SlideShare uma empresa Scribd logo
1 de 43
1
Bioscience & Chemistry Programme
Professional & Scientific Practice 3
Evaluating the pathogenesis of Lung Adenocarcinoma in relation to
NICE treatment guidelines and futuristic treatment developments
By Adam Boulger
B7040016
Submitted Date: 21/02/2020
2
Table of Contents
Abstract ………………………………………………………………………………..3
1. Introduction……………………………………………………………………… 3-5
2. Genetic analysis to identify underlying pathological mechanisms…………. 5-9
2.1 MAPK & PI3K/mTOR pathway dysregulation drives pathogenesis…… ..10-15
2.2 Cell Cycle propelled by MAPK/PI3K effectors……………………………...16-17
3 Current treatment approach to NSCLC Adenocarcinoma…………………...18-19
3.1 Futuristic treatment approach: Personalised Pharmacogenomics……… 19-23
4.0 Conclusion…………………………………………………………………….. 23-24
Bibliography……………………………………………………………………….. 25-43
3
Evaluating the pathogenesis of Lung Adenocarcinoma in relation to
NICE treatment guidelines and futuristic treatment developments.
Abstract
Non-small cell lung cancer (NSCLC) is the deadliest form of cancer, accentuating the
clinical demand for more effective treatments in addition to the introduction of early
detection programs. Genomic advancements are driving the understanding around
the underlying pathogenesis by identifying recurrent MAPK/PI3K and cell cycle
pathway dysregulation. Current targeted treatment approach against epidermal
growth factor receptor (EGFR) and programmed death-ligand 1 (PD-L1) enable
median survival of 216 days against non-targeted 203 days. An underappreciation
towards copy number alterations could be a contributing factor and in recent years,
advancements are focusing upon human epidermal growth factor receptor-2 (Her-2),
Kirsten rat sarcoma (K-Ras) and cyclin-dependent kinases 4/6 (CDK-4/6) inhibitors
4
present the potential to increase disease survival rates. Although, rapid and
considerable progress are required due to the poor survival and a lack of specific
patient cohorts, hindering developments through a bottleneck in clinical trials.
1. Introduction to Lung Adenocarcinoma
Lung cancer is the second most prevalent type of cancer for both males and females across
the UK (UK Office for National Statistics, 2018). UK government figures from 2017 outlined
38,888 cancer registrations, consisting of 12.7% of the total cancer burden (UK Office for
National Statistics, 2019) forming the deadliest form of cancer and attributed as the fifth
leading causation of death with around 30570 deaths during 2017 (UK Office for National
Statistics, 2017). This figure, which is greater than breast, colon and pancreatic cancers
combined (Zappa, & Mousa, 2016), exemplifies the importance of reviewing and researching
lung cancer to improve the approach of diagnosis and treatment of lung cancers. Lung cancer
is divided into 13% patients with small-cell and 87% NSCLC (National Health Service, 2020).
Although, this is potentially simplified as a major flaw was identified by NHS statistics stating
only 72% of lung cancer patients are confirmed by phenotypic or molecular genotyping
(NICE, 2017). It could be suggested that this is in relation to around 70% of individuals being
diagnosed at an advanced disease state, commonly stated as terminal (Molina, Yang,
Cassivi, Schild, & Adjei, 2008). However, research concurs that the major histological form
of NSCLC is adenocarcinoma and is accepted as the leading cause of cancer deaths
worldwide (Collison et al, 2014), with Reis et al identifying the relative frequency being 90.9%
of cases (Reis et al, 2020). Although, comparative analysis with a 2019 publication
commissioned and implemented by NHS identified adenocarcinoma at 60% (Crosbie et al,
2019). This agreement outlined the most prevalent sub-type, whilst highlighting clear issues
with data standards and a lack of consistency. Until recently, it was thought major sub-types
could be characterised by their antithetical anatomical tumour locations, with
5
adenocarcinoma being peripheral (Kadara, Scheet, Ignacio, & Spira, 2016), but this is no
longer assumed following Moon et al’s publication highlighting 13.3% of cases being central
bound (Moon, Lee, Sung, & Park, 2016). This could suggest reasoning behind the
discrepancy identified in the data, therefore, these statistics may not be comparable or
accurate. A current focus is upon early diagnosis, with a Manchester research screening pilot
across 1384 high risk individuals produced diagnostic results showing 80.4% patients
diagnosed at stages I/II compared to the standard of 31% and advanced stages III/IV at
19.6% compared to standard of 69%, with the biggest decrease being for stage IV from 48%
to 11% (Crosbie et al, 2019). Furthermore, another screening trial reduced mortality by 26%
in men and up to 61% in females (Yousaf-Khan et al, 2017). This directly correlates to findings
by McPhail et al, stating that an earlier diagnosis directly correlates to an increased rate of
survival (McPhail, Johnson, Greenberg, Peake, & Rous, 2015). This review will evaluate the
current understanding of pathogenesis in relation to current and futuristic treatment
approaches.
