RAS Mutations in Human Cancers: Roles in Precision Medicine: Seminars in Cancer Biology

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Review

RAS mutations in human cancers: Roles in precision medicine


Avaniyapuram Kannan Murugana, , Michele Griecob, Nobuo Tsuchidaa,
⁎ ⁎⁎

a
Department of Molecular Cellular Oncology and Microbiology, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549 Japan
b
DiSTABiF, Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Seconda Università di Napoli, via Vivaldi 43, Caserta 81100 Italy

ARTICLE INFO ABSTRACT

Keywords: Ras proteins play a crucial role as a central component of the cellular networks controlling a variety of signaling
Mutation pathways that regulate growth, proliferation, survival, differentiation, adhesion, cytoskeletal rearrangements
Precision medicine and motility of a cell. Almost, 4 decades passed since Ras research was started and ras genes were originally
Oncogene discovered as retroviral oncogenes. Later on, mutations of the human RAS genes were linked to tumorigenesis.
HRAS
Genetic analyses found that RAS is one of the most deregulated oncogenes in human cancers. In this review, we
KRAS
summarize the pioneering works which allowed the discovery of RAS oncogenes, the finding of frequent mu-
NRAS
Cancer tations of RAS in various human cancers, the role of these mutations in tumorigenesis and mutation-activated
Personalized medicine signaling networks. We further describe the importance of RAS mutations in personalized or precision medicine
particularly in molecular targeted therapy, as well as their use as diagnostic and prognostic markers as ther-
apeutic determinants in human cancers.

1. Introduction that Rap (Ras-associated protein-1), Rit (Ras-like without CAAX 1), Ral
(Ras-like proto-oncogene), Rras2 (Ras-related 2) and Ras are belonging
Ras proteins are small GTPases which comprise three RAS proto- to the same branch in the Ras family tree. Each member of the
oncogenes such as HRAS, KRAS, and NRAS in Homo sapiens (humans), branching Ras family has more than one isoform. The main branch of
and they are orthologues of Hras (Harvey rat sarcoma virus oncogene), the Ras family tree consists of several members such as Rap1A, Rap1B,
Kras (Kirsten rat sarcoma virus oncogene) and Nras (Neuroblastoma ras Rap2A, Rap2B, and Rap2C (Ras-associated protein-1A, B and 2A-2C,
oncogene) in mice. The Ras genes are involved in the modulation of member of Ras oncogene family), Rit1 (Ras-like without CAAX 1), Rit2
various key cellular signaling pathways and regulate vital cellular (Ras-like without CAAX 2), RalA isoform 1 (Ras-like proto-oncogene A
functions such as differentiation, cell proliferation, growth, cell cycle, isoform 1), RalA isoform 2 (Ras-like proto-oncogene A isoform 2), RalB
and motility. In this article, Ras gene nomenclature is used as re- (Ras-like proto-oncogene B), Eras (ES cell-expressed Ras), Hras isoform
commended by HGNC (HUGO Gene Nomenclature Committee) and as 1 (HRas proto-oncogene isoform 1), Hras isoform 2 (HRas proto-onco-
coined previously [1]. We use accordingly here, as, ras for viral origin, gene isoform 2), Kras2A (KRas proto-oncogene isoform 2A), Kras2B
RAS for the human origin and Ras for both viral and human origins. The (KRas proto-oncogene isoform 2B), Nras (NRas proto-oncogene), Mras
RAS is implicated in many human diseases including cancer and it has (Muscle Ras oncogene homolog), Rras (Ras-related) and Rras2 (Ras-
been set as a "hallmark cancer gene" as it is the most mutated oncogene related 2) [6,7]. Though there are more than 39 various proteins, many
in human cancers [1–4]. The term "ras" was originally derived from the of them were not very well characterized except for HRAS, KRAS, and
rat sarcoma virus and it denotes a common set of genes of rat-derived NRAS. In normal cells, most of the characterized ras family proteins
murine sarcoma retroviruses responsible for transformation and tu- have been found to be involved in the signaling pathways which reg-
morigenesis [5]. ulate important cellular functions. When deregulated, these proteins
Ras encoded proteins belongs to the family of small GTPases. The induce abnormal cellular proliferation, growth, survival, cytoskeletal
Ras family of small GTPases genes consists of approximately 36 mem- rearrangements, adhesion, motility, and differentiation resulting in
bers, which encode about 39 Ras proteins. The molecular weight of malignant tumor formation [8]. Here, we summarize the origin and
these encoded Ras proteins ranges from 20 to 29 kDa. It has been shown evolution of Ras research in its golden epochs, deregulated RAS genes in

Corresponding author. Present address: Department of Molecular Oncology, King Faisal Specialist Hospital & Research Centre, PO Box 3354, Research Center

(MBC 03), Riyadh, 11211, Saudi Arabia.


⁎⁎
Corresponding author.
E-mail addresses: akmurugan@gmail.com (A.K. Murugan), ntutid.eme@tmd.ac.jp (N. Tsuchida).

https://doi.org/10.1016/j.semcancer.2019.06.007
Received 28 November 2018; Received in revised form 13 May 2019; Accepted 7 June 2019
1044-579X/ © 2019 Elsevier Ltd. All rights reserved.

Please cite this article as: Avaniyapuram Kannan Murugan, Michele Grieco and Nobuo Tsuchida, Seminars in Cancer Biology,
https://doi.org/10.1016/j.semcancer.2019.06.007
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 1. Timeline: milestones in the Ras research.


Timeline shows the important key discoveries of Ras oncogenes in the field of cancer research. The related reference numbers [10,12,13,16,17,19,25,28,42–44,
49,50–54,56,58,69,71,74,78,83,89,96,102,143–146] are mentioned within the bracket at the end of the sentences.

human cancer, mutated RAS-induced signaling, RAS mutations and transfer to newborn Wister-Furth rats [13]. Secondly, Peters et al iso-
their importance in precision medicine particularly in molecular tar- lated BALB-MSV from a leukemic mouse in 1974 [14]. Thirdly, in 1978,
geted therapy, as diagnostic and prognostic markers in human cancers Rasheed et al, isolated stable rat sarcoma virus from cultured rat cells
is discussed. [15]. Further, molecular cloning and characterization of Ha-MSV were
accomplished by Gordon L. Hager in 1979 [16]. Most of the sequences
2. Discovery of ras oncogenes: at the bench responsible for the oncogenic properties of these acute transforming
viruses were molecularly defined in the early 1980s.
The pioneering work that led to the discovery of the ras oncogenes Molecular DNA clones of Ha-MSV and Ki-MSV genomes were iso-
was started almost half a century ago and boomed in the early 1980s lated and characterized by Ed Scolinick and his colleagues and Nobuo
and still, ras-related research findings are hot pop-ups in the field of Tsuchida's laboratory, respectively [16–18]. In 1981, Ed Scolnick and
molecular and cellular biology. Moreover, cancer biology is expecting his associates including Nobuo Tsuchida established that the Harvey
every day yet other findings on the role of RAS genes in human cancer. (Ha-MSV) and Kirsten (Ki-MSV) strains were recombinant viruses that
Retrovirus researches eventually lead to the finding of ras oncogenes. carry sequences derived from the rat 30S RNA genome [19]. KiMSV
As illustrated in Fig. 1, the discovery of the Rous sarcoma virus (RSV) clone 4 had been presented in a Cold Spring Harbor Laboratory (CSHL)
can be traced back to approximately 100 years ago. In 1911, Peyton meeting of RNA tumor viruses (abstract, page 35 in 1980) much before
Rous discovered the RSV virus, when he was working at The Rockefeller the publication of Ki-MSV cloning paper of clone 4, in 1981 [17]. Much
University in New York. He injected cell-free extract of a chicken tumor earlier to this publication, sub-clones of Ki-MSV clone 4 and Hi-MSV
into healthy Plymouth Rock chickens, the extract induced a tumor of clone H-1 were constructed and compared thus allowing to estimate the
the connective tissue (a sarcoma) [9,10]. Therefore, it was termed as area of the p21 transforming genes, then called src. Harvey and Kirstein
Rous sarcoma virus (RSV). The RSV is a retrovirus, causes sarcoma in murine sarcoma viruses originated from divergent members of a family
chickens and is the first oncovirus to have been described. Rous was of normal vertebrate genes (Hras in Ha-MSV and Kras in Ki-MSV) [19].
awarded the Nobel Prize for his discovery in 1966, almost 55 years after Among sub-clones, KBE2 and HiHi3 for Kras, and HB-11, and BS-9 for
his discovery of RSV when Rous was 87 years old [11]. Hras were used in most laboratories except for Aaronson’s and Barba-
In 1964, Jennifer Harvey observed sarcomas in new-born rodents cid’s labs, where human RAS genes were cloned by themselves and
which were inoculated a preparation of a murine leukemia virus, taken identified using v-bas (mouse Hras) probes.
from a leukemic rat [12]. Harvey's findings initiated ras discoveries, The first report of human KRAS sequence was published in August
subsequently; three additional studies found that three retroviruses also 1983 [20,21], one year after Nobuo Tsuchida's publication of the viral
carry ras oncogenes. Firstly, in 1967, Kirsten demonstrated that he Kras oncogene sequence which had transforming activity, published in
could obtain two serial passages of MSV (moloney sarcoma virus) cell- September 1982 [18]. Thus, the sequence of Kras in Tsuchida's paper is
free extract (later it was called Kirsten-MSV) which he was able to the first Kras sequence to be reported. Robert Weinberg's laboratory

2
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Table 1
Somatic mutation of the RAS oncogenes in human cancers.
S. No HRAS mutations (%)* KRAS mutations (%)* NRAS mutations (%)*
Primary tumor tissues Primary tumor tissues Primary tumor tissues

