpopulations of Patients With Metastatic Colorectal Cancer (INTRINSIC). ClinicalTrials.gov identifier: NCT04929223. Available athttps://clinicaltrials.gov/study/NCT04929223 (accessed22 November 2024).

70
Copy DOIDOI Copied
Visit DOI Link

 Shi ZWeng JFan Xet alAbstract 932: Discovery of D-1553, a novel and selective KRAS-G12C inhibitor with potent anti-tumor activity in a broad spectrum of tumor cell lines and xenograft modelsCancer Res2021;81(Suppl.13):Abstr.932DOI10.1158/1538-7445.AM2021-932.

71
Copy DOIDOI Copied
Visit DOI Link

 Shi ZWeng JFan Xet alAbstract 1056: potent in vivo anti-tumor activity of D-1553 as a single agent and in combination with targeted therapeutics in a broad spectrum of patient-derived xenograft tumor models with KRAS G12C mutationCancer Res. 2021;81(Suppl.13):Abstr.1056. DOI10.1158/1538-7445.AM2021-1056.

72
Copy DOIDOI Copied
Visit DOI Link

 Xu R-HXu YYan Det al550O Safety and efficacy of D-1553 in combination with cetuximab in KRAS G12C mutated colorectal cancer (CRC): A phase II studyAnn Oncol2023;34(Suppl.2):S4101DOI10.1016/j.annonc.2023.09.1741.

73
Copy DOIDOI Copied
Visit DOI Link

 Kondo SYan DGanju Vet al1622P D-1553 in patients with KRAS G12C mutated advanced pancreatic cancer (PCA)Ann Oncol. 2023;34(Suppl.2):S8989. DOI10.1016/j.annonc.2023.09.2571.

74
Copy DOIDOI Copied
Visit DOI Link

 Wang JZhao JZhong Jet al653O Glecirasib (KRAS G12C inhibitor) in combination with JAB-3312 (SHP2 inhibitor) in patients with KRAS p.G12C mutated solid tumorsAnn Oncol2023;34(Suppl.2):S459DOI10.1016/j.annonc.2023.09.1839.

75
Copy DOIDOI Copied
Visit DOI Link

 Falchook GLi BTMarrone KAet alOA03.03 Sotorasib in combination with RMC-4630, a SHP2 inhibitor, in KRAS p.G12C-mutated NSCLC and other solid tumorsJ Thorac Oncol2022;17(Suppl.):S8DOI10.1016/j.jtho.2022.07.022.

76
Copy DOIDOI Copied
Visit DOI Link

 Yaeger RMezzadra RSinopoli Jet alMolecular characterization of acquired resistance to KRASG12C-EGFR inhibition in colorectal cancerCancer Discov2023;13:4155DOI10.1158/2159-8290.CD-22-0405.

77
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govAdagrasib in Combination With Nab-Sirolimus in Patients With Advanced Solid Tumors and Non-Small Cell Lung Cancer With a KRAS G12C Mutation (KRYSTAL -19). ClinicalTrials.gov identifier: NCT05840510. Available athttps://clinicaltrials.gov/study/NCT05840510 (accessed22 November 2024).

78
Copy DOIDOI Copied
Visit DOI Link

 Canon JRex KSaiki AYet alThe clinical KRAS(G12C) inhibitor AMG 510 drives anti-tumour immunityNature2019;575:21723. DOI10.1038/s41586-019-1694-1.

79
Copy DOIDOI Copied
Visit DOI Link

 Ramalingam SFakih MStrickler Jet alAbstract P05-01: A phase 1b study evaluating the safety and efficacy of sotorasib: A KRASG12C inhibitor, in combination with trametinib, a MEK inhibitor, in KRAS p.G12C-mutated solid tumorsMol Cancer Ther2021;20(Suppl.12):P0501. DOI10.1158/1535-7163.TARG-21-P05-01.