2. Genetic analysis to identify underlying pathological mechanisms
Across adenocarcinoma, difficulties identifying the cell of origin have historically hampered
the understanding of initiation and progression pathogenesis studies (Devarakonda,
Morgensztern, & Govindan, 2015), with alveolar type II cells now accepted as the origin of
adenocarcinoma (Kim et al, 2005., Lin et al, 2012). Critically, the issues with cell of origin still
exist with central adenocarcinoma. To elucidate the pathogenesis of adenocarcinoma, The
Cancer Genome Atlas Research Network (TCGARN) published a large-scale genome project
(Collison et al, 2014) to identify prevalent recurrent genetic alterations to be identified in those
with adenocarcinoma. This data can be mapped in accordance to affected pathways to
suggest the basis of underlying pathological mechanisms. Critically, a major issue with
6
TCGARN’s publication was the focus upon point mutations rather than copy number
alterations, although the copy number alteration data was provided.
Figure 1: 2-D Bar Chart presenting percentage (%) Lung adenocarcinoma patients with somatic point mutation
(missense, nonsense, in-frame deletion, frameshift) against correlating gene. Selected all >10%. Created using
The Cancer Genome Atlas Research Network’s publication of data (Collison etal,2014).Loss of function (LOF)
mutation to TP53, KEAP1, STK11 and NF1. Gain of function (GOF) mutation to KRAS, EGFR, BRAF.
0 5 10 15 20 25 30 35 40 45 50
BRAF
NF1
EGFR
SKT11
KEAP1
KRAS
TP53
% Patients
Geneaffected
RecurrentLung adenocarcinomapatient point mutations
7
Figure 2: 2-D Bar Chart presenting % patients presenting with low-medium copy number amplification to a
gene. Selected all >10%. Constructed using raw genetic data from supplementary data from The Cancer
Genome Atlas Research Network 2014 publication (Collison etal,2014).
0 10 20 30 40 50 60 70
RBM10
HRAS
AKT1
CCNE1
U2AF1
MDM2
CDK4
NRAS
PI3KCA
ARID1A
KRAS
TERC
CCND1
NKX2-1
MET
NF1
BRAF
ERBB2
EGFR
TERT
MYC
RIT1
% Patients
GeneAffected Recurrentlow-mediumcopy number amplifications inLung
adenocarcinomapatients
8
0 2 4 6 8 10 12 14 16 18 20
TERT
RIT1
NKX2-1
MYC
MDM2
EGFR
CDK4
KRAS
% Patients
GeneAffected Recurrenthighcopy number amplificationinlung
adenocarcinomapatients
Figure 3: 2-D Bar Chart presenting % patients presenting with a specific high copy number amplification to a
gene. Selected all >5%. Constructed using raw genetic data from supplementary data from The Cancer
Genome Atlas Research Network 2014 publication (Collison etal,2014).
9
The underlying disease mechanisms are yet to be understood (Chen et al, 2018)
with Zhang et al highlighting this difficulty due to the complex heterogeneity (Zhang,
Chang, & Yang, 2019). However, research has a collective acceptance that the
MAPK and PI3K pathways are essential drivers (Wu et al 2019., Pradhan et al
2019). This in relation to 75% of patients presenting with direct oncogenic alterations
driving Ras activity (Inamura, 2017) and 62% of patients presenting direct mutations
to KRAS and EGFR (Kadara et al, 2016., Yap et al, 2014) with an average of 8.9
mutations per million bases (Collison et al, 2014).
0 10 20 30 40 50 60
CDKN2A
SETD2
RB1
KEAP1
SMARCA44
STK11
MGA
TP53
% patients
GeneAffected
Recurrent gene deletions in lung adenocarcinoma
patients
Figure 4 : 2-D Bar Chart presenting % patients presenting with genetic absence of a gene. Heterozygous loss is
coded as blue and homozygous loss (2L) is orange. Heterozygous loss selected top 8 most common, majority of
Homozygous loss only present as affecting CDKN2A, second most common was gene MGA with 3.04% patients
affected. Constructed using raw genetic data from supplementary data from The Cancer Genome Atlas
Research Network 2014 publication (Collison etal,2014).
CDKN2A
10
2.1 MAPK & PI3K/mTOR pathway dysregulation drives pathogenesis
Figure 5: Created using data outlined in Figures 1, 2, 3 and 4 to review the major pathway modifications to
signalling pathways involved in propagation and pathogenesis by identifying recurrent pathogenic drivers to
suggest potential treatment approaches to lung adenocarcinoma. Diagram created to review and collate the
plethora of recent advancements and publications in the field of adenocarcinoma utilising information from
(Collison etal,2014.,Singh et al,2013., Reis et al,2020., Cheung, & Nguyen, 2015., Cheng, Alexander, &
MacLennan, 2012., Greulich,2010., Berger, Imielinski,& Duke, 2014., Kadara et al,2016., Korneeva, Song,
Gram, Edens, & Rhoads, 2015.,Siddiqui,& Sonenberg, 2015., Jahangiri,& Weiss,2013.,Gul,Leyland-Jones,
Dey, & De, 2018., Hemmings, & Restuccia,2012.,Ozenne, Eymin, Brambilla,& Gazzeri, 2010).Key: green
11
arrows represent activation, red arrows represent inhibition, orange boxes highlight oncogenes and grey boxes
highlight tumour suppressor genes. Abbreviations: GOF, gain of function, LOF, loss of function, LMA, low
medium amplification, HA, high amplification, -1hl heterozygous loss, -2hl, homozygous loss.