1 Salivary gland 24/164 (15%) Pancreas 3247/5656 (57%) Skin 918/5368 (17%)
2 Cervix 23/264 (9%) Large intestine 13177/38080 (35%) Hemato. & lymphoid 929/8882 (10%)
3 Upper aero-digestive tract 110/1202 (9%) Biliary tract 491/1766 (28%) Thyroid 330/4755 (7%)
4 Urinary tract 165/1765 (9%) Lung 2872/17472 (16.43%) Autonomic ganglia 6/102 (6%)
5 Penis 2/28 (7%) Small intestine 63/377 (17%) Adrenal gland 9/171 (5%)
6 Skin 134/2168 (6%) Endometrium 332/2268 (15%) Soft tissue 27/509 (5%)
7 Prostate 29/558 (5%) Ovary 436/3192 (14%) Large intestine 62/1731 (4%)
8 Soft tissue 37/740 (5%) Gastrointestinal tract 7/73 (10%) Upper aero-digestive 24/860 (3%)
9 Stomach 14/384 (4%) Prostate 94/1214 (8%) Liver 8/310 (3%)
10 Testis 5/130 (4%) Cervix 46/637 (7%) Ovary 5/191 (3%)
11 Thyroid 153/4231 (4%) Peritoneum 6/87 (7%) Testis 8/283 (3%)
12 Pituitary 10/300 (3%) Soft tissue 73/1100 (7%) Cervix 2/132 (2%)
13 Bone 3/199 (2%) Stomach 195/2814 (7%) Biliary tract 6/287 (2%)
14 Thymus 1/46 (2%) Liver 31/507 (6%) Pancreas 5/306 (2%)
15 Adrenal gland 1/136 (1%) Hemato. & lymphoid 310/6116 (5%) Stomach 5/215 (2%)
16 Breast 5/720 (1%) Genital tract 1/25 (4%) Urinary tract 10/873 (1%)
17 Endometrium 3/291 (1%) Testis 17/432 (4%) Breast 7/508 (1%)
18 Esophagus 2/161(1%) Penis 1/28 (4%) Prostate 8/588 (1%)
19 Autonomic ganglia 0/63 (0%) Urinary tract 47/1099 (4%) Central nervous system 8/1069 (1%)
20 Biliary tract 0/153 (0%) Eye 4/90 (4%) Endometrium 2/314 (1%)
21 Central nervous system 0/964 (0%) Autonomic ganglia 2/63 (3%) Lung 28/3084 (1%)
22 Eye 0/33 (0%) Breast 24/789 (3%) Eye 1/106 (1%)
23 Gastrointestinal tract 0/1 (0%) Esophagus 13/375 (3%) Fallopian tube 0/2 (0%)
24 Hematopoietic& lymphoid 8/3076 (0.2%) Salivary gland 6/173 (3%) Bone 0/207(0%)
25 Kidney 1/273 (0.3%) Upper aero-digestive 54/1673 (3%) Gastrointestinal tract 0/2 (0%)
26 Large intestine 2/717 (0%) Skin 39/1532 (3%) Kidney 2/435 (0%)
27 Liver 1/271 (0%) Thyroid 116/5259 (2%) Meninges 0/77 (0%)
28 Lung 9/2094 (0%) Thymus 4/186 (2%) Esophagus 0/161 (0%)
29 Meninges 0/75 (0%) Bone 2/252 (1%) Parathyroid 0/100 (0%)
30 Ovary 0/152 (0%) Kidney 4/704 (1%) Penis 0/28 (0%)
31 Parathyroid 0/100 (0%) Central nervous system 9/1175 (1%) Pituitary 0/300 (0%)
32 Peritoneum 0/3 (0%) Fallopian tube 0/3 (0%) Placenta 0/2 (0%)
33 Placenta 0/2 (0%) Adrenal gland 1/211 (0%) Pleura 0/30 (0%)
34 Pleura 0/19 (0%) Meninges 0/62 (0%) Salivary gland 0/48 (0%)
35 Pancreas 0/279 (0%) Parathyroid 0/100 (0%) Small intestine 0/5 (0%)
36 Small intestine 0/5 (0%) Pituitary 0/300 (0%) Thymus 0/46 (0%)
37 Vulva 0/16 (0%) Placenta 0/10 (0%) Vagina 0/1 (0%)
37 – – Pleura 0/45 (0%) Vulva 0/16 (0%)
39 – – Vagina 0/2 (0%) – –
40 – – Vulva 0/16 (0%) – –
742/21,783 (3.41%) 21,724/95,963 (23%) 2410/32,104 (7.51%)

* Mutated tumor Samples/Total number of tumor sample analyzed (%); Hemato., hematopoietic tumors. The data were derived from COSMIC (Catalogue Of
Somatic Mutation in Cancer) database.

was the first to report the transformation of NIH-3T3 cells with genomic DNAs [32]. Wigler's lab and Barbacid's lab in collaboration with Stuart
DNA of chemically transformed mouse cells [22] and subsequently, Aaronson's lab, independently reported similar findings when they used
they also showed the presence of an Alu repetitive sequence in the NIH- probes specific for v-Hras/v-Bas and v-Kras to hybridize the DNA iso-
3T3 cells which had been transformed with DNA isolated from human lated from cells which were transformed by EJ bladder (EJ) and lung
tumor cell lines [23]. Alu repetitive sequences are human-specific and (LX-1) carcinoma cell line DNA, respectively [33,34]. Interestingly four
they allowed the identification of human gene fragments inserted by years later, Weinberg’s group found that the initial NIH-3T3 transfor-
transfection in the mouse genome. In late 1981, Wigler and his group mants reported by them were carrying Kras oncogene [35].
reported the presence of Alu-containing DNA fragments in foci which The functional differences caused by single nucleotide were found
were obtained transfecting DNA from human tumor cell lines in mouse after cloning the normal and oncogenic allele of human HRAS in 1983
cells [24]. In 1982, Weinberg and his group collaborating with Ed [25,27,36]. Unlike other oncogenes which were established as onco-
Scolnick described a point mutation of human HRAS in bladder carci- genes in the "post-ras-genomic era", the ras genes did not take a cake-
noma cells [25]. The Levinson's group used the KBE2 HiHi3 probe for walk to the "cancer-podium", contrary, they took many criticisms when
cloning human KRAS [26], while the Wigler's group used an Alu-spe- they were considered as oncogenes due to a single point mutation in the
cific probe in transfected-NIH-3T3 cells [27]. However, the uses of viral oncogenic allele. The question was raised because of two reasons:
Kras sub-clones were essential from the point that some of EcoRI firstly, as many mutations as arise in somatic cells every day, however
fragments of human KRAS DNA do not contain Alu sequence. Le- no tumor is formed until old age [37,38]; secondly, ras genes, unlike
vingson's group used HiHi3 to clone human KRAS sequence [28]. In their retroviral-counterparts oncogenes, were not able to transform
Early 1982, human transforming genes were cloned from T24 and EJ primary cells unless immortalized cells such as NIH-3T3 cells [39–41].
bladder carcinoma cell lines and reported independently by Weinberg, Moreover, the c-terminus of the RAS molecule was shown to be re-
Wigler and Barbacid laboratories [29–31]. quired for anchoring to the cell membrane and transformation [42].
The molecular nature of these human oncogenes was resolved when Eventually, a study effectively showed that the cooperation of a second
Channing Der used retroviral probes to hybridize DNA isolated from immortalizing oncogene such as myc or adenovirus E1A was required to
NIH-3T3 cells which had been transformed with various human tumor induce tumor growth in nude mice [39,41]. Besides, a new human

3
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

transforming gene was identified in 1983. This gene was termed as mutations has been identified in malignant melanomas (17%). A rela-
NRAS, which later became the third members of the Ras gene family, tively high incidence of the mutation has also been found in malig-
and it was also shown to be mutated in human cancers [21,43]. How- nancies of hematopoietic and lymphoid tissue and thyroid cancer re-
ever, the production of an NRAS deficient mouse showed that it is not presenting somatic mutation of NRAS 10% and 7%, respectively.
important for the growth and development but essential for immune Though the mutations of NRAS were also often detected in other tu-
response and T-cell function [44]. mors, their prevalence is relatively low, accounting for 0–5% (Table 1).
The first RAS mutation in human cancer tissue was identified in
1984 when the KRAS gene was found to be mutated in the lung tumor 4. Roles of RAS mutations in human cancers
but not in the blood cells of the patient [34]. In the later years, mutation
analyses found a frequent mutation of KRAS gene in the colon [45,46], The RAS mutations play an important role both in upstream and
lung [47] and pancreatic [48] cancers and it has been experimentally downstream of the onset of the disease, cancer. The upstream of the
demonstrated that the KRAS could cause early-onset lung cancer in onset disease shows the course of developing the disease that con-
mice if somatically activated [49]. The Kras but not Hras and Nras were stitutes the acquiring the RAS mutations, developing the disease and
shown to be important for the embryonic development in mouse [50]. exhibiting the first or initial phenotype of the disease with primary
Further, RAS was implicated in tumor maintenance [51]. In 1988, the symptoms. In a more simplified way the carcinogenesis process.
3D structure of human HRAS was determined [52], and the nucleotide Further, the downstream of the onset covers the possible outcome of the
exchange-mediated activation of RAS and farnesyl transferase activity disease and the frequency with which the disease could be expected in
was determined [53,54]. Further, studies were expanded to other iso- the future to occur. In a simplified way, the downstream of the cancer
forms of RAS, such as HRAS, and NRAS. In the following years, it has onset is the prognosis, a prediction of the course of a disease following
been found that RAS is one of the frequently mutated oncogenes in the disease onset.
human cancers [2,3,7,45]. In 1973, many studies were conducted on
the nucleic acid sequences of Kirsten-MSV and a model was proposed 4.1. Oncogenic mutations of RAS genes
for the formation of a mammalian RNA-containing sarcoma virus by
Scolnick et al and sarcoma virus-related RNA sequences were found in RAS oncogenes are mutated with different prevalence in human
normal rat cells by Tsuchida et al in 1974 [55–57]. Viral Kras and Hras cancers (Table 1). No specific pattern can be identified and/or specu-
genes were found to be originated from normal cellular genes. Although lated based on etiological and ethnic factors except some types of
after finding of ras genes opened the first door to the field of cancer cancers such as oral cancer, pancreatic cancer, and lung cancer. For
research in human cancers, which could never be accomplished from example, a relatively high incidence of HRAS mutations have been
src or mos researches since the src and mos genes mutations were never identified in oral cancers obtained from patients who were exposed to
been found in human cancers or it could be a rare event if there is. In some nitrosamine containing carcinogens through the habit of tobacco
1976, Michael Bishop and Harold E. Varmus in collaboration with Peter chewing, smoking and betel quid, etc. [60,61]. The possible role ex-
K. Vogt discovered that the DNA specific to the transforming avian erted by these carcinogens has been proved in experimental models by
sarcoma virus was present in normal avian DNA [58]. This research inducing HRAS single nucleotide point mutations in the tumor of a
paved the way for the Nobel Prize in physiology or medicine in 1989. mouse using nitrosomethylurea [62]. Many animal models have been
The Nobel Prize was jointly awarded to J. Michael Bishop and Harold E. demonstrated to show that tobacco-derived compounds could induce
Varmus for the discovery of the cellular origin of retroviral oncogenes KRAS mutations and that these mutations have occurred at the begin-
(Fig. 1). However, human src sequence has not been found as trans- ning of the tumorigenesis process [63]. Approximately, 7–8 fold in-
forming oncogene. Thus transforming RAS is the first human oncogene. creased incidence of KRAS mutation has been observed in a pancreatic
The history of human oncogenes and ras contribution to cancer re- cancer patient group with higher occupational exposure to organic
search was recently reviewed by Tsuchida et al [1]. solvents [64]. Similarly, tobacco smoke-induced KRAS mutation has
been a well-known fact for the past 20 years [8,65].
3. Identification of frequent RAS mutations in human cancers Although the RAS mutations are widely distributed throughout the
RAS genes, some amino acid residues are predominantly found to be
Finding of mutation in lung tumor ignited the researchers to search mutated in cancer (Fig. 2). For example, three amino acid residues such
for somatic mutations in other tumor types. Almost 35 years passed as G12, G13, and Q61 of all HRAS, KRAS, and NRAS are highly mutated
since the first mutation was found in 1984 [34]. Overall, RAS is mu- in human cancers (Figs. 2 & 3 ). These mutations prevent the intrinsic
tated approximately 30% of human cancers. To date, in HRAS, overall, and GAP catalyzed hydrolysis of GTP, thereby generating constantly
741 mutations have been found in 21,793 various tumor samples ac- active ras molecule [66]. In a normal cell, equilibrium is tightly
counting for 3.41% mutations. The prevalence of the mutation varies in maintained between the active and inactive state even though RAS
different types of primary tissues. For example, the highest incidence of proteins have a minimal and measurable activity on their own [61].
HRAS mutation has been found in salivary gland tumors (15%). This This condition is strictly maintained by GAPs (GTPase activating pro-
HRAS mutational incidence has truly been reflected also in the in- teins) and GEFs (Guanine nucleotide exchange factors) in the normal
dividual studies from various cohorts in oral squamous cell carcinomas physiological condition of the cell. The GAPs such as (NF1 and
(OSCC) of head and neck [59–61] and a relatively high incidence of p120GAP) accelerate the GTP hydrolysis of ras and the antagonist GEFs
mutations have also been found in cervical, upper aero-digestive tract such as ras-GRFs (p140 RAS-GRF1 and p130 RAS-GRF2) and RAS-GRPs
and urinary tract (9%) (Table1). (RASGRP1 and RASGRP 2) catalyze and weaken the GDP replacing with
In the KRAS, 21,724 mutations have been found from 95,963 GTP [67,68]. In the pathophysiological condition of a cell where RAS is
samples analyzed and accounting for 23% of mutations in human mutated, the equilibrium between the GTP and GDP-bound state is
cancers. The KRAS is found to be the most mutated oncogene in human impaired. The constitutively activated RAS molecule triggers the sig-
cancers. The highest incidence of mutations has been found in pan- naling on the downstream effectors that result in hyperactivation of
creatic cancer (57%). Relatively, a high incidence of mutations has also cellular pathways which control important cellular functions such as
been found in malignancies of the large intestine (35%), biliary tract growth, cell proliferation, survival, cytoskeleton rearrangements, ad-
(28%), small intestine (17%), lung (16%), endometrium (15%) and hesion, motility, and differentiation [66]. By this uncontrolled activa-
ovary (14%) as listed in Table 1. In the NRAS, 2410 mutations have tion, the cell undergoes the neoplastic transformation that results in
been identified in 32,104 tumors overall accounting for 7.51% of mu- cancer. RAS molecules and their various modulators in human cancer
tations in various primary tumor tissues. A very high incidence of NRAS were recently reviewed [2].