80
Copy DOIDOI Copied
Visit DOI Link

 Zdanov SMandapathil MAbu Eid Ret alMutant KRAS conversion of conventional T cells into regulatory T cellsCancer Immunol Res. 2016;4:35465. DOI10.1158/2326-6066.CIR-15-0241.

81
Copy DOIDOI Copied
Visit DOI Link

 Hamarsheh SGroß OBrummer TZeiser RImmune modulatory effects of oncogenic KRAS in cancerNat Commun. 2020;11:5439. DOI10.1038/s41467-020-19288-6.

82
Copy DOIDOI Copied
Visit DOI Link

 Liou G-YDöppler HNecela Bet alMutant KRAS-induced expression of ICAM-1 in pancreatic acinar cells causes attraction of macrophages to expedite the formation of precancerous lesionsCancer Discov2015;5:5263DOI10.1158/2159-8290.CD-14-0474.

83
Copy DOIDOI Copied
Visit DOI Link

 Baud VKarin MIs NF-kappab a good target for cancer therapy? Hopes and pitfallsNat Rev Drug Discov2009;8:3340. DOI10.1038/nrd2781.

84
Copy DOIDOI Copied
Visit DOI Link

 Salgia RPharaon RMambetsariev Iet alThe improbable targeted therapy: KRAS as an emerging target in non-small cell lung cancer (NSCLC)Cell Rep Med2021;2:100186DOI10.1016/j.xcrm.2020.100186.

85
Copy DOIDOI Copied
Visit DOI Link

 Ghimessy ARadeczky PLaszlo Vet alCurrent therapy of KRAS-mutant lung cancerCancer Metastasis Rev2020;39:115977. DOI10.1007/s10555-020-09903-9.

86
Copy DOIDOI Copied
Visit DOI Link

 Li BTFalchook GSDurm GAet alOA03.06 CodeBreaK 100/101: First report of safety/efficacy of sotorasib in combination with pembrolizumab or atezolizumab in advanced KRAS p.g12c NSCLCJ Thorac Oncol2022;17(Suppl.):S101DOI10.1016/j.jtho.2022.07.025.

87
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govPhase 2 Trial of Adagrasib Monotherapy and in Combination With Pembrolizumab and a Phase 3 Trial of Adagrasib in Combination in Patients With a KRAS G12C Mutation KRYSTAL-7. ClinicalTrials.gov identifier: NCT04613596. Available athttps://clinicaltrials.gov/study/NCT04613596 (accessed22 November 2024).

88
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govPhase I Trial of Adagrasib (MRTX849) in Combination With Cetuximab and Irinotecan in Patients With Colorectal Cancer. ClinicalTrials.gov identifier: NCT05722327. Available athttps://clinicaltrials.gov/study/NCT05722327 (accessed22 November 2024).

89
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govPhase 3 Study of MRTX849 With Cetuximab vs Chemotherapy in Patients With Advanced Colorectal Cancer With KRAS G12C Mutation (KRYSTAL-10). ClinicalTrials.gov identifier: NCT04793958. Available athttps://clinicaltrials.gov/study/NCT04793958 (accessed22 November 2024).

90
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govAdagrasib in Combination With TNO155 in Patients With Cancer (KRYSTAL 2). ClinicalTrials.gov identifier: NCT04330664. Available athttps://clinicaltrials.gov/study/NCT04330664 (accessed22 November 2024).

91
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govAdagrasib in Combination With BI 1701963 in Patients With Cancer (KRYSTAL 14). ClinicalTrials.gov identifier: NCT04975256. Available athttps://clinicaltrials.gov/study/NCT04975256 (accessed22 November 2024).

92
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Phase 1/2 Study of MRTX0902 in Solid Tumors With Mutations in the KRAS MAPK Pathway. ClinicalTrials.gov identifier: NCT05578092. Available athttps://clinicaltrials.gov/study/NCT05578092 (accessed2 December 2022).