Chen et al highlighted that although the underlying disease mechanisms have yet to
be elucidated, their research analysing KEGG pathway enrichment highlights the
importance of the cell cycle and cancer pathways (Chen et al, 2018). Recent
publications by Pradhan et al and Wu et al highlight the importance of MAPK and PI3K
cancer pathways as essential for pathogenesis by driving the cell cycle (Wu et al
2019., Pradhan et al 2019).
Across the genomic landscape of adenocarcinoma, Figure 5 presents the recurrent
receptor alterations to MET, ERBB1and ERBB2 which directly induce MAPK and PI3K
signalling (Collison et al, 2014) leading to proliferation, evasion of apoptosis and
angiogenesis (Sun et al, 2015., Liu, Jin, Wang, & Wang, 2017). The most prevalent is
ERBB1, presenting two recurrent gain of function (GOF) aberrations which consist for
90% total mutation burden with exon 21 p.L858R (Wan, Wright, & Coveney, 2012) and
micro-deletion p.del19 which directly increase pro-oncogenic downstream signalling
(Imielinks et al, 2012., Jakobsen, Santoni-Rugiu, Grauslund, Melchior, & Sørensen,
2018) in addition to 47% of patients presenting copy number amplifications (Collison
et al, 2014). This is supported by Kadara et al and Reungwetwattana et al indicating
the involvement of ERBB1 and Ras family alterations as the most prominent driver
mutations (Kadara et al, 2016., Reungwetwattana, Weroha, & Molina, 2012), with Lee
et Bae highlighting K-ras as an important event in the early initiation of
adenocarcinoma (Lee, & Bae, 2016). As illustrated in Figure 5, the intercalation
between MAPK and PI3K can be driven by K-ras, presenting 33% and 33.5% of
patients with GOF and amplifications respectively (Collison et al, 2014). Jordan et al
identified the most prevalent somatic alteration as p.G12C across both primary and
12
metastatic patients (Jordan et al, 2017), which links to alternative research highlighting
this mutation leads to greater oncogenic activity by higher extracellular signal-related
kinase 1/2 (ERK) downstream activatory phosphorylation in comparison to p.G12D
(Yang, Liang, Schmid, & Peng, 2019., Li et al, 2018) which drives downstream MAPK
activity. This suggests that analysing gene heterogeneity could lead to an improved
pharmacogenomic approach.
The Kegg publication identified the key effectors of the MAPK pathway as c-Jun, c-
Fos, Ets1, MSK1 and c-MYC (Kyoto University Bioinformatics Centre, 2019). ERK’s
phosphorylate and activate ETS-Like-1 protein (Elk-1) and Ets-1 transcription factors
which rapidly upregulate the Jun and Fos proto-oncogene families, with Kegg
highlighting the up-regulation of c-Fos and c-Jun proteins as key which form activator
protein-1 (AP-1) heterodimers (Kyoto University Bioinformatics Centre, 2019.,
Atsaves, Leventaki, Rassidakis, & Claret, 2019) . A landmark paper identified the c-
Fos:c-Jun heterodimer exhibits higher transactivation in comparison to alternatives
such as JunB or JunD (Chiu, Angel, & Karin, 1989). This activity is activated upon the
transcriptional activation domain of c-Jun at sites p.Ser63 and p.Ser73 being
phosphorylated by c-Jun N-terminal terminal kinases (JNK) (Zhao, Wang, & Tony,
2015), with Tanos et al identifying c-Fos is activated by P38 MAPK phosphorylation
(Tanos et al, 2005). A key finding, highlighted by Alonso et al, is c-Jun promoter and
AP-1 exhibiting a high affinity, therefore, it could be suggested that a positive feedback
loop exists with this perpetual and self-activatory action exacerbating pathogenesis by
increasing AP-1 levels (Alonso et al, 2018), with Figure 5 outlining AP-1 upregulating
cyclins, CDK’s and activating E2F transcription factors.
13
The identification of an underlying driver could lead to rapid progression in the
treatment of NSCLC adenocarcinoma. Elangovan et al’s breakthrough research
discovered Fos family, Fos-like antigen-1 (FOSL-1) as a major effector of RAS-MAPK
signalling, presenting FOSL-1 expression as a critical determinant of the
tumourigenesis of NSCLC adenocarcinoma, highlighting that emerging data is
indicating that FOSL-1 expression and poor survival are directly correlated (Elangovan
et al, 2018). These findings were developed upon Vallejo et al’s publication, with data
identifying FOSL-1 up regulation in mutant K-ras mutant (p.G12D) cell lines by multiple
downstream kinases via an autonomous mechanism (Vallejo et al, 2017). With the key
transcriptional regulators being ATF-2, MYC and AP-1 (Lopez-Bergami, Lau, & Ronai,
2010), with a finding identifying a key role of MSK1, by phosphorylating histone H3S10
to induce the transcriptional elongation of FOSL-1, C-Jun and C-Fos (Zippo et al,
2009). This links directly back to Kegg publication highlighting c-Jun, c-Fos, Ets1,
MSK1 and c-MYC as the major factors (Kyoto University Bioinformatics Centre, 2019)
whilst portraying the symbiotic self-activatory activities of the constituents.