4
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 2. Schematic diagrams of the RAS oncogenes showing mutations reported in various human cancers.
A. Schematic illustration of HRAS gene. The HRAS is located on chromosome 11p15.5 and contains 4 exons with intervening sequences. Shown are various oncogenic
somatic mutations of the HRAS gene identified in various human cancers. B. Schematic sketch of KRAS gene. The KRAS has two sub-types such as KRAS A and B. The
KRAS is located on chromosome 12p12.1 and contains 4 exons with intervening sequences. All the oncogenic somatic mutations of the KRAS gene are marked in the
illustration. C. Schematic picture of NRAS gene. The NRAS is located on chromosome 1p13.2 and contains 4 exons with intervening sequences. Various human cancer-
associated oncogenic somatic mutations of NRAS gene are marked here. The missense somatic mutations identified in RAS genes of various human cancers were
derived and analyzed from COSMIC (www.sanger.ac.uk/genetics/CGP/cosmic/).

5
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 3. Space-fill model 3D structure and differential interacting network of the RAS oncoproteins.
A. Figure shows the space-fill model of HRAS. B. The space-fill model of KRAS. C) It shows the space-fill model of NRAS. Shown are the space-fill diagrams of a native
modeled structure of HRAS, KRAS, and NRAS, respectively. The protein structure PDB ID of HRAS, KRAS and NRAS are 121 P, 5VP7, and 5UHV, respectively. Amino
acid residues of the frequently mutated codons in human cancer were traced and plotted in RAS protein structures and protein 3D molecules were visualized by NGL
(WebGL) viewer. D. An interacting network of HRAS. E. An interacting network of KRAS. F. An interacting network of NRAS. Interacting network for each member of the
RAS protein was built by the STRING version 10.5 setting with highest confidence interaction score (0.900) and kmeans clustering with a minimum 5 number of
clusters. The result of the network analysis was imported as high-resolution portable network graphic (PNG) file.

6
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

Fig. 4. Mechanisms of the Ras activation and


its major signaling pathways.
The active-inactive cycle of RAS is tightly
controlled by GAPs (GTPase-activating pro-
teins) and GEFs (guanine-nucleotide exchange
factors). Mutated RAS is constitutively acti-
vated as it is constantly bound to GTP. Upon
the RAS activation, the MAPK, and PI3K/AKT,
the two major downstream signaling pathways
are activated and these pathways are asso-
ciated with growth, differentiation, develop-
ment, proliferation and survival.

4.2. Mutant RAS-induced constitutive activation of vital signaling pathways with the RAS-GTP bound form [80,81]. The association of PI3K activity
with RAS activation was reported by Lapetina and colleagues in 1991
Constitutively activated RAS activates various downstream sig- [82]. Subsequently, three years later, in 1994, the direct interaction of
naling molecules such as PI3K (Phosphatidylinositol 3-kinase), RAF (A p110α of PI3K with the RAS was demonstrated by Julian Downward
family of three serine/threonine-specific protein kinase, an acronym for and his group [78]. More specifically, binding of RAS to (p110α) PI3K
Rapidly Accelerated Fibrosarcoma), Rin1 (Ras and Rab Interactor 1), is necessary for RAS driven tumorigenesis [83]. Among the three RAS,
RASSF (Ras association domain family member), AF6 (Afadin protein), HRAS has been shown to be more efficient than KRAS in activating PI3K
PLCε (Phospholipase C-epsilon), RalGEF (Ras-like Guanine nucleotide [84]. RAS-mediated activation of PI3K generates abundantly the second
Exchange Factor), Tiam1 p190 (T-lymphoma invasion and metastasis 1 messenger PIP3 (phosphatidylinositol-3,4,5-triphosphate) from PIP2
p190), PKCξ (Protein kinase C epsilon type), etc., which control many (phosphatidylinositol-4,5-biphosphate). The tumor suppressor PTEN
cellular processes such as growth, cell proliferation, survival, cytoske- antagonizes PI3K activity by dephosphorylating the PIP3 to PIP2
leton rearrangements, adhesion, motility and differentiation including [80,81]. Generation of the second messenger, PIP3 recruits pleckstrin-
tumorigenesis [2,3,7,8,66]. As illustrated in Fig. 4, receptor tyrosine homology (PH) domain-containing proteins such as PDK1 and AKT/
kinases signaling were shown to be linked to Ras signaling via SH2 and PKB to the plasma membrane, and facilitates the activation and phos-
SH3 domain-containing adapter GRB2 [69]. The RAF1 mediated MAPK phorylation of AKT at threonine 308 by PDK1 [85]. AKT1 has been
cascade and the PI3K/AKT signaling pathways are the two key down- known to be involved in cell proliferation and survival, AKT2 has been
stream pathways of constitutively activated RAS signaling involved in linked to insulin-mediated metabolic process while AKT3 has been in-
tumorigenesis. The Ser/Thr kinase RAF1 and its interaction with RAS volved in cell size and cell number, altogether proven to play an im-
was found in 1993 by independently four groups and RAF1 was the first portant role in tumorigenesis [86]. Activated AKT initiates protein
mammalian RAS effector to be found [70–73]. Subsequent extensive synthesis through a cascade of its effector proteins such as mTOR
research found that the RAF1 signal through mitogen-activated protein (mammalian target of rapamycin) acting on to two critical downstream
kinases (MAPK). RAS interaction with RAF1 kinase facilitates the targets, S6K (p70S6K) and 4EBP1 (eukaryotic initiation factor 4E-
phosphorylation of two distinct serine residues of the dual specificity binding protein 1). MTORC2 phosphorylates and thereby additionally
protein kinases MEK1/2, which, in turn, activate by phosphorylating activates AKT at serine 473 [2,87]. AKT also regulates transcription-
threonine and tyrosine residues in the activation loop of the serine/ induced phosphorylation-dependent degradation of FOXO1 (forkhead
threonine-specific protein kinases ERK1/2 that results in constitutive box O transcription factor). Active PI3K also activates Rac [88].
activation of the MAPK signaling pathway. In addition, it has been Moreover, it has been found that RAS and RHO families of GTPases
shown that CRAF but not BRAF is required for KRAS (G12D)-mediated were shown to directly regulate various isoforms of PI3K [89]. Re-
initiation of lung cancer [74]. The activated MAPK cascade promotes cently, noncoding RNAs (ncRNAs) were shown to act as regulators of
cell differentiation, growth and cell cycle. Subsequent remarkable stu- RAS oncogenes, these ncRNAs epigenetically modulate the expression
dies on RAS-induced MAPK signaling found that this cascade is suffi- of RAS genes and loss of RAS-repressing ncRNAs results in upregulation
cient and important for RAS-induced transformation of murine cell lines of the RAS oncogenes [90]. These RAS mutation-induced constitutive
[75–77]. activation of various vital signaling molecules collectively result in
In addition to RAS-induced RAF1 mediated MAPK cascade, the PI3K uncontrolled cell proliferation and growth, cell survival and motility.
signaling pathway is another important RAS-induced pathway [78]. The ncRNAs-mediated epigenetic regulation also suggests an alter-
PI3K is a heterodimeric lipid kinase which is composed of a catalytic native and mutation-independent activation mechanism of RAS onco-
subunit (p110α) and a regulatory subunit (p85) [79]. The p110α con- genes (Fig. 4).
tains a RAS-binding domain (RBD) which enables p110α to interact