93
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Phase I Study of Adagrasib and Durvalumab for Treatment of Advanced Non-Small Cell Lung Cancers and Gastro-Intestinal Cancers Harboring KRAS G12C Mutations. ClinicalTrials.gov identifier: NCT05848843. Available athttps://clinicaltrials.gov/study/NCT05848843 (accessed25 April 2023).

94
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study of INCB099280 in Combination With Adagrasib in Adults With Advanced Solid Tumors Harboring A KRASG12C Mutation. ClinicalTrials.gov identifier: NCT06039384. Available athttps://clinicaltrials.gov/study/NCT06039384 (accessed22 November 2024).

95
Copy DOIDOI Copied
Visit DOI Link

 Yaeger RLanger CJRuffinelli JCet alA phase 1b study of sotorasib combined with panitumumab as second-line treatment of kras G12C  mutated colorectal cancerJ Clin Oncol2024;42:128128DOI10.1200/JCO.2024.42.3_suppl.128.

96
Copy DOIDOI Copied
Visit DOI Link

 Hong DSKuboki YStrickler JHet alSotorasib (soto) plus panitumumab (pmab) and FOLFIRI for previously treated kras G12C-mutated metastatic colorectal cancer (mcrc): codebreak 101 phase 1b safety and efficacyJ Clin Oncol2023;41:35133513. DOI10.1200/JCO.2023.41.16_suppl.3513.

97
Copy DOIDOI Copied
Visit DOI Link

 Pietrantonio FSalvatore LEsaki Tet alLBA10 sotorasib plus panitumumab versus standard-of-care for chemorefractory KRAS G12C-mutated metastatic colorectal cancer (mcrc): codebreak 300 phase III studyAnn Oncol2023;34:S1266DOI10.1016/j.annonc.2023.10.016.

98
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govSotorasib Activity in Subjects With Advanced Solid Tumors With KRAS p.G12C Mutation (CodeBreak 101). ClinicalTrials.gov identifier: NCT04185883. Available athttps://clinicaltrials.gov/study/NCT04185883 (accessed22 November 2024).

99
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govSHP2 Inhibitor BBP-398 in Combination With Sotorasib in Patients With Advanced Solid Tumors and a KRAS-G12C Mutation (Argonaut). ClinicalTrials.gov identifier: NCT05480865. Available atwww.clinicaltrials.gov/study/NCT05480865 (accessed22 November 2024).

100
Copy DOIDOI Copied
Visit DOI Link

 Kondo SYan DGanju Vet alD-1553 (Garsorasib) in Patients with KRAS G12C mutated Advanced Pancreatic Cancer (PCa)Available atwww.cabrini.com.au/app/uploads/29.-Gary-Richardson-D-1553-in-patients-with-KRAS-G12C-mutated-advanced-pancreatic-cancer.pdf (accessed22 November 2024).

101
Copy DOIDOI Copied
Visit DOI Link

 Li JShen LGu Yet alPreliminary activity and safety results of KRAS G12C inhibitor glecirasib (JAB-21822) in patients with pancreatic cancer and other solid tumorsJ Clin Oncol2024;42:604604DOI10.1200/JCO.2024.42.3_suppl.604.

102
Copy DOIDOI Copied
Visit DOI Link

 JacobioJacobio Announces China CDE Clearance for Phase III Clinical Trial of KRAS G12C Inhibitor Glecirasib in Patients with Colorectal Cancer2024Available atwww.jacobiopharma.com/en/news/Jacobio_Announces_China_CDE_Clearance_for_Phase_III_Clinical_Trial_of_KRAS_G12C_Inhibitor_Glecirasib_in_Patients_with_Colorectal_Cancer (accessed22 November 2024).

103
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govA Study of JAB-21822 in Patients With KRAS p.G12C Mutated Pancreatic Cancer. ClinicalTrials.gov: NCT06008288. Available athttps://clinicaltrials.gov/study/NCT06008288 (accessed22 November 2024).