Furthermore, as indicated by Figure 5, Warne et al identified the MAPK/PI3K
intercalation by Ras activation of PI3K (Rodriguez-Viciana, Warne, Vanhaesebroeck,
Waterfield, & Downward, 1996), with Castellano et Downard’s publication presenting
the PI3K pathway as essential in RAS mutant in-vivo tumourigenesis studies with
PI3KCA’s p.Lys227 being essential in this intercalation activation (Castellano, &
Downward, 2011), with 24% of patients presenting amplifications (Collison et al,
2014). However, Nussinov et al recently highlighted the difficulty in identifying the
mechanism by which this occurs (Nussinov, Tsai, & Jang, 2019). An investigation into
whether Ras heterogeneity (such as K-ras p.G12C>p.G12D discussed above) can
14
induce variable levels of PI3K activity could lead to breakthrough findings. This
essential Ras:PI3KCA interaction was disrupted by Castellano et al in Ras driven
adenocarcinoma and this led to regression by an autonomous mechanism (Castellano
et al, 2013). This was also found by Murillo et al in mutant ERBB1 cells, where
disrupting Ras:PI3KCA inhibited tumourigenesis and also induced regression in
established adenocarcinoma (Murillo et al, 2018). Furthermore, Veen et al recently
published findings with mutant BRAF (p.V600E) cells unable to initiate
adenocarcinoma tumourigenesis unless co-expressed with PI3KCA pathway mutant
(p.H1047R), compared to mutant K-ras (p.G12D) being able to solely induce
tumourigenesis, which is directly associated to RAS:PI3K cross-interaction (Veen et
al, 2019). Upon this, it could be suggested that this interaction is a major underlying
factor in the pathogenesis, even with a critical flaw in Veen et al’s methodology using
PI3KCA mutant not present in TCGARN’s data (Collison et al, 2014). Wang et al
highlighted across their study into PI3KCA mutant patients that 86% had coexisting
mutations in ERBB1 or K-Ras (Wang et al, 2014). However, Tang et al presented
findings which suggested all patients present with co-existing mutations (Tang, Zhang,
& Lu, 2018). Although, it could be suggested that a key flaw across papers is the
incomparable selection of individuals, therefore, this could lead to inaccurate
comparative analysis between papers. Meng et al presents an example of this by
focusing only upon Chinese patients (Meng et al, 2019).
Furthermore, TCGARN highlight unknown mechanisms of MAPK/PI3K activation exist
(Collison et al, 2014), which could link to the under appreciation of copy number
alterations mostly under appreciating the alterations to HRAS, NRAS and RIT1 genes.
TCGARN data presents 1% of patients present with somatic mutations to these genes
15
(Collison et al, 2014). However, including analysis of copy number alterations
identifies high levels of amplifications, with 16%, 23% and 74% patients presenting
amplifications respectively. Critically, Figure 5 shows patients can present with loss of
function (11%) or amplifications (37%) to NF1, which Yap et al outline is a tumour
suppressor by suppressing Ras activity (Yap et al, 2014). This would indicate NF1
reduces MAPK/PI3K activity, although, questioning the understanding of the role of
NF1 could lead to novel findings around the functionality as recurrent amplifications to
a tumour suppressor with current understanding would not assume to induce
adenocarcinoma. This is yet to be analysed in relation to adenocarcinoma, but breast
cancer patients present 17% NF1 amplification, with Philpott et al suggesting it is
important for the pathogenesis (Philpott, Tovell, Frayling, Cooper, & Upadhyaya,
2017). It could be suggested that NF1 presents oncogenic properties with certain
genomic profiles, potentially an investigation into whether NF1 repressing RAS: MAPK
leads to an increase of RAS:PI3K interaction.
16
2.2 Cell Cycle propelled by MAPK/PI3Keffectors
Figure 6: Constructed upon Figures 1, 2, 3, 4 and 5 to review the major pathway modifications to the cell cycle
pathway. Key: green arrows represent activation, red arrows represent inhibition, orange boxes highlight
oncogenes, pink highlight transcription factors and grey boxes highlight tumour suppressor genes. References;
(Collison etal,2014., Singh et al,2013.,Reis et al,2020., Cheung, & Nguyen, 2015., Cheng et al,2012.,
Greulich,2010., Berger et al,2014., Kadara et al,2016).
The role of MAPK/PI3K outlined in Figure 5, in addition to Figure 6, highlights the recurrent
cell cycle alterations, which is a clear presentation of the correlation and symbiotic nature of
17
pathway alterations leading to R-point passage for S-phase which is understood as the ‘point
of no return’ for cellular proliferation (Matson, & Cook, 2017). As presented by Figure 6, PI3K
effector ATK1 inhibits P53 with ATK1 and MDM2 both present with recurrent amplifications,
highlighting the essential inhibition of P53 (Collison et al, 2014). This is also evident by 61%
patients presenting a form of CDK2NA loss and this prevents the inhibition of MDM2.
(Collison et al, 2014). Furthermore, this is also evident by 46% and 55% of patients
presenting with TP53 LOF or heterozygous deletion respectively. This finding is key as in
cases of heterozygosity, mutant P53 exhibits the ability to antagonise wild-type P53 in a
dominant-negative manner by inhibiting tetramer formation (Rivlin, Brosh, Oren, & Rotter,
2011). Alexandrova et al suggests this could underestimate true P53 loss of function
(Alexandrova, Mirza, & Xu, 2017). Although, Lee et Bae stated that it is currently unclear
whether TP53 inactivation is the progression or initiation of pathogenesis (Lee, & Bae, 2016).