7
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

5. Role of RAS mutations in precision medicine for human cancer from bench to bedside. Recently, this task has been achieved only for
the G12C KRAS mutated form developing a small molecule inhibitor
Precision medicine can be defined as the tailoring of the therapeutic that actively bound covalently to cysteine and was shown to inhibit
strategies (developing effective, safe medications and doses) based on oncogenic RAS [94,95]. As seen in Fig. 5, it has been shown that a RAS
genetic (genomics, transcriptomics, proteomics), environmental and inhibitor, ARS853 could selectively interact to the GDP-bound KRAS
lifestyle factors. Pharmacogenomics becomes a part of the precision and block the nucleotide exchange which is important for the produc-
medicine as it deals the effect of genes on a person’s response to par- tion of GTP-bound form by the mutated codon 12 [96]. Similarly,
ticular drugs. Although "precision medicine" and "personalized medi- acetylation-mediated inhibition of oncogenic activity has been shown
cine overlap each other, National Research Council (NRC), USA ap- for the codon lysine 104 KRAS [97]. On the other hand, inhibition of
proved and preferred the term "precision medicine" rather than the nucleotide exchange itself demonstrated as an alternative therapeutic
term "personalized medicine" as the later one is relatively older and strategy, wherein a synthetic peptide has been shown to bind to the RAS
could be misinterpreted nevertheless the two terms are still used in- and inhibit the interaction of Son of Sevenless (SOS) that could result in
terchangeably by some people. RAS mutations could be used as an the prevention of GEF-induced nucleotide exchange [98]. These mole-
important factor in the determination of the therapeutic option for cular insights exploited the nucleotide exchange mechanisms-mediated
human cancer. RAS is one of the most sought therapeutic targets, di- activation of RAS molecules that helps to lock the mutant RAS mole-
agnostic and prognostic markers in human cancers. cules to the GDP-bound state as constantly inactive. Moreover, an al-
ternative mechanism has been suggested to explore, i.e., understanding
5.1. Targeted therapy for RAS mutation in human cancers differential activation of various effector pathways and utilize them
using various inhibitors as mutant alleles are not alike each other [99].
Developing an effective small molecule against the molecular target Furthermore, Tipifarnib – a RAS pathway inhibitor has been approved
is the initial phase of precision medicine. Direct inhibition of mutant by USFDA for the treatment of acute leukemia, especially in elderly
RAS has been attempted in various ways, however; it has been a big cases. Moreover, some peptidomimetics and bi-substrate inhibitors are
challenge forever with unique difficulties in each methodology due to under clinical trials (FTI 276, FTI 277, B956, B1086, L731, L735, L739,
multifaceted personalities of RAS oncoproteins. Principally RAS iso- L750, BMS-214662, and L778123) and they are likely to pave the way
forms displayed their variable activation across the human cancers that for precision therapy in RAS-mutant positive cases [100]. Recently, it
reflected the differential effects of RAS mutations [7,61,91]. Therefore, has been shown that inhibition of galectin-3, a glycoprotein that binds
to bypass these difficulties, the downstream signaling pathway of RAS to integrin αvβ3 sensitizes the mutant KRAS-addicted tumors to therapy
was targeted. Unlike other proteins, RAS signals through multiple and integrin αvβ3 might be used as a biomarker to identify mutant
mediators and effectors [7]. This complexity raised the question "which KRAS-addicted tumors. Moreover, galectin-3 directly binds to the cell
pathway should be targeted?” On the other hand, cancer is a multi- surface receptor, named integrin αvβ3 that result in KRAS addiction via
factorial genetic disease, potentially more than a gene is required for acquiring various functions of KRAS in transformed cells and that
the tumor initiation, maintenance, and metastasis and it would be makes the galectin-3 as a lynchpin for KRAS-addicted tumors [101].
practically not feasible to use a single inhibitor which could conquer the Recently, various small-molecule pan-RAS ligands have been synthe-
tumorigenesis. Therefore, more than an inhibitor is necessitated to see a sized and a compound (named 3144) was shown to bind to RAS pro-
clinically significant response in a setup/pathway where many genes teins. The small molecule found to interact with the oncogenic KRAS
are deregulated. Besides, it is also not possible to use more than two particularly with adjacent sites on the surface of KRAS. This pan-RAS
drugs/inhibitors as the drug itself could cause serious side-effects and inhibiting molecule showed promising and metabolically stable anti-
most of the cancer drugs targets invariably both the normal and cancer cancer effect in the xenograft mouse models and pose an effective anti-
cells. Therefore, minimal drug and clinically a maximal effect are cancer therapeutic strategy for human cancers with RAS mutations
needed. This prompted the researchers to narrow down the number of [102]. Although drugging RAS has initial failures, after great effort and
targets based on the direct target and closely signaling pathways which understanding the complexities of RAS now armed with new directions
are involved in proliferation, growth, tumorigenesis, survival, and anti- with various technologies [103]. Recently, the NCI (National Cancer
apoptosis. Institute) has initiated a suite for next-generation anti-RAS drug dis-
Well-characterized and the direct downstream of the RAS signaling covery [4]. This initiative gives a big hope that could develop new
are the MAPK and PI3K signaling pathways [92] and MEK and AKT armor which alters the undruggable RAS to druggable. This would help
inhibitors block these two pathways, respectively. Though the MAPK in utilizing various pathway-specific inhibitors which are in the market
pathway is preferentially activated by RAS, it has also been known to and clinical trials for the cancer patients bearing RAS mutants with
interact with p110α, the catalytic subunit of PI3K and PI3K signaling is codon-specific pathway activation.
firmly established under RAS [83]. Furthermore, in KRAS driven col-
orectal tumor, the PIK3CA mutation increase the resistance to EGFR 5.2. Inhibition of RAS molecules in human cancers and the mechanism of
therapy [93]. Therefore, the PI3K pathway inhibition becomes neces- resistance
sary. The use of these inhibitors may be an alternative, appropriate and
a promising strategy for targeting RAS-mediated signaling pathways in Targeting RAS itself is a cumbersome matter as each mutant of these
human cancers because this therapeutic strategy not only inhibits RAS- RAS molecules exhibits differential phenotypes; moreover, the majority
mediated signaling, but it paves also the way for inhibiting other ge- of the cancer therapies fails as a result of resistance. It has been clearly
netically altered upstream and downstream effectors of RAS. Moreover, demonstrated that the inhibition of RAS oncogenes could cause a dra-
this strategy also inhibits many other cancer-associated activated pro- matic regression of various tumors in mouse models. Nevertheless,
tein molecules which trigger the growth signal through AKT and MEK. emerging of tumor resistance as a result of various new RAS-in-
This may have a synergistic effect on both PI3K/AKT pathway and RAS- dependent clones was observed which were mostly shown to be driven
RAF-MEK-ERK cascade. The molecular targeted therapy (MTT) acting by the YAP1 gene [104]. Similarly, this phenomenon has been alter-
on these two pathways should be taken into account at least in a subset natively demonstrated showing that YAP1 gene overexpression could
of patients with RAS mutations. rescue cancer cells from the depletion of KRAS molecules [105]. Cur-
In the future, the precision medicine will depend mainly on the rently, HRAS is inhibited using farnesyl transferase inhibitors which are
genetic makeup of the patients as their genotype could drive the best- presently used in clinical trials and mechanism of resistance if any
targeted therapies. To this end, finding allele-specific inhibitors for RAS would tend to emerge in the future. The major advantage of the RAS
mutants would predominately bring success to the precision medicine inhibitors is that they could likely relieve on the feedback loop in the

8
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

pancreatic intra-epithelial neoplasia (PanIN) lesions. Accuracy rates of


distinguishing mice with early or late stage lesions versus their re-
spective controls were 82.1% and 81.5%, respectively [108]. Effective
management of pancreatic intraductal papillary mucinous neoplasms
(IPMN) and mucinous cystic neoplasms (MCN) are exclusively based on
distinguishing accurately the mucinous cyst from other pancreatic
cystic lesions. In fact, a recent preoperative testing KRAS in the diag-
nosis of pancreatic mucinous cysts showed KRAS mutations in 28 (68%)
IPMNs, 7 (78%) IPMNs with adenocarcinoma, and 1 (6%) MCN. KRAS
showed 100% specificity and 65% sensitivity for mucinous differ-
entiation. In the case of IPMNs, KRAS had 98% specificity and 84%
sensitivity [109]. In addition, the integration of KRAS testing in 618
pancreatic cysts showed a specificity of 100%, but a sensitivity of 54%
for mucinous differentiation [110]. The KRAS is frequently mutated in
colon cancer next to pancreatic cancer.
A study using the chip-based digital PCR for detection of circulating
tumor DNA in metastatic colon cancer revealed the detection of ctDNA
Fig. 5. Trapping mechanisms of the KRAS G12C allele-specific inhibitor. (circulating tumor DNA) in 11/16 cases (68.75%) with KRAS-mutated
A RAS inhibitor, ARS853 selectively interact to the GDP-bound KRAS codon tumors. It was consistent with the detection rate of the chip-based dPCR
G12C allele and block the nucleotide exchange which is important for the (96%) and KRAS mutation was detected in 18 wild-type tumor cases
production of GTP-bound form by the mutated codon 12 whereby lock the with a sensitivity and specificity of 69% and 100%, respectively [111].
mutant KRAS molecules to the GDP-bound state as constantly inactive. Moreover, the fecal immunochemical test (FIT) has also been used as
one of the diagnostic tests to detect the KRAS mutation for diagnosis of
colon cancer [112]. Although RAS mutations incidence is relatively low
upstream growth factor receptors. Although oncogenic mutations of in bladder cancer, RAS is used as a potential diagnostic marker in urine
RAS, BRAF, and MEK genes are mutually exclusive in this MAPK samples especially, the HRAS along with the TERT, FGFR3, and PIK3CA.
pathway, combination therapy may be necessary if RAS inhibitors are The concomitant presence of these gene mutations in urine samples was
used in clinics to overcome a concomitant mutation of BRAF/MEK. not often revealed [113].
Recently, it has been demonstrated that Kras G12D “knockin’’ mutant- In the thyroid, nodules are evaluated by fine-needle aspiration
generated primary murine acute myeloid leukemia (AML) responded to (FNA) cytology and 20–30% of FNAs often shows indeterminate cy-
MEK inhibitor PD0325901 (PD901) and showed intrinsic drug re- tology that challenges the relevant management options [114]. Mostly,
sistance at relapse. On the other hand, they found that the resistance follicular or oncocytic (FN)/suspicious follicular or oncocytic neoplasia
cases were sensitive to MEK inhibition when the wild-type KRAS is lost. (SFN) is the common indeterminate diagnosis that poses the cancer risk
This was also reflected in the colon cancer cell lines when the WT KRAS of ∼15–30%. A ThyroSeq v2 next-generation sequencing assay-based
was changed to a mutant allele that caused the heterozygous mutant- approach for the diagnosis of cancer in thyroid nodules including FN/
bearing cells sensitive to treatment while the KRAS mutants were al- SFN identified NRAS followed by KRAS as the most commonly mutated
ready highly sensitive to MAPK inhibition in HCT116 cells. They also oncogenes in the nodules. This assay showed 90% sensitivity and 93%
showed that in advanced cancer cases, 642 of 1168 (55%) patients with specificity with 92% accuracy and the positive and negative predictive
KRAS mutations showed allelic imbalance suggesting that the genetic value was 83% and 96%, respectively [115]. A recent study found 17 of
changes at KRAS locus are common in human cancer and they are likely 362 nodules positive for RAS mutations out of which 8 (8/17, 47%)
to be dependent (sensitive) on MEK inhibition [106]. proved to be malignant after surgery and also showed that the RAS-
positive nodules had an indolent behavior for many years as no extra-
5.3. The utility of RAS mutations as diagnostic markers in human cancers thyroidal extension, metastases, or lymphovascular invasion with low
risk [116].
The currently available diagnostic tests mainly and mostly show a
lack of sensitivity and specificity to identify the markers for early stages 5.4. Use of RAS mutations as prognostic markers in human cancers
of cancer regardless of molecular, metabolomics, invasive, non-invasive
nature of the test. Many studies have been carried out to determine the RAS is one of the most mutated oncogenes in human cancers. EGFR
diagnostic utility of KRAS mutations but the results were highly het- is one of the other most genetically altered oncogenes in human cancers
erogeneous and contradictory. Pancreatic cancer is one of the most and it is an upstream effector of RAS signaling. The EGFR is ther-
lethal human cancers with less than 3% overall 5-year survival rate and apeutically targeted using anti-EGFR antibody-mediated therapy,
requires more clinically meaningful tests for early diagnosis. KRAS is especially in lung cancer and colon cancer. Recently, mutation status of
the most mutated oncogene in pancreatic cancer. Various methods have KRAS is determined before the EGFR-targeted therapy in human can-
been used to detect KRAS mutations in cytological tissues as a mole- cers, especially in lung and colon cancers [117,118]. Furthermore, the
cular marker for the diagnosis of pancreatic cancer [107]. It has been determination of the presence or absence of the KRAS mutation has
reported that KRAS mutations were mostly detected in cells derived been considered as an important key factor to predict prognosis and/or
from invasive methods such as fine needle aspiration (FNA), endoscopic the treatment method in these cancers [117,118]. Because of the pre-
retrograde cholangiopancreatography (ERCP), and surgery. When the sence of a KRAS mutation is observed as an adverse prognostic factor
first two (FNA and ERCP) were compared with surgery, the pooled [119] and also a predictor of failure to benefit from EGFR-targeted
sensitivity was 77% versus 54% and the specificity was 88% and the therapy [120–122]. Apart from the lack of response, it has been
diagnostic odds ratio (DOR) was 20.2 versus 7.52, respectively. Further, documented that KRAS mutations worsen the response when EGFR-
the study suggested that diagnostic accuracy was significantly im- targeted therapy was added to their standard chemotherapy [121].
proved when two methods were combined. A mouse model with age- Approximately, 40% of colorectal cancers harbor with KRAS mu-
matched wild-type mice compared to p48-Cre/LSL-KrasG12D showed tations [123], among them, 90% of mutations are detected in the codon
that malignant progression of pancreatic ductal adenocarcinoma 12 and 13 while the remaining ones occur in codon 61 and 146
(PDAC) is analogous to the human disease stages via early and late [124,125]. It has been predicted that mutation in the codon 12, 13 and

