KRAS

Source: Wikipedia, the free encyclopedia.
KRAS
Gene ontology
Molecular function
Cellular component
Biological process
Sources:Amigo / QuickGO
Ensembl
UniProt
RefSeq (mRNA)

NM_004985
NM_033360
NM_001369786
NM_001369787

NM_021284

RefSeq (protein)

NP_004976
NP_203524
NP_001356715
NP_001356716
NP_004976.2

Location (UCSC)Chr 12: 25.21 – 25.25 MbChr 6: 145.16 – 145.2 Mb
PubMed search[3][4]
Wikidata
View/Edit HumanView/Edit Mouse

KRAS (

proto-oncogene
.

The K-Ras protein is a GTPase, a class of enzymes which convert the nucleotide guanosine triphosphate (GTP) into guanosine diphosphate (GDP). In this way the K-Ras protein acts like a switch that is turned on and off by the GTP and GDP molecules. To transmit signals, it must be turned on by attaching (binding) to a molecule of GTP. The K-Ras protein is turned off (inactivated) when it converts the GTP to GDP. When the protein is bound to GDP, it does not relay signals to the nucleus.

The

cell membranes because of the presence of an isoprene group on its C-terminus. There are two protein products of the KRAS gene in mammalian cells that result from the use of alternative exon 4 (exon 4A and 4B respectively): K-Ras4A and K-Ras4B. These proteins have different structures in their C-terminal region and use different mechanisms to localize to cellular membranes, including the plasma membrane.[8]

Function

KRAS acts as a molecular on/off switch, using

Guanine Nucleotide Exchange Factor (GEF) class (such as SOS1), which forces the release of bound nucleotide (GDP). Subsequently, KRAS binds GTP present in the cytosol
and the GEF is released from ras-GTP.

Other members of the Ras family include: HRAS and NRAS. These proteins all are regulated in the same manner and appear to differ in their sites of action within the cell.[citation needed]

Clinical significance when mutated

This

lung adenocarcinoma,[10] mucinous adenoma, ductal carcinoma of the pancreas and colorectal cancer.[11][12]

Several

Somatic KRAS mutations are found at high rates in leukemias, colorectal cancer,[15] pancreatic cancer[16] and lung cancer.[17]

Colorectal cancer

The impact of KRAS mutations is heavily dependent on the order of mutations. Primary KRAS mutations generally lead to a self-limiting hyperplastic or borderline lesion, but if they occur after a previous

APC mutation it often progresses to cancer.[18] KRAS mutations are more commonly observed in cecal cancers than colorectal cancers located in any other places from ascending colon to rectum.[19][20]

As of 2006, KRAS mutation was predictive of a very poor response to panitumumab (Vectibix) and cetuximab (Erbitux) therapy in colorectal cancer.[21]

As of 2008, the most reliable way to predict whether a colorectal cancer patient will respond to one of the

EGFR-inhibiting drugs was to test for certain “activating” mutations in the gene that encodes KRAS, which occurs in 30%–50% of colorectal cancers. Studies show patients whose tumors express the mutated version of the KRAS gene will not respond to cetuximab or panitumumab.[22]

As of 2009 although presence of the wild-type (or normal) KRAS gene does not guarantee that these drugs will work, a number of large studies[23][24] had shown that cetuximab had efficacy in mCRC patients with KRAS wild-type tumors. In the Phase III CRYSTAL study, published in 2009, patients with the wild-type KRAS gene treated with Erbitux plus chemotherapy showed a response rate of up to 59% compared to those treated with chemotherapy alone. Patients with the KRAS wild-type gene also showed a 32% decreased risk of disease progression compared to patients receiving chemotherapy alone.[24]

As of 2012 it was known that emergence of KRAS mutations was a frequent driver of acquired resistance to cetuximab anti-EGFR therapy in colorectal cancers. The emergence of KRAS mutant clones can be detected non-invasively[how?] months before radiographic progression. It suggests to perform an early initiation of a MEK inhibitor as a rational strategy for delaying or reversing drug resistance.[25]