Investigations by Junttila et al & Feldser et al induced adenocarcinoma by mutating Ras/TP53
and reversing this led to regression (Junttila et al, 2010., Feldser et al, 2010). Furthermore,
Junttila et al identified Ras mutants are a potent trigger of P53 and the oncogenic driving
action of Ras mutants is selective for the acquisition of mutant TP53 genes (Junttila et al,
2010), whilst Feldser et al highlighted MAPK signalling as a critical determinant of
adenocarcinoma (Feldser et al, 2010). This is the basis which enables the extreme
accumulation of mutations.
18
3.0 Current Treatment Approach to NSCLC Adenocarcinoma
Figure 7: Adapted from NICE guidelines illustrating current clinical algorithm (National Institutefor Health and
Care Excellence, 2019).Altered to remove ALK analysis following TCGARN data (34) which presented <1%
adenocarcinoma patients with ALK mutant. This is a flaw in the current practice due to grouping into
‘squamous’ and ‘non-squamous’ forms.
19
Khakwani analysed UK NSCLC survival trends identifying a slight survival
improvement due to increased surgery rate, even with an increasing proportion of
NSCLC adenocarcinoma (Khakwani et al, 2013). The current treatment algorithm is
fundamentally flawed with the NHS 2019 Lung Cancer audit stating 67.3% and 37%
of patients achieved three-month survival and one-year survival respectively with only
18.4% of patients undergoing surgical cancer removal and 40% of stage IIIB patients
not receiving treatment (Royal College of Physicians, 2018). Furthermore, a 2020
audit on molecular testing across 1157 advanced ‘non-squamous’ patients 83% of
patients analysed following guidelines for EGFR, PD-L1 and ALK alterations (Royal
College of Physicians, 2020). Although, clinical data identifies 3.5% of non-squamous
patients present with ALK, which does not appear in adenocarcinoma patients
(Collison et al, 2014), questioning the rationale behind this. Across the 16.5% EGFR+
patients, 75% received treatment. Median survival was 216 days (Royal College of
Physicians, 2020). This is a slight improvement from the median survival of 203 days
(2011) (NICE, 2011). It could, therefore, be suggested novel therapeutics are an
essential clinical demand.
3.1 Futuristic treatmentapproaches: Personalised Pharmacogenomics
The current outdated and generalised histopathological and anatomical classification
into squamous or non-squamous should be updated in line with advancements of
genomic understanding and to introduce a personalised targeted approach for
adenocarcinoma patients. A logical solution suggested by TCGARN’s publication
was to characterise by point mutations (Collison et al, 2014), with additions of NF1,
STK11 and B-Raf (Collison et al, 2014) to Wilkerson et al’s TP53, K-Ras and ERBB1
suggestions (Wilkerson et al, 2012). Although, it could be suggested this is an
20
incomplete solution with TCGARN stating copy number alterations, presented by
Figures 5 and 6, were not considered as pathogenesis driver alterations in their
publication (Collison et al, 2014). Potential futuristic approaches based upon
pharmacogenomic analysis of Figures 1-6 could include; Her-2, K-RAS, CDK-4/6,
PI3KCA and MET.
Reviewing Figure 7 suggests a clear issue with the treatment algorithm such as PD-
L1 negative patients treated with Atelozimub (anti-PD1 agent). It could be suggested
that directing this individual towards clinical trial testing could lead to a better
outcome. Furthermore, osimertinib is currently available towards EGFR+ patients
and this drug targets Her-2+ (National Institute for Health and Care Excellence,
2019). A critical and fundamental flaw across this protocol is the lack of personalised
genomic analysis, with Her-2 not possible to request by oncologists (Royal College
of Physicians, 2020). Shengwu et al suggest that Her-2 is the single direct
oncogenic driver for 6% of adenocarcinoma patients, but their breakthrough finding
was the Her-2 amplification acquisition as an underlying mechanism in the
resistance towards EGFR inhibitors (Shengwu et al, 2018). Mouse models indicated
monotherapy-osimertinib presented robust anti-tumour efficacy, suggesting a
potential alternative to the current “unmet clinical demand” (Shengwu et al, 2018).
Although, a previous phase II clinical trial by Gatzemeier et al indicated a poor
response to trastuzumab, with only 1.5 month increase in progression-free survival
(Gatzemeier et al, 2004). A potential flaw in the methodology was the requirement
for ‘untreated patients’ upon EGFR resistance findings (Shengwu et al, 2018).
Recently, Pillai et al’s trial stated patient group administered Her-2 targeted therapies
had median survival of 2.1 years against non-Her-2 therapies 1.4 year survival,
21
suggesting further investigations (Pilla et al, 2017). Upon this, Ogoshi et al identified
that cells with co-contaminant Her-2 and K-Ras mutants do not respond to Her-2
targeted therapies, but results using neratinib against cell lines H2170 and Calu-3
cell lines led inhibition of proliferation, whilst , H1781 cell line indicated a strong
cytotoxic affect (Ogoshi et al, 2019). The current focus is upon phase II clinical trial
(NCT03845270) following prior CLEOPATRA breast cancer methodology using
trastuzumab, pertuzumab and docetaxel (Clinical Trials, 2019) could lead to
promising findings, with CLEOPATRA presenting 37% of patients alive after 8 years
(Swain et al, 2015). Similar to Her-2 amplification, Rehman et al state MET
amplification is another method by which EGFR resistance is acquired, highlighting a
key issue with MET progression by a severe lack of patients leading to a bottleneck
of trials (Rehman, & Dy, 2018). However, a potential solution, as highlighted prior, is
adenocarcinoma patients being grouped into ‘non-squamous’ and analysed for ALK
alterations (National Institute for Health and Care Excellence, 2019., Royal College
of Physicians, 2020 ). If positive, Crizotinib, a ALK/MET inhibitor is used (Chen,
Zhao, & Zhang, 2018), back-tracking adenocarcinoma patients receiving crizotinib
could produce relevant data. Although, critically, less than 1% of adenocarcinoma
patients possess ALK alterations (Collison et al, 2014).