9
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

61 may show a lack of response to cetuximab while codon 146 muta- cancer and the KRAS G12D mutant could be a poor prognostic marker
tions did not affect cetuximab efficacy [125]. In contrast, EGFR ex- in this cancer [140]. Similarly, non–small cell lung cancer (NSCLC)
pression in colon cancer failed to demonstrate predictive value for anti- patients with KRAS-G12C/G12 V exhibited worse progression-free sur-
EGFR monoclonal antibody therapy. The intensity of EGFR staining by vival compared with the patients bearing other KRAS mutations/wild-
IHC analysis showed no correlation with the response rate to cetuximab type. Interestingly, NSCLC cell lines with KRAS-G12C/G12 V mutants
and panitumumab [126,127]. Therefore, KRAS mutation in human triggered the Ral signaling and abolished the growth factor dependence
cancer is an established predictor of response to EGFR-targeted therapy. for AKT activation. On the other hand, KRAS-G12D mutants activated
As it is well evidenced, obtaining a comprehensive mutational profile is both the PI3K/AKT and MAPK signaling. Molecular modeling on these
clinically highly necessary and this may help in treatment selection in a mutants revealed that different confirmations resulted from various
molecular targeted therapy. mutants likely to have differential binding efficiency with the down-
stream signaling members which transduce heterogeneous mutant-
5.5. RAS mutations alter the outcome of the disease specific signaling [136]. These data suggest that KRAS mutants have
differential oncogenic signaling and patient outcome. Moreover, the
Although all RAS genes are mutated at various extents in human differential effect of KRAS mutants implies that therapeutic strategies to
cancers, the KRAS mutations show the most frequent mutations and be catered to the tumors based on the status of amino acid-specific
influence the clinical outcome of the KRAS-associated tumors to the KRAS mutation. Recently, KRAS-specific inhibitors (Deltarasin & Sal-
worst condition. KRAS mutations are associated with clinical outcomes, irasib) have been synthesized and clinically evaluated [100]. NCI in-
such as inferior survival in colon cancer patients [128,129] and me- itiative on drugging RAS may bring hope for patients with RAS muta-
tastasis and invasion in pancreatic cancer [130]. The highest incidences tions particularly for KRAS as this gene is predominantly implicated in
of codons 12 and 13 mutations have been detected in colorectal cancer human cancers especially in aggressive phenotypes such as pancreatic,
with liver metastasis (CRLM) and these mutations (G12V/C/S) were colon and lung cancer, and the KRAS seems to be much more important,
correlated with poor overall survival [131]. In a mouse system, PI3K followed by NRAS and HRAS [4,141]. Nonetheless, biochemistry has
activity had been reported to be essential both for the initiation and the been relatively advanced with HRAS but that of KRAS was slightly
maintenance of tumors with the KRASG12D genotype and this activity delayed. Recently, it has been demonstrated that cancers with KRAS
requires KRASG12D-PIK3CA (p110α) binding [3,83]. In addition, RAS mutations could be targeted with a covalent G12C-specific small mo-
interaction with PIK3CA (p110α) is required for maintenance of lung lecule inhibitor [142]. Further, a recent study traced the evolutionary
tumor and tumor-induced angiogenesis [132, 133]. In human carcino- routes of KRAS dosage variation and their role in cancer phenotypic
genesis, the mutated PIK3CA was demonstrated to be associated with difference, early tumorigenesis, and metastasis in pancreatic cancer
invasion [134]. Meanwhile, mCRC (metastatic colorectal cancer) pa- [143]. This uncovers the truth behind the differential cancer pheno-
tients with brain metastasis predominantly had the KRASG12D geno- types for a specific codon of KRAS.
type rather than the other genotypes of codon 12 [135]. The
KRASG12D in NSCLC cell lines has more preferential signaling to the 7. Conclusion and future prospects
PI3K pathway than to the Raf pathway [136]. It was reported that the
same mutant was a stronger stimulant of the Raf-ERK pathway than of The RAS is one of the genetically most deregulated oncogenes in
the PI3K-AKT pathway in lymphoid neoplasia [137]. Therefore, further human cancers. HRAS, KRAS, and NRAS are mutated with the highest
studies are needed to elucidate the detailed mechanisms of signaling to frequency in salivary gland (15%), pancreatic (57%) and skin cancer
downstream effectors, including differences in tumor cell types, KRAS (17%), respectively. More than three decades of RAS research have
genotypes (amino acid substitutions), and isotypes (KRAS4A/KRAS4B). unraveled the important role played by RAS genes in human cancers.
It has been reported that KRAS4A mRNA was widely expressed in ex- Detection and exploitation of RAS mutations for molecular targeted
amined human cancer cell lines, though KRAS4B mRNA was present at therapy and as a diagnostic and prognostic marker in precision medi-
higher levels, but, amounts approximately equal in most cases of 17 cine is an indicator of the best therapeutic approach. In future, a po-
fresh human colorectal tumors [138]. tential and effective RASopathy would exclusively be depending on
designing various potent inhibitors which could significantly inhibit the
6. KRAS: a prime focus unique, multifaceted and differential personalities of various alleles of
mutant RAS molecules and their closely cooperating partners with least
Overall KRAS oncogene is the most frequently mutated (∼23%) side effects.
oncogene in human cancers compared with the other counter RAS
subtypes, HRAS (3.4%) and NRAS (7.5%). Moreover, KRAS is specifi- Acknowledgements
cally deregulated in the most aggressive human cancers such as pan-
creatic (∼57%), large intestine/colon (∼35%), biliary tract (˜∼28), This work was partly supported by Grants-in-Aid from The Ministry
lung (∼17%) suggesting an important role of KRAS in these carcino- of Education, Culture, Sports, Science and Technology (MEXT), Japan
geneses. This observation paved way for PRIME and CRYSTAL, two [Grant #06044072], Japan Society for the Promotion of Sciences
large-scale clinical trials. The prime idea was to incorporate anti-EGFR [Grant #257132] and National Institute of Health (NIH), U.S.A. [Grant
mAbs as first-line chemotherapy for metastatic CRC patients however, #CA22701] to N. Tsuchida as the P.I. We (A.K. Murugan and N.
this therapy exhibited clear survival benefit only for cases with wt- Tsuchida) thank Government of Japan for awarding “Monbukagakusho:
KRAS and NRAS. Moreover, this therapy showed worse/no benefit on MEXT Scholarship" to A.K. Murugan. We apologize that we could not
overall survival for cases bearing mutant KRAS/NRAS. Therefore, cur- cite some of the remarkable work of our colleagues mainly due to space
rently, cetuximab and panitumumab-mediated treatments are re- limitations.
commended only for the patients bearing wt-RAS suggesting that
evolving a KRAS-dependent precision medicine for mCRC [139]. The References
KRAS codon 12 mutation subtypes seem to be different in biologic ac-
tivities while KRAS of G12D mutation is associated with poor prognosis. [1] N. Tsuchida, A.K. Murugan, M. Grieco, Kirsten Ras* oncogene: significance of its
Recent findings showed that the KRAS mutations have a differential discovery in human cancer research, Oncotarget 7 (2016) 46717–46733.
[2] D.K. Simanshu, D.V. Nissley, F. McCormick, RAS proteins and their regulators in
role even in a specific tumor type. The KRAS G12D mutants showed human disease, Cell 170 (2017) 17–33.
poorer median survival when compared with Non-G12D KRAS mutants [3] K.M. Haigis, KRAS alleles: the devil is in the detail, Trends Cancer 3 (2017)
and they mostly represented with advanced tumor stage in ampullary 686–697.