KRAS amplification

KRAS gene can also be

EGFR inhibitors. Although KRAS amplification is infrequent in colorectal cancer, as of 2013 it was hypothesized to be responsible for precluding response to anti-EGFR treatment in some patients.[26] As of 2015 amplification of wild-type Kras has also been observed in ovarian,[27] gastric, uterine, and lung cancers.[28]

Lung cancer

Whether a patient is positive or negative for a mutation in the epidermal growth factor receptor (EGFR) will predict how patients will respond to certain EGFR antagonists such as erlotinib (Tarceva) or gefitinib (Iressa). Patients who harbor an EGFR mutation have a 60% response rate to erlotinib. However, the mutation of KRAS and EGFR are generally mutually exclusive.[29][30][31] Lung cancer patients who are positive for KRAS mutation (and the EGFR status would be wild type) have a low response rate to erlotinib or gefitinib estimated at 5% or less.[29]

Different types of data including mutation status and gene expression did not have a significant prognostic power.[32] No correlation to survival was observed in 72% of all studies with KRAS sequencing performed in non-small cell lung cancer (NSCLC).[32] However, KRAS mutations can not only affect the gene itself and the expression of the corresponding protein, but can also influence the expression of other downstream genes involved in crucial pathways regulating cell growth, differentiation and apoptosis. The different expression of these genes in KRAS-mutant tumors might have a more prominent role in affecting patient's clinical outcomes.[32]

A 2008 paper published in Cancer Research concluded that the in vivo administration of the compound oncrasin-1 "suppressed the growth of K-ras mutant human lung tumor xenografts by >70% and prolonged the survival of nude mice bearing these tumors, without causing detectable toxicity", and that the "results indicate that oncrasin-1 or its active analogues could be a novel class of anticancer agents which effectively kill K-Ras mutant cancer cells."[33]

Pancreatic cancer

Over 90% of

pancreatic ductal adenocarcinomas (PDACs) have a KRAS mutation.[34][35][36] There is one approved drug, sotorasib, that targets the KRAS G12C mutation, but only ~1% of PDACs have this mutation.[34] Another KRAS inhibitor, MRTX1133 targets G12D mutation which is present in over 40% of PDACs[37][38] is currently in clinical trials to treat solid tumors including pancreatic adenocarcinoma.[39]

KRAS testing

In July 2009, the US Food and Drug Administration (FDA) updated the labels of two anti-EGFR monoclonal antibody drugs indicated for treatment of metastatic colorectal cancer, panitumumab (Vectibix) and cetuximab (Erbitux), to include information about KRAS mutations.[40]

In 2012, the FDA cleared a genetic test by

QIAGEN named therascreen KRAS test, designed to detect the presence of seven mutations in the KRAS gene in colorectal cancer cells. This test aids physicians in identifying patients with metastatic colorectal cancer for treatment with Erbitux. The presence of KRAS mutations in colorectal cancer tissue indicates that the patient may not benefit from treatment with Erbitux. If the test result indicates that the KRAS mutations are absent in the colorectal cancer cells, then the patient may be considered for treatment with Erbitux.[41]

As a therapeutic target

As of 2014, driver mutations in KRAS were known to underlie the pathogenesis of up to 20% of human cancers.[42] Hence KRAS is an attractive drug target, but as of 2018 lack of obvious binding sites had hindered pharmaceutical development.[43] One potential drug interaction site is where GTP/GDP binds, but due to the extraordinarily high affinity of GTP/GDP for this site, it appeared unlikely as of 2018 that drug-like small molecule inhibitors could compete with GTP/GDP binding. Other than where GTP/GDP binds, there are no obvious high affinity binding sites for small molecules.[44]

G12C mutation

Surface model of a KRASG12C protein, showing a GDP molecule (orange) in its high-affinity binding site and the covalent inhibitor sotorasib (aqua) occupying an adjacent "cryptic" binding pocket. Sotorasib forms an irreversible bond with a cysteine residue and disrupts function of the mutated protein. From PDB: 6OIM​.[45][46]