104
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govStudy of JDQ443 in Patients With Advanced Solid Tumors Harboring the KRAS G12C Mutation (KontRASt-01). ClinicalTrials.gov identifier: NCT04699188. Available athttps://clinicaltrials.gov/study/NCT04699188 (accessed22 November 2024).

105
Copy DOIDOI Copied
Visit DOI Link

 ClinicalTrials.govPlatform Study of JDQ443 in Combinations in Patients With Advanced Solid Tumors Harboring the KRAS G12C Mutation (KontRASt-03). ClinicalTrials.gov identifier: NCT05358249. Available athttps://clinicaltrials.gov/study/NCT05358249 (accessed22 November 2024).

106
Copy DOIDOI Copied
Visit DOI Link

 Maciag AEStice JWang Bet alAbstract ND07: BBO-8520, a first-in-class, direct inhibitor of KRASG12C (ON), locks GTP-bound KRASG12C in the state 1 conformation resulting in rapid and complete blockade of effector bindingCancer Res2024;84:D07ND07DOI10.1158/1538-7445.AM2024-ND07.

107
Copy DOIDOI Copied
Visit DOI Link

 Jänne PABigot FPapadopoulos Ket alAbstract PR014: preliminary safety and anti-tumor activity of RMC-6291, a first-in-class, tri-complex KRASG12C(ON) inhibitor, in patients with or without prior KRASG12C(OFF) inhibitor treatmentMol Cancer Ther2023;22:R014PR014. DOI10.1158/1535-7163.TARG-23-PR014.

4

Article Information

Disclosure

Lorenza Rimassa consults, advises, is on the speakers’ bureau and received grants from AstraZeneca, Eisai, Incyte, Ipsen and Roche. She consults, advises and is on the speakers’ bureau for Bayer, Bristol Myers Squibb and Servier. She consults, advises and received grants from Eli Lilly, Exelixis, MSD, Nerviano Medical Sciences and Zymeworks. She consults and advises for Basilea, Genenta, Hengrui, IQVIA, Jazz Pharmaceuticals and Taiho Oncology. She is a member of the speakers’ bureau for Gilead, Merck Serono and Sanofi. She received grants from Agios, BeiGene and Fibrogen. Mohamad B Sonbol has received consulting income from Novartis. Tanios Bekaii-Saab has received consulting income from Boehringer Ingelheim, Treos Bio, Sobi, Ipsen, Array Biopharma, Seattle Genetics, Bayer, Genentech, Incyte, Merck, Boston Biomedical, Bayer, Amgen, Merck, Celgene, Lilly, Ipsen, Clovis, Seattle Genetics, Array Biopharma, Genentech, Abgenomics and Incyte. Celine Hoyek, Angelo Pirozzi and Jeremy Jones have no financial or non-financial relationships or activities to declare in relation to this article.

Compliance With Ethics

This article involves a review of literature and does not report on new clinical data, or any studies with human or animal subjects performed by any of the authors.

Review Process

Double-blind peer review.

Authorship

All named authors meet the criteria of the International Committee of Medical Journal Editors for authorship for this manuscript, take responsibility for the integrity of the work as a whole and have given final approval for the version to be published.

Correspondence

Celine HoyekDepartment of Hematology and Oncology, Mayo Clinic5777 East Mayo BlvdPhoenixAZ 85054USAHoyek.Celine@mayo.edu

Support

No funding was received for the publication of this article.

Access

This article is freely accessible at touchONCOLOGY.com. ©Touch Medical Media 2024.

Data Availability

Data sharing is not applicable to this article as no datasets were generated or analysed during the writing of this article.

Received

2024-07-09

5

Further Resources

Share
Facebook
X (formerly Twitter)
LinkedIn
Via Email
Mark CompleteCompleted
BookmarkBookmarked
Copy LinkLink Copied
Download as PDF
Close Popup