10
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[4] D. Esposito, A.G. Stephen, T.J. Turbyville, M. Holderfield, New weapons to pe- for the acquisition of transforming properties by the T24 human bladder carci-
netrate the armor: novel reagents and assays developed at the NCI RAS initiative noma oncogene, Nature 300 (1982) 149–152.
to enable discovery of RAS therapeutics, Semin. Cancer Biol. (2018), https://doi. [37] R.J. Muschel, G. Khoury, P. Lebowitz, R. Koller, R. Dhar, The human c-ras1H
org/10.1016/j.semcancer.2018.02.006 pii: S1044-579X(17)30279-1. oncogene: a mutation in normal and neoplastic tissue from the same patient,
[5] M. Barbacid, ras genes, Annu. Rev. Biochem. 56 (1987) 779–827. Science 219 (1983) 853–856.
[6] J. Colicelli, Human RAS superfamily proteins and related GTPases, Sci. STKE 250 [38] P.H. Duesberg, Activated proto-onc genes: sufficient or necessary for cancer?
(2004) RE13. Science 228 (1985) 669–677.
[7] A.E. Karnoub, R.A. Weinberg, Ras oncogenes: split personalities, Nat. Rev. Mol. [39] H. Land, L.F. Parada, R.A. Weinberg, Tumorigenic conversion of primary embryo
Cell Biol. 9 (2008) 517–531. fibroblasts requires at least two cooperating oncogenes, Nature 304 (1983)
[8] M. Malumbres, M. Barbacid, RAS oncogenes: the first 30 years, Nat. Rev. Cancer 3 596–602.
(2003) 459–465. [40] R.F. Newbold, R.W. Overell, Fibroblast immortality is a prerequisite for transfor-
[9] P.A. Rous, Transmissible avian neoplasm. (sarcoma of the common fowl), J. Exp. mation by EJ c-Ha-ras oncogene, Nature 304 (1983) 648–651.
Med. 12 (1910) 696–705. [41] H.E. Ruley, Adenovirus early region 1A enables viral and cellular transforming
[10] P.A. Rous, Sarcoma of the fowl transmissible by an agent separable from the tumor genes to transform primary cells in culture, Nature 304 (1983) 602–606.
cells, J. Exp. Med. 13 (1911) 397–411. [42] B.M. Willumsen, A. Christensen, H.L. Hubbert, A.G. Papageorge, D.R. Lowy, The
[11] H.L. Van Epps, Peyton Rous: father of the tumor virus, J. Exp. Med. 201 (2005) p21ras c-terminus is required for transformation and membrane association,
320. Nature 310 (1984) 583–586.
[12] J.J. Harvey, An unidentified virus which causes the rapid production of tumors in [43] A. Hall, C.J. Marshall, N.K. Spurr, R.A. Weiss, Identification of transforming gene
mice, Nature 204 (1964) 1104–1105. in two human sarcoma cell lines as a new member of the ras gene family located
[13] W.H. Kirsten, L.A. Mayer, Morphologic responses to a murine erythroblastosis on chromosome 1, Nature 303 (1983) 396–400.
virus, J. Natl. Cancer Inst. 39 (1967) 311–335. [44] H. Umanoff, W. Edelmann, A. Pellicer, R. Kucherlapati, The murine N-ras gene is
[14] R.L. Peters, L.S. Rabstein, R. VanVleck, G.J. Kelloff, R.J. Huebner, Naturally oc- not essential for growth and development, Proc. Natl. Acad. Sci. U. S. A. 92 (1995)
curring sarcoma virus of the BALB/cCr mouse, J. Natl. Cancer Inst. 53 (1974) 1709–1713.
1725–1729. [45] J.L. Bos, Ras oncogenes in human cancer: a review, Cancer Res. 49 (1989)
[15] S. Rasheed, M.B. Gardner, R.J. Huebner, In vitro isolation of stable rat sarcoma 4682–4689.
viruses, Proc. Natl. Acad. Sci. U. S. A. 75 (1978) 2972–2976. [46] K. Forrester, C. Almoguera, K. Han, W.E. Grizzle, M. Perucho, Detection of high
[16] G.L. Hager, E.H. Chang, H.W. Chan, C.F. Garon, M.A. Israel, M.A. Martin, et al., incidence of K-ras oncogenes during human colon tumorigenesis, Nature 327
Molecular cloning of the Harvey sarcoma virus closed circular DNA intermediates: (1987) 298–303.
initial structural and biological characterization, J. Virol. 31 (1979) 795–809. [47] S. Rodenhuis, M.L. van de Wetering, W.J. Mooi, S.G. Evers, N. van Zandwijk,
[17] N. Tsuchida, S. Uesugi, Structure and functions of the Kirsten murine sarcoma J.L. Bos, Mutational activation of the K-ras oncogene. A possible pathogenetic
virus genome: molecular cloning of biologically active Kirsten murine sarcoma factor in adenocarcinoma of the lung, N. Engl. J. Med. 317 (1987) 929–935.
virus DNA, J. Virol. 38 (1981) 720–727. [48] C. Almoguera, D. Shibata, K. Forrester, J. Martin, N. Arnheim, M. Perucho, Most
[18] N. Tsuchida, E. Ohtsubo, T. Ryder, Nucleotide sequence of the oncogene encoding human carcinomas of the exocrine pancreas contain mutant c-K-ras genes, Cell 53
the p21 transforming protein of Kirsten murine sarcoma virus, Science 217 (1982) (1988) 549–554.
937–939. [49] L. Johnson, K. Mercer, D. Greenbaum, R.T. Bronson, D. Crowley, D.A. Tuveson,
[19] R.W. Ellis, D. Defeo, T.Y. Shih, M.A. Gonda, H.A. Young, N. Tsuchida, et al., The et al., Somatic activation of the K-ras oncogene causes early onset lung cancer in
p21 src genes of Harvey and Kirsten sarcoma viruses originate from divergent mice, Nature 410 (2001) 1111–1116.
members of a family of normal vertebrate genes, Nature 292 (1981) 506–511. [50] K. Koera, K. Nakamura, K. Nakao, J. Miyoshi, K. Toyoshima, T. Hatta, et al., K-ras
[20] K. Shimizu, M. Goldfarb, Y. Suard, M. Perucho, Y. Li, T. Kamata, et al., Three is essential for the development of the mouse embryo, Oncogene 15 (1997)
human transforming genes are related to the viral ras oncogenes, Proc. Natl. Acad. 1151–1159.
Sci. U. S. A. 80 (1983) 2112–2116. [51] L. Chin, A. Tam, J. Pomerantz, M. Wong, J. Holash, N. Bardeesy, et al., Essential
[21] K. Shimizu, D. Birnbaum, M.A. Ruley, O. Fasano, Y. Suard, L. Edlund, et al., role for oncogenic Ras in tumour maintenance, Nature 400 (1999) 468–472.
Structure of the Ki-ras gene of the human lung carcinoma cell line Calu-1, Nature [52] A.M. de Vos, L. Tong, M.V. Milburn, P.M. Matias, J. Jancarik, S. Noguchi, et al.,
304 (1983) 497–500. Three-dimensional structure of an oncogene protein: catalytic domain of human c-
[22] C. Shih, B.Z. Shilo, M.P. Goldfarb, A. Dannenberg, R.A. Weinberg, Passage of H-ras p21, Science 239 (1988) 888–893.
phenotypes of chemically transformed cells via transfection of DNA and chro- [53] J. Downward, R. Riehl, L. Wu, R.A. Weinberg, Identification of a nucleotide ex-
matin, Proc. Natl. Acad. Sci. U. S. A. 76 (1979) 5714–5718. change-promoting activity for p21ras, Proc. Natl. Acad. Sci. U. S. A. 87 (1990)
[23] C. Shih, L.C. Padhy, M. Murray, R.A. Weinberg, Transforming genes of carcinomas 5998–02.
and neuroblastomas introduced into mouse fibroblasts, Nature 290 (1981) [54] Y. Reiss, J.L. Goldstein, M.C. Seabra, P.J. Casey, M.S. Brown, Inhibition of purified
261–264. p21ras farnesyl:protein transferase by Cys-AAX tetrapeptides, Cell 62 (1990)
[24] M. Perucho, M. Goldfarb, K. Shimizu, C. Lama, J. Fogh, M. Wigler, Human-tumor- 81–88.
derived cell lines contain common and different transforming genes, Cell 27 [55] P. Duesberg, Antecedents of a nobel prize, Nature 343 (1990) 302–303.
(1981) 467–476. [56] E.M. Scolnick, E. Rands, D. Williams, W.P. Parks, Studies on the nucleic acid se-
[25] C.J. Tabin, S.M. Bradley, C.I. Bargmann, R.A. Weinberg, A.G. Papageorge, quences of Kirsten sarcoma virus: a model for formation of a mammalian RNA-
E.M. Scolnick, et al., Mechanism of activation of a human oncogene, Nature 300 containing sarcoma virus, J. Virol. 12 (1973) 458–463.
(1982) 143–149. [57] N. Tsuchida, R.V. Gilden, M. Hatanaka, Sarcoma-virus-related RNA sequences in
[26] J.P. McGrath, D.J. Capon, D.H. Smith, E.Y. Chen, P.H. Seeburg, D.V. Goeddel, normal rat cells, Proc. Natl. Acad. Sci. U. S. A. 71 (1974) 4503–4507.
et al., Structure and organization of the human Ki-ras proto-oncogene and a re- [58] D. Stehelin, H.E. Varmus, J.M. Bishop, P.K. Vogt, DNA related to the transforming
lated processed pseudogene, Nature 304 (1983) 501–506. gene(s) of avian sarcoma viruses is present in normal avian DNA, Nature 260
[27] E. Taparowsky, Y. Suard, O. Fasano, K. Shimizu, M. Goldfarb, M. Wigler, (1976) 170–173.
Activation of the T24 bladder carcinoma transforming gene is linked to a single [59] A.K. Munirajan, B.K.C. Mohanprasad, G. Shanmugam, N. Tsuchida, Detection of a
amino acid change, Nature 300 (1982) 762–765. rare point mutation at codon 59 and relatively high incidence of H-ras mutation in
[28] D.J. Capon, P.H. Seeburg, J.P. McGrath, J.S. Hayflick, U. Edman, A.D. Levinson, Indian oral cancer, Int. J. Oncol. 13 (1998) 971–974.
et al., Activation of Ki-ras2 gene in human colon and lung carcinomas by two [60] A.K. Murugan, N.T. Hong, T.T. Cuc, N.C. Hung, A.K. Munirajan, M.A. Ikeda,
different point mutations, Nature 304 (1983) 507–513. N. Tsuchida, Detection of two novel mutations and relatively high incidence of H-
[29] C. Shih, R.A. Weinberg, Isolation of a transforming sequence from a human RAS mutations in Vietnamese oral cancer, Oral Oncol. 45 (2009) e161–6.
bladder carcinoma cell line, Cell 29 (1982) 161–169. [61] A.K. Murugan, A.K. Munirajan, N. Tsuchida, Ras oncogenes in oral cancer: the past
[30] M. Goldfarb, K. Shimizu, M. Perucho, M. Wigler, Isolation and preliminary char- 20 years, Oral Oncol. 48 (2012) 383–392.
acterization of a human transforming gene from T24 bladder carcinoma cells, [62] S. Sukumar, V. Notario, D. Martin-Zanca, M. Barbacid, Induction of mammary
Nature 296 (1982) 404–409. carcinomas in rats by nitroso-methylurea involves malignant activation of H-ras-1
[31] S. Pulciani, E. Santos, A.V. Lauver, L.K. Long, K.C. Robbins, M. Barbacid, locus by single point mutations, Nature 306 (1983) 658–661.
Oncogenes in human tumor cell lines: molecular cloning of a transforming gene [63] R. Lochan, H.L. Reeves, A.K. Daly, R.M. Charnley, The role of tobacco-derived
from human bladder carcinoma cells, Proc. Natl. Acad. Sci. U. S. A. 79 (1982) carcinogens in pancreas cancer, ISRN Oncol. 2011 (2011) 249235.
2845–2849. [64] J. Alguacil, M. Porta, N. Malats, T. Kauppinen, M. Kogevinas, F.G. Benavides,
[32] Krontiris T.G. Der CJ, G.M. Cooper, Transforming genes of human bladder and et al., Occupational exposure to organic solvents and K-ras mutations in exocrine
lung carcinoma cell lines are homologous to the ras genes of Harvey and Kirsten pancreatic cancer. PANKRAS II Study Group, Carcinogenesis 23 (2002) 101–106.
sarcoma viruses, Proc. Natl. Acad. Sci. U. S. A. 79 (1982) 3637–3640. [65] F. Le Calvez, A. Mukeria, J.D. Hunt, O. Kelm, R.J. Hung, P. Tanière, et al., P53 and
[33] L.F. Parada, C.J. Tabin, C. Shih, R. Weinberg, Human EJ bladder carcinoma on- KRAS mutation load and types in lung cancers in relation to tobacco smoke: dis-
cogene is homologue of Harvey sarcoma virus ras, Nature 297 (1982) 474–478. tinct patterns in never, former, and current smokers, Cancer Res. 65 (2005)
[34] E. Santos, D. Martin-Zanca, E.P. Reddy, M.A. Pierotti, G. Della Porta, M. Barbacid, 5076–5083.
Malignant activation of a K-ras oncogene in lung carcinoma but not in normal [66] K. Rajalingam, R. Schreck, U.R. Rapp, S. Albert, Ras oncogenes and their down-
tissue of the same patient, Science 223 (1984) 661–664. stream targets, Biochim. Biophys. Acta 1773 (2007) 1177–1195.
[35] L.F. Parada, R.A. Weinberg, Presence of a Kirsten murine sarcoma virus ras on- [67] P. Polakis, F. McCormick, Structural requirements for the interaction of p21ras
cogene in cells transformed by 3-methylcholanthrene, Mol. Cell. Biol. 3 (1983) with GAP, exchange factors, and its biological effector target, J. Biol. Chem. 268
2298–2301. (1993) 9157–9160.
[36] E.P. Reddy, R.K. Reynolds, E. Santos, M. Barbacid, A point mutation is responsible [68] I.M. Ahearn, K. Haigis, D. Bar-Sagi, M.R. Philips, Regulating the regulator: post-