One fairly frequent driver mutation is KRASG12C which is adjacent a shallow binding site. As of 2019, this allowed the development of

nucleophilic sulfur atom of Cys-12 and hence selectively target KRASG12C and leave wild-type KRAS untouched.[47]

In 2021, the U.S. FDA approved one KRASG12C mutant

non-small cell lung cancer (NSCLC), the first KRAS inhibitor to reach the market and enter clinical use.[48][49]

A second is

investigational new drug (IND) approval to start clinical trials.[52] An antisense oligonucleotide (ASO) targeting KRAS, AZD4785 (AstraZeneca/Ionis Therapeutics), completed a phase I study[53] but in 2019 was discontinued from further development because of insufficient knockdown of the target.[54]

A phase Ia/Ib dose escalation trial of the oral selective KRAS G12C inhibitor divarasib was published in 2023, where the drug was tested in non-small cell lung cancer, colorectal cancer, and other solid tumors with KRAS G12C mutations.[55] It continues in phase I and II studies for several cancer types as of August 2023.[56][57][58][59]

G12D mutation

The most common KRAS mutation is G12D which is estimated to be present in up to 37% pancreatic cancers and over 12% of colorectal cancers. Normally amino acid position 12 of the KRAS protein is occupied by glycine but in G12D it is occupied by aspartic acid.[60]

As of 2023, there are no commercial drug candidates targeting the KRAS G12D mutation in the clinical phase of development.

A novel inhibitor finding strategy for mutated G12D KRAS molecules was described in.[61] The KRAS mutations in the 12th residue position inhibit the bound of the regulatory GAP molecule to the mutated KRAS, causing uncontrolled cell growth. The novel strategy proposes finding small glue molecules, which attach the mutated KRAS to the GAP, prohibiting uncontrolled cell growth and restoring the normal function. For this goal a theoretical KRAS-GAP conformation was designed with a several Å gap between the molecules, and a high-throughput in silico docking was performed for finding gluing agents. As a proof of concept, two novel molecules were described with satisfying biological activity.

As of 2021, there were a number of drug candidates in

investigational new drug (IND) approval in H1:2021 to start clinical trials.[62] As of 2022 Revolution Medicines was exploring a small molecule therapy and reported anti-tumor activity in KRAS-G12D mutant tumor models.[63]

In 2021, the first clinical trial of a gene therapy targeting KRAS G12D was recruiting patients, sponsored by the National Cancer Institute.[64]

In June 2022, a case report was published about a 71-year-old woman with metastatic pancreatic cancer after extensive treatment (

Whipple Surgery, radiation and multiple agent chemotherapy) who received a single infusion of her blood with engineered T cells with 2 genes encoding T cell receptors, directed to both the G12D mutation and an HLA allele (HLA-C*08:02). Her tumor regressed persistently. But another similarly treated patient died from the cancer.[65]

Interactions

KRAS has been shown to

interact
with:

References

  1. ^ a b c GRCh38: Ensembl release 89: ENSG00000133703Ensembl, May 2017
  2. ^ a b c GRCm38: Ensembl release 89: ENSMUSG00000030265Ensembl, May 2017
  3. ^ "Human PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  4. ^ "Mouse PubMed Reference:". National Center for Biotechnology Information, U.S. National Library of Medicine.
  5. PMID 6287573
    .
  6. .
  7. .
  8. .
  9. .
  10. .
  11. .
  12. .
  13. .
  14. .
  15. .
  16. .
  17. .
  18. .
  19. .
  20. .
  21. .
  22. ^ van Epps HL (Winter 2008). "Bittersweet Gene". CURE (Cancer Updates, Research and Education). Archived from the original on 2009-02-07.
  23. PMID 19114683
    .
  24. ^ .
  25. .
  26. .
  27. .
  28. .
  29. ^ .
  30. .
  31. .
  32. ^ .
  33. .
  34. ^ .
  35. .
  36. ^ Rosenzwieg A (8 June 2021). "5 Things to Know About Targeting Mutant KRAS in Pancreatic Cancer". Pancreatic Cancer Action Network.
  37. S2CID 263967602
    .
  38. .
  39. ^ Clinical trial number NCT05737706 for "Study of MRTX1133 in Patients With Advanced Solid Tumors Harboring a KRAS G12D Mutation" at ClinicalTrials.gov
  40. ^ OncoGenetics.Org (July 2009). "FDA updates Vectibix and Erbitux labels with KRAS testing info". OncoGenetics.Org. Archived from the original on November 9, 2014. Retrieved 2009-07-20.
  41. ^ FDA: Medical devices: therascreen® KRAS RGQ PCR Kit – P110030, accessed 20 Jone 2014
  42. PMID 25323927
    .
  43. .
  44. .
  45. .
  46. .
  47. .
  48. ^ "FDA Approves First Targeted Therapy for Lung Cancer Mutation Previously Considered Resistant to Drug Therapy". U.S. Food and Drug Administration (FDA). 28 May 2021. Retrieved 28 May 2021.
  49. ^ NCI Staff (2021-06-25). "Sotorasib is First KRAS Inhibitor Approved by FDA - NCI". Cancer Currents. National Cancer Institute. Retrieved 2022-06-04.
  50. ^ Clinical trial number NCT03785249 for "MRTX849 in Patients With Cancer Having a KRAS G12C Mutation" at ClinicalTrials.gov
  51. ^ Kaiser J (2019-10-30). "Two new drugs finally hit 'undruggable' cancer target, providing hope for treatments". Science Magazine. AAAS. Retrieved 2019-11-04.
  52. PMID 31267080
    .
  53. ^ Clinical trial number NCT03101839 for "Phase I Dose-Escalation Study of AZD4785 in Patients With Advanced Solid Tumours" at ClinicalTrials.gov
  54. ^ Plieth J (26 April 2019). "Astra's first attempt fails, but there's no giving up on KRAS". Evaluate.
  55. S2CID 261098837
    .
  56. ^ "Study Record | A Study of Multiple Therapies in Biomarker-Selected Patients With Resectable Stages IB-III Non-Small Cell Lung Cancer". clinicaltrials.gov. Retrieved 2023-08-26.
  57. ^ "Study Record | A Study Evaluating the Safety and Efficacy of Targeted Therapies in Subpopulations of Patients With Metastatic Colorectal Cancer (INTRINSIC)". clinicaltrials.gov. Retrieved 2023-08-26.
  58. ^ "Study Record | A 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. Retrieved 2023-08-26.
  59. ^ "Study Record | A Study to Evaluate the Efficacy and Safety of Multiple Targeted Therapies as Treatments for Participants With Non-Small Cell Lung Cancer (NSCLC) (B-FAST)". clinicaltrials.gov. Retrieved 2023-08-26.
  60. PMID 35046095
    .
  61. .
  62. ^ "Mirati Therapeutics Reports Investigational Adagrasib (MRTX849) Preliminary Data Demonstrating Tolerability and Durable Anti-Tumor Activity as well as Initial MRTX1133 Preclinical Data". ir.mirati.com. Retrieved 26 July 2021.
  63. ^ "RMC-9805 (RM-036), a First-in-Class, Orally-Bioavailable, Tri-Complex Covalent KRAS-G12D(ON) Inhibitor, Drives Profound Anti-Tumor Activity in KRAS-G12D Mutant Tumor Models". Revolution Medicines. Retrieved 2023-01-29.
  64. ^ "A Phase I/II Study Administering Peripheral Blood Lymphocytes Transduced With a Murine T-Cell Receptor Recognizing the G12D Variant of Mutated RAS in HLA-A*11:01 Patients". clinicaltrials.gov. 28 January 2021. Retrieved 26 July 2021.
  65. PMID 35648703
    .
  66. ^ .
  67. .
  68. .
  69. .
  70. .
  71. .

Further reading

External links

This page is based on the copyrighted Wikipedia article: KRAS. Articles is available under the CC BY-SA 3.0 license; additional terms may apply.Privacy Policy