Gopalan et al’s Phase II clinical trial (NCT01291017) using CDK-4/6 inhibitor
palbociclib in previously treated advanced patients (CDK2NA loss and wild-type
RB1) achieved stable disease in 50% of patients but did not affect overall
progression-free survival (Gopalan et al, 2017., Clinical Trials, 2016). Nie et al
suggested a similar finding to Her-2 findings, with palbociclib treated cells
overcoming EGFR resistance to afatinib, this could suggest combination therapy as
22
a method of treatment whilst results are indicating this also reduces the risk of
relapse (Nie et al, 2019). Whilst, Thangavel et al identified novel findings with
palbociclib treated RB1-proficient cells suggested RB1-induced apoptosis,
suggesting the basis for a clinical trial (Thangavel et al, 2018). However, even with
this accumulating support for palbociclib, only one current trial (NCT02664935) for
lung adenocarcinoma is ongoing with 2021 completion (Clinical Trials, 2019-2).
Combination with, Gendicine, a recombinant adenovirus which expresses wild-type
P53 could prove beneficial, as off-label monotherapy has not yielded major results
(Zhang et al, 2017,. Chen et al, 2014., Ning, Sun, & Wang, 2011). A similar issue
with a lack of analysis can be seen with recent breakthrough treatment of a first
generation PI3KCA inhibitor, Alpelisib, which passed phase III breast cancer trials
presenting a 2-fold increase in progression-free survival (Andre et al, 2019). This is
currently being investigated by phase II clinical trial (NCT02276027) in NSCLC
adenocarcinoma patients but the clinical team are not publicly publishing results
(Clinical Trials, 2020). In relation to 86% of PI3KCA mutant patients presenting with
K-Ras or EGFR mutations, therefore, combination therapy could generate promising
results (Wang et al, 2014).
K-Ras mutants are highlighted clearly as a recurrent factor in the underlying
pathogenesis throughout Section 2. A critical issue arises with this common
alteration as no current targeted therapy is available. Recent breakthroughs by
Amgen produced AMG-510, the first K-Ras p.G12C mutant irreversible inhibitor in
clinical development, in pre-clinical testing (NCT03600883) patients remain on AMG-
510 after 42 weeks of use, presenting positive factors such as regression and
synergy with PD-L1 treatments (Canon, Rex, & Saiki, 2019). Upon these findings,
23
AMG-510 has received fast tracking, with two clinical trials (NCT04185883 and
NCT03600883) utilising combination PD-L1 inhibitor:AMG-510 treatment and
monotherapy AMG-510 respectively (Clinical Trials, 2020-2., Clinical Trials, 2020-3).
This focus upon p.G12C links back to Jordan et al’s finding, supporting p.G12C as
the most common K-Ras mutant across both primary and metastatic patients
(Jordan et al, 2017). The promising initial results presenting greater survival in
comparison against the current, suggest AMG-510 as the most promising treatment
in clinical development. Although, in addition to the research areas suggested
throughout, further research into ATK-1 (novel MK-2206), FOSL-1 (novel LY-1816),
and the development of c-MYC inhibitors could lead to alternative promising
treatments in line with pharmacogenomic development (Jansen, Mayer, & Arteaga,
2016., Carabet, Rennie, & Cherkasov, 2018., Yang et al, 2019).
4.0 Conclusion to Review
Analysing copy number and point mutations across NSCLC adenocarcinoma patients
supports findings supporting the basis of adenocarcinoma as MAPK/PI3K/Cell cycle
pathway dysregulation, highlighting previously unappreciated alterations. The
development and introduction of patient personalised analysis is rapidly advancing to
provide targeted therapies to improve patient outcomes, with current EGFR and PD-
L1 analysis enabling targeted treatments leading to a median survival of 216 days
against 203 days without. Forthcoming developments correlate directly to
MAPK/PI3K/Cell cycles with K-ras, HER-2 and CDK-4/6 inhibitors presenting
promising findings. Suggested futuristic scope to improve early diagnosis,
personalised analysis and targeted treatments will lead to major increases in survival
rates.
24
25
Bibliography
Alexandrova, E., Mirza, S., & Xu, S. (2017). p53 loss-of-heterozygosity is a necessary prerequisite for
mutant p53 stabilization and gain-of-function in vivo. Cell Death Dis, 8, e2661.
doi:10.1038/cddis.2017.80
Alonso, I., Liang, H., Turner, S., Lagger, S., Merkel, O., & Kenner, L. (2018). The Role of Activator
Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers, 10(4), 93.
doi:10.3390/cancers10040093
Andre, F., Ciruelos, E., Rubovszky, G., Campone, M., Loibl, S., Hope, S., …, & Lu, Y. (2019).