11
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

translational modification of RAS, Nat. Rev. Mol. Cell Biol. 13 (2011) 39–51. Multivalent small-molecule pan-RAS inhibitors, Cell 168 (2017) 878-89.e29.
[69] E.J. Lowenstein, R.J. Daly, A.G. Batzer, W. Li, B. Margolis, R. Lammers, A. Ullrich, [103] B. Papke, C.J. Der, Drugging RAS: know the enemy, Science 355 (2017)
et al., The SH2 and SH3 domain-containing protein GRB2 links receptor tyrosine 1158–1163.
kinases to Ras signalling, Cell 70 (1992) 431–442. [104] A. Kapoor, W. Yao, H. Ying, S. Hua, A. Liewen, Q. Wang, et al., Yap1 activation
[70] S.A. Moodie, B.M. Willumsen, M.J. Weber, A. Wolfman, Complexes of Ras.GTP enables bypass of oncogenic Kras addiction in pancreatic cancer, Cell 158 (2014)
with Raf-1 and mitogen-activated protein kinase kinase, Science 260 (1993) 185–197.
1658–1661. [105] D.D. Shao, W. Xue, E.B. Krall, A. Bhutkar, F. Piccioni, X. Wang, et al., KRAS and
[71] P.H. Warne, P.R. Viciana, J. Downward, Direct interaction of Ras and the amino- YAP1 converge to regulate EMT and tumor survival, Cell 158 (2014) 171–184.
terminal region of Raf-1 in vitro, Nature 364 (1993) 352–355. [106] M.R. Burgess, E. Hwang, R. Mroue, C.M. Bielski, A.M. Wandler, B.J. Huang, et al.,
[72] X.F. Zhang, J. Settleman, J.M. Kyriakis, E. Takeuchi-Suzuki, S.J. Elledge, KRAS allelic imbalance enhances fitness and modulates MAP kinase dependence in
M.S. Marshall, et al., Normal and oncogenic p21ras proteins bind to the amino- cancer, Cell 168 (2017) 817–829.
terminal regulatory domain of c-Raf-1, Nature 364 (1993) 308–313. [107] J. Yang, J. Li, R. Zhu, H. Zhang, Y. Zheng, W. Dai, et al., K-ras mutational status in
[73] A.B. Vojtek, S.M. Hollenberg, J.A. Cooper, Mammalian Ras interacts directly with cytohistological tissue as a molecular marker for the diagnosis of pancreatic
the serine/threonine kinase Raf, Cell 74 (1993) 205–214. cancer: a systematic review and meta-analysis, Dis. Markers 2014 (2014) 573783.
[74] F.A. Karreth, K.K. Frese, G.M. DeNicola, M. Baccarini, D.A. Tuveson, C-Raf is re- [108] J.J. LaConti, E.C. Laiakis, A.D. Mays, I. Peran, S.E. Kim, J.W. Shay, et al., Distinct
quired for the initiation of lung cancer by K-RasG12D, Cancer Discov. 1 (2011) serum metabolomics profiles associated with malignant progression in the
128–136. KrasG12D mouse model of pancreatic ductal adenocarcinoma, BMC Genomics 16
[75] S.J. Leevers, H.F. Paterson, C.J. Marshall, Requirement for Ras in Raf activation is (2015) S1.
overcome by targeting Raf to the plasma membrane, Nature 369 (1994) 411–414. [109] A.D. Singhi, M.N. Nikiforova, K.E. Fasanella, K.M. McGrath, R.K. Pai, N.P. Ohori,
[76] D. Stokoe, S.G. Macdonald, K. Cadwallader, M. Symons, J.F. Hancock, Activation et al., Preoperative GNAS and KRAS testing in the diagnosis of pancreatic muci-
of Raf as a result of recruitment to the plasma membrane, Science 264 (1994) nous cysts, Clin. Cancer Res. 20 (2014) 4381–4389.
1463–1467. [110] M.N. Nikiforova, A. Khalid, K.E. Fasanella, K.M. McGrath, R.E. Brand,
[77] R. Khosravi-Far, P.A. Solski, G.J. Clark, M.S. Kinch, C.J. Der, Activation of Rac1, J.S. Chennat, et al., Integration of KRAS testing in the diagnosis of pancreatic
RhoA, and mitogen-activated protein kinases is required for Ras transformation, cystic lesions: a clinical experience of 618 pancreatic cysts, Mod. Pathol. 26 (2013)
Mol. Cell. Biol. 15 (1995) 6443–6453. 1478–1487.
[78] P. Rodriguez-Viciana, P.H. Warne, R. Dhand, B. Vanhaesebroeck, I. Gout, M.J. Fry, [111] D. Sefrioui, N. Sarafan-Vasseur, L. Beaussire, M. Baretti, A. Gangloff, F. Blanchard,
et al., Phosphatidylinositol-3-OH kinase as a direct target of Ras, Nature 370 F. Clatot, J.C. Sabourin, R. Sesboüé, T. Frebourg, P. Michel, F. Di Fiore, Clinical
(1994) 527–532. value of chip-based digital-PCR platform for the detection of circulating DNA in
[79] B. Vanhaesebroeck, S.J. Leevers, G. Panayotou, M.D. Waterfield, Phosphoinositide metastatic colorectal cancer, Dig. Liver Dis. 47 (2015) 884–890.
3-kinases: a conserved family of signal transducers, Trends Biochem. Sci. 22 [112] T.R. Levin, D.A. Corley, C.D. Jensen, A.R. Marks, W.K. Zhao, A.M. Zebrowski,
(1997) 267–272. et al., Genetic biomarker prevalence is similar in fecal immunochemical test po-
[80] L. Zhao, P.K. Vogt, Class I PI3K in oncogenic cellular transformation, Oncogene 27 sitive and negative colorectal cancer tissue, Dig. Dis. Sci. 62 (2017) 678–688.
(2008) 5486–5496. [113] R. Critelli, F. Fasanelli, M. Oderda, S. Polidoro, M.B. Assumma, C. Viberti, et al.,
[81] A.K. Murugan, N.T. Hong, Y. Fukui, A.K. Munirajan, N. Tsuchida, Oncogenic Detection of multiple mutations in urinary exfoliated cells from male bladder
mutations of the PIK3CA gene in head and neck squamous cell carcinomas, Int. J. cancerpatients at diagnosis and during follow-up, Oncotarget 7 (2016)
Oncol. 32 (2008) 101–111. 67435–67448.
[82] A. Sjölander, K. Yamamoto, B.E. Huber, E.G. Lapetina, Association of p21ras with [114] M. Xing, Clinical utility of RAS mutations in thyroid cancer: a blurred picture now
phosphatidylinositol 3-kinase, Proc. Natl. Acad. Sci. U. S. A. 88 (1991) 7908–7912. emerging clearer, BMC Med. 14 (2016) 12.
[83] S. Gupta, A.R. Ramjaun, P. Haiko, Y. Wang, P.H. Warne, B. Nicke, et al., Binding of [115] Y.E. Nikiforov, S.E. Carty, S.I. Chiosea, C. Coyne, U. Duvvuri, R.L. Ferris, et al.,
ras to phosphoinositide 3-kinase p110alpha is required for ras-driven tumorigen- Highly accurate diagnosis of cancer in thyroid nodules with follicular neoplasm/
esis in mice, Cell 129 (2007) 957–968. suspicious for a follicular neoplasm cytology by ThyroSeq v2 next-generation se-
[84] J. Yan, S. Roy, A. Apolloni, A. Lane, J.F. Hancock, Ras isoforms vary in their ability quencing assay, Cancer 120 (2014) 3627–3634.
to activate Raf-1 and phosphoinositide 3-kinase, J. Biol. Chem. 273 (1998) [116] M. Medici, N. Kwong, T.E. Angell, E. Marqusee, M.I. Kim, M.C. Frates, et al., The
24052–24056. variable phenotype and low-risk nature of RAS-positive thyroid nodules, BMC
[85] M. Cully, J. Downward, SnapShot: ras signaling, Cell 133 (2008) 1292. Med. 13 (2015) 184.
[86] M.P. Scheid, J.R. Woodgett, Phosphatidylinositol 3′ kinase signaling in mammary [117] Y. Loriot, P. Mordant, E. Deutsch, K.A. Olaussen, J.C. Soria, Are RAS mutations
tumorigenesis, J. Mammary Gland Biol. Neoplasia 6 (2001) 83–99. predictive markers of resistance to standard chemotherapy? Nat. Rev. Clin. Oncol.
[87] D.D. Sarbassov, D.A. Guertin, S.M. Ali, D.M. Sabatini, Phosphorylation and reg- 6 (2009) 528–534.
ulation of Akt/PKB by the rictor-mTOR complex, Science 307 (2005) 1098–1101. [118] N. Normanno, S. Tejpar, F. Morgillo, A. De Luca, E. Van Cutsem, F. Ciardiello,
[88] H.C. Welch, W.J. Coadwell, L.R. Stephens, P.T. Hawkins, Phosphoinositide 3-ki- Implications for KRAS status and EGFR-targeted therapies in metastatic CRC, Nat.
nase-dependent activation of Rac, FEBS Lett. 546 (2003) 93–97. Rev. Clin. Oncol. 6 (2009) 519–527.
[89] R. Fritsch, I. de Krijger, K. Fritsch, R. George, B. Reason, M.S. Kumar, et al., RAS [119] J.L. Marks, S. Broderick, Q. Zhou, D. Chitale, A.R. Li, M.F. Zakowski, et al.,
and RHO families of GTPases directly regulate distinct phosphoinositide 3-kinase Prognostic implications of EGFR and KRAS mutations in resected lung adeno-
isoforms, Cell 153 (2013) 1050–1063. carcinoma, J. Thorac. Oncol. 3 (2008) 111–116.
[90] A.K. Murugan, A.K. Munirajan, A.S. Alzahrani, MicroRNAs: modulators of the Ras [120] W. Pao, T.Y. Wang, G.J. Riely, V.A. Miller, Q. Pan, M. Ladanyi, et al., KRAS mu-
oncogenes in oral cancer, J. Cell. Physiol. 231 (2016) 1424–1431. tations and primary resistance of lung adenocarcinomas to gefitinib or erlotinib,
[91] S. Gysin, M. Salt, A. Young, F. McCormick, Therapeutic strategies for targeting ras PLoS Med. 2 (2005) e17.
proteins, Genes Cancer 2 (2011) 359–372. [121] D.A. Eberhard, B.E. Johnson, L.C. Amler, A.D. Goddard, S.L. Heldens, R.S. Herbst,
[92] E. Castellano, J. Downward, RAS interaction with PI3K more than just another et al., Mutations in the epidermal growth factor receptor and in KRAS are pre-
effector pathway, Genes Cancer 2 (2011) 261–274. dictive and prognostic indicators in patients with non-small-cell lung cancer
[93] A. Sartore-Bianchi, M. Martini, F. Molinari, S. Veronese, M. Nichelatti, S. Artale, treated with chemotherapy alone and in combination with erlotinib, J. Clin.
et al., PIK3CA mutations in colorectal cancer are associated with clinical resistance Oncol. 23 (2005) 5900–5909.
to EGFR-targeted monoclonal antibodies, Cancer Res. 69 (2009) 1851–1857. [122] E. Massarelli, M. Varella-Garcia, X. Tang, A.C. Xavier, N.C. Ozburn, D.D. Liu, et al.,
[94] J.M. Ostrem, U. Peters, M.L. Sos, J.A. Wells, K.M. Shokat, K-Ras(G12C) inhibitors KRAS mutation is an important predictor of resistance to therapy with epidermal
allosterically control GTP affinity and effector interactions, Nature 503 (2013) growth factor receptor tyrosine kinase inhibitors in non-small-cell lung cancer,
548–551. Clin. Cancer Res. 13 (2007) 2890–2896.
[95] S.M. Lim, K.D. Westover, S.B. Ficarro, R.A. Harrison, H.G. Choi, M.E. Pacold, et al., [123] R.G. Amado, M. Wolf, M. Peeters, E. Van Cutsem, S. Siena, D.J. Freeman, et al.,
Therapeutic targeting of oncogenic K-Ras by a covalent catalytic site inhibitor, Wild-type KRAS is required for panitumumab efficacy in patients with metastatic
Angew. Chem. Int. Ed. Engl. 53 (2014) 199–204. colorectal cancer, J. Clin. Oncol. 26 (2008) 1626–1634.
[96] P. Lito, M. Solomon, L.S. Li, R. Hansen, N. Rosen, Allele-specific inhibitors in- [124] C.J. Allegra, J.M. Jessup, M.R. Somerfield, S.R. Hamilton, E.H. Hammond,
activate mutant KRAS G12C by a trapping mechanism, Science 351 (2016) D.F. Hayes, et al., American society of clinical oncology provisional clinical opi-
604–608. nion: testing for KRAS gene mutations in patients with metastatic colorectal car-
[97] M.H. Yang, S. Nickerson, E.T. Kim, C. Liot, G. Laurent, R. Spang, et al., Regulation cinoma to predict response to anti-epidermal growth factor receptor monoclonal
of RAS oncogenicity by acetylation, Proc. Natl. Acad. Sci. U. S. A. 109 (2012) antibody therapy, J. Clin. Oncol. 27 (2009) 2091–2096.
10843–10848. [125] W. De Roock, B. Claes, D. Bernasconi, J. De Schutter, B. Biesmans, G. Fountzilas,
[98] A. Patgiri, K.K. Yadav, P.S. Arora, D. Bar-Sagi, An orthosteric inhibitor of the Ras- et al., Effects of KRAS, BRAF, NRAS, and PIK3CA mutations on the efficacy of
Sos interaction, Nat. Chem. Biol. 7 (2011) 585–587. cetuximab plus chemotherapy in chemotherapy-refractory metastatic colorectal
[99] M.J. Smith, M. Ikura, Integrated RAS signaling defined by parallel NMR detection cancer: a retrospective consortium analysis, Lancet Oncol. 11 (2010) 753–762.
of effectors and regulators, Nat. Chem. Biol. 10 (2014) 223–230. [126] D. Cunningham, Y. Humblet, S. Siena, D. Khayat, H. Bleiberg, A. Santoro, et al.,
[100] V. Asati, D.K. Mahapatra, S.K. Bharti, K-Ras and its inhibitors towards persona- Cetuximab monotherapy and cetuximab plus irinotecan in irinotecan-refractory
lized cancer treatment: pharmacological and structural perspectives, Eur. J. Med. metastatic colorectal cancer, New England J. Med. Surg. Collat. Branches Sci. 351
Chem. 125 (2017) 299–314. (2004) 337–345.
[101] L. Seguin, M.F. Camargo, H.I. Wettersten, S. Kato, J.S. Desgrosellier, T. von [127] J. Hecht, E. Mitchell, J. Baranda, Panitumumab antitumor activity in patients (pts)
Schalscha, et al., Galectin-3, a druggable vulnerability for KRAS-addicted cancers, with metastatic colorectal cancer (mCRC) expressing low (1–9%) or negative
Cancer Discov. 7 (2017) 1464–1479. (< 1%) levels of epidermal growth receptor (EGFr), J. Clin. Oncol. 24 (2006)
[102] M.E. Welsch, A. Kaplan, J.M. Chambers, M.E. Stokes, P.H. Bos, A. Zask, et al., 3506.

12
A.K. Murugan, et al. Seminars in Cancer Biology xxx (xxxx) xxx–xxx

[128] H.H. Yoon, D. Tougeron, Q. Shi, S.R. Alberts, M.R. Mahoney, G.D. Nelson, et al., initiate leukemia in vivo, Blood 121 (2013) 4884–4893.
KRAS codon 12 and 13 mutations in relation to disease-free survival in [138] F.D. Tsai, M.S. Lopes, M. Zhou, H. Court, O. Ponce, J.J. Fiordalisi, et al., K-Ras4A
BRAF–wild-type stage III colon cancers from an adjuvant chemotherapy trial splice variant is widely expressed in cancer and uses a hybrid membrane-targeting
(N0147 alliance), Clin. Cancer Res. 20 (2014) 3033–3043. motif, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) 779–784.
[129] W. Li, T. Qiu, W. Zhi, S. Shi, S. Zou, Y. Ling, et al., Colorectal carcinomas with [139] T. Ushijima, T. Yoshino, The moment that KRAS mutation started to evolve into
KRAS codon 12 mutation are associated with more advanced tumor stages, BMC precision medicine in metastatic colorectal cancer, Cancer Res. 76 (2016)
Cancer 15 (2015) 1340. 6443–6444.
[130] S. Rachagani, S. Senapati, S. Chakraborty, M.P. Ponnusamy, S. Kumar, L.M. Smith, [140] N.P. Valsangkar, T. Ingkakul, C. Correa-Gallego, M. Mino-Kenudson, R. Masia,
et al., Activated KrasG12D is associated with invasion and metastasis of pancreatic K.D. Lillemoe, et al., Survival in ampullary cancer: potential role of different KRAS
cancer cells through inhibition of E-cadherin, Br. J. Cancer 53 (1985) 1038–1048. mutations, Surgery 157 (2015) 260–268.
[131] J. Downward, RAS’s cloak of invincibility slips at last, Çancer Cell 25 (2014) 5–6. [141] T.G. Bivona, Dampening oncogenic RAS signaling, Science 363 (2019)
[132] E. Castellano, C. Sheridan, M.Z. Thin, E. Nye, B. Spencer-Dene, M.K. Diefenbacher, 1280–1281.
et al., Requirement for interaction of PI3-kinase p110 with RAS in lung tumor [142] M.R. Janes, J. Zhang, L.S. Li, R. Hansen, U. Peters, X. Guo, et al., Targeting KRAS
maintenance, Cancer Cell 24 (2013) 617–630. mutant cancers with a covalent G12C-specific inhibitor, Cell 172 (2018) 578-
[133] M.M. Murillo, S. Zelenay, E. Nye, E. Castellano, F. Lassailly, G. Stamp, et al., RAS 589.e17.
interaction with PI3K p110α is required for tumor-induced angiogenesis, J. Clin. [143] S. Mueller, T. Engleitner, R. Maresch, M. Zukowska, S. Lange, T. Kaltenbacher,
Invest. 124 (2014) 3601–3611. et al., Evolutionary routes and KRAS dosage define pancreatic cancer phenotypes,
[134] Y. Samuels, L.A. Diaz Jr, O. Schmidt-Kittler, J.M. Cummins, L. Delong, I. Cheong, Nature 554 (2018) 62–68.
et al., Mutant PIK3CA promotes cell growth and invasion of human cancer cells, [144] K. Shimizu, M. Goldfarb, M. Perucho, M. Wigler, Isolation and preliminary char-
Cancer Cell 7 (2005) 561–573. acterization of the transforming gene of a human neuroblastoma cell line, Proc.
[135] R. Yaeger, E. Cowell, J.F. Chou, A.N. Gewirtz, L. Borsu, E. Vakiani, et al., RAS Natl. Acad. Sci. U. S. A. 80 (1983) 383–387.
mutations affect pattern of metastatic spread and increase propensity for brain [145] J.L. Bos, E.R. Fearon, S.R. Hamilton, M. Verlaan-de Vries, J.H. van Boom, A.J. van
metastasis in colorectal cancer, Cancer 121 (2015) 1195–2003. der Eb, B. Vogelstein, Prevalence of ras gene mutations in human colorectal
[136] N.T. Ihle, L.A. Byers, E.S. Kim, P. Saintigny, P.J. Lee, G.R. Blumenschein, et al., cancers, Nature 327 (1987) 293–297.
Effect of KRAS oncogene substitutions on protein behavior: implications for sig- [146] J.R. Kovalski, A. Bhaduri, A.M. Zehnder, P.H. Neela, Y. Che, G.G. Wozniak,
naling and clinical outcome, J. Natl. Cancer Inst. 104 (2012) 228–239. P.A. Khavari, The functional proximal proteome of oncogenic ras includes
[137] A. Shieh, A.F. Ward, K.L. Donlan, E.R. Harding-Theobald, J. Xu, C.G. Mullighan, mTORC2, Mol. Cell 73 (2019) 830–844.
et al., Defective K-Ras oncoproteins overcome impaired effector activation to

13

You might also like