Alpelisib for PI3KCA-mutated, hormone receptor positive advanced breast cancer. New
England Journal of Medicine, 380, 1929-1940. doi:10.1056/negmoa1813904
Atsaves, V., Leventaki, V., Rassidakis, G., & Claret, F. (2019). AP-1 transcription factors as
regulators of immune responses in cancer. Cancers, 11(7), 1037.
doi:10.3390/cancers11071037
Auerbach, O., Stout, A., Hammond, C., & Garfinkel, L. (1961). Changes in Bronchial Epithelium in
relation to cigarette smoking and in relation to Lung Cancer. The New England Journal of
Medicine, 265, 253-267. Doi:10.10156/NEJM196108102650601
Berger, A., Imielinski, M., & Duke, F. (2014). Oncogenic RIT1 mutations in lung
adenocarcinoma. Oncogene,33, 4418–4423. doi:10.1038/onc.2013.581
26
Canon, J., Rex, K., & Saiki, A. (2019). The clinical KRAS(G12C) inhibitor AMG 510 drives anti-
tumour immunity. Nature, 575, 217–223. doi:10.1038/s41586-019-1694-1
Carabet, L., Rennie, P., & Cherkasov, A. (2018). Therapeutic Inhibition of Myc in Cancer. Structural
Bases and Computer-Aided Drug Discovery Approaches. International journal of molecular
sciences, 20(1), 120. doi:10.3390/ijms20010120
Castellano, E., & Downward, J. (2011). RAS Interaction with PI3K: More Than Just Another Effector
Pathway. Genes & cancer, 2(3), 261–274. Doi:10.117/1947601911408079
Castellano, E., Sheridan, C., Thin, M., Nye, E., Spencer-Dene, B., Diefenbacher, M., …, &
Downward, J. (2013). Requirement for interaction of PI3-kinase p110α with RAS in lung
tumor maintenance. Cancer cell, 24(5), 617–630. https://doi.org/10.1016/j.ccr.2013.09.012
Chen, G., Zhang, S., He, X., Liu, S., Ma, C., & Zou, X. (2014). Clinical utility of recombinant
adenoviral human p53 gene therapy: current perspectives. OncoTargets and therapy, 7, 1901–
1909. doi:10.2147/OTT.S50483
Chen, J., Dong, X., Lei, X., Xia, Y., Zeng, Q., Que, P., … , & Peng, B. (2018). Non-small-cell lung
cancer pathological subtype-related gene selection and bioinformatics analysis based on gene
expression profiles. Molecular and clinical oncology, 8(2), 356–361.
DOI:10.3892/MCO.2017.1516
27
Chen, R., Zhao, J., & Zhang, X. (2018). Crizotinib in advanced non-small-cell lung cancer with
concomitant ALK rearrangement and c-Met overexpression. BMC Cancer 18, 1171.
doi:10.1186/s12885-018-5078-y
Cheng, L., Alexander, R., & MacLennan, G. (2012). Molecular pathology of lung cancer: key to
personalized medicine. Mod Pathology, 25, 347–369. doi:10.1038/modpathol.2011.215
Cheung, W., & Nguyen, D. (2015). Lineage factors and differentiation states in lung cancer
progression. Oncogene 34, 5771–5780. doi:10.1038/onc.2015.85
Chiu, R., Angel, P., & Karin, M. (1989). Jun-B differs in its biological properties form, and is a
negative regulator of, c-Jun. Cell, 59(6), 979-986. doi:10.1016/0092-8674(89)90754-X
Clinical Trials. (2016). Cyclin Dependent Kinase (CDK) 4/6 inhibitor, PD0332991 in advanced non-
small cell lung cancer NSCLC. Retrieved from
https://clinicaltrials.gov/ct2/show/results/NCT01291017#base
Clinical Trials. (2019-2). National Lung matrix trial: Multi-drug phase II trial in non-small cell lung
cancer. Retrieved from
https://clinicaltrials.gov/ct2/show/NCT02664935?term=palbociclib&cond=Adenocarcinoma
+of+Lung&draw=2&rank=1
Clinical Trials. (2019). Her2-positive lung cancer treated with dedicated drug R2D2. Retrieved from
https://clinicaltrials.gov/ct2/show/NCT03845270
28
Clinical Trials. (2020-2). A Phase 1/2, Study Evaluating the Safety, Tolerability, PK, and Efficacy
of AMG 510 in Subjects With Solid Tumors With a Specific KRAS Mutation (CodeBreak
100). Retrieved from https://clinicaltrials.gov/ct2/show/NCT03600883?term=amg-
510&draw=2&rank=2
Clinical Trials. (2020-3). AMG 510 Activity in Subjects With Advanced Solid Tumors With KRAS
p.G12C Mutation (CodeBreak 101). Retrieved from
https://clinicaltrials.gov/ct2/show/NCT04185883?term=amg-510&draw=2&rank=1
Clinical Trials. (2020). A Phase II, Open Label, Multiple Arm Study of AUY922, BYL719, INC280,
LDK378 and MEK162 in Chinese Patients With Advanced Non-small Cell Lung Cancer.
Retrieved from
https://clinicaltrials.gov/ct2/show/NCT02276027?term=alpelisib&cond=Lung+Adenocarcino
ma&draw=2&rank=1
Collison, E., Campbell, J., Brooks, A., Berger, A., William, L., Chmielecki, J., …, Meyerson, M.
(2014). Comprehensive molecular profiling of Lung Adenocarcinoma. Nature, 511, 543–550.
doi:10.1038/nature13385
Crosbie, A., Balata, H., Evison, M., Atack, M., Colligan, D., Duerden, R., …, Booton, R. (2019).
Implementing Lung Cancer Screening: baseline results from a community-based ‘Lung
Health Check’ pilot in deprived areas of Manchester. BMJ Thorax, 74(4), 405-409.
Doi:10.1136/thoraxjnl-2017-211377
29
Desai, T., Brownfield, D., & Krasnow, M. (2014). Alveolar progenitor and stem cells in lung
development, renewal and cancer. Nature, 507(7491), 190–194. Doi:10.1038/nature12930
Devarakonda, S., Morgensztern, D., & Govindan, R. (2015). Genomic Alterations in Lung
Adenocarcinoma. The Lancet Oncology, 16(7), 342,351. Doi:10.1016/s1470-2045(15)00077-
7
Elangovan, I., Vaz, M., Tamatam, C., Potteti, H., Reddy, N., & Reddy, S.(2018). FOSL1 Promotes
Kras-induced Lung Cancer through Amphiregulin and Cell Survival Gene
Regulation. American journal of respiratory cell and molecular biology, 58(5), 625–635.
doi:10.1165/rcmb.2017-0164OC
Feldser, D., Kostova, K., Winslow, M., Taylor, S., Cashman, C., Whittaker, C., …, & Jacks, T.
(2010). Stage-specific sensitivity to p53 restoration during lung cancer
progression. Nature, 468(7323), 572–575. doi:10.1038/nature09535
Gainor, J., Varghese, A., Ignatius, S., Kabraji, S., Awad, M., Katayama, R., …, & Shaw, A. (2013).
ALK rearrangements are mutually exclusive mutations in EGFR and KRAS: An analysis of
1683 patients with non-small cell lung cancer. American Association for Cancer Research,
19(15). doi:10.1158/1078-0432.ccr-13-0318
Gatzemeier, U., Groth, G., Butts, C., Zandwijk, N., Shepherd, F., Ardizzoni, A., …, & Hirsh, V.
(2004). Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in
HER2-positive non-small cell lung cancer. Oncology, 15(1), 19-27.
doi:10.1093/annonc/mdh031
30
Gopalan, P., Pinder, M., Chiappori, A., Ivey, A., Villegas, A., & Kaye, F. (2017). A phase II clinical
trial of the CDK 4/6 inhibitor palbociclib (PD 0332991) in previously treated advanced non-
small cell lung cancer patients with inactivated CDKN2A. Journal of Clinical Oncology,
published before print. doi:10.1200/jco.2014.32.15_suppl.8077
Greulich, H. (2010). The genomics of lung adenocarcinoma: opportunities for targeted
therapies. Genes & cancer, 1(12), 1200–1210. doi:10.1177/1947601911407324
Gul, A., Leyland-Jones, B., Dey, N., & De, P. (2018). A combination of the PI3K pathway inhibitor
plus cell cycle pathway inhibitor to combat endocrine resistance in hormone receptor-positive
breast cancer: a genomic algorithm-based treatment approach. American journal of cancer
research, 8(12), 2359–2376. PMCID:PMC6325472
Hart, J., & Vogt, P. (2011). Phosphorylation of AKT: a mutational analysis. Oncotarget, 2(6), 467–
476. doi:10.18632/oncotarget.293
Hemmings, B., & Restuccia, D. (2012). PI3K-PKB/Akt pathway. Cold Spring Harbor perspectives in
biology, 4(9), a011189. doi:10.1101/cshperspect.a011189
Imielinksi, M., Berger, A., Hammerman, P., Hernandez, B., Pugh, T., Hodis, E., …, & Meyerson, M.
(2012). Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel
Sequencing. Cell, 150(6), 1107-1120. Doi:10.1016/j.cell.2012.08.029
Inamura, K. (2017). Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO
Classification. Frontiers in oncology, 7, 193. doi:10.3389/fonc.2017.00193
31
Jahangiri, A., & Weiss, W. (2013). It takes two to tango: Dual inhibition of PI3K and MAPK in
Rhabdomyosarcoma. Clinical Cancer Research, 19(21). Doi:10.1158/1078-0432.ccr.13-2177
Jakobsen, J., Santoni-Rugiu, E., Grauslund, M., Melchior, L., & Sørensen, J. (2018). Concomitant
driver mutations in advanced EGFR-mutated non-small-cell lung cancer and their impact on
erlotinib treatment. Oncotarget, 9(40), 26195–26208. doi:10.18632/oncotarget.25490
Jansen, V., Mayer, I., & Arteaga, C. (2016). Is there a future for AKT inhibitors in the treatment of
Cancer?. Clinical Cancer Research, 22(11), 2599-2601. doi:10.1158/1078-0432.CCR-16-
0100
Jordan, E., Kim, H., Arcila, M., Barron, D., Chakravarty, D., Gao, J., … & Riely, G.(2017).
Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for
Efficient Patient Matching to Approved and Emerging Therapies. Cancer discovery, 7(6),
596–609. doi:10.1158/2159-8290.CD-16-1337
Junttila, M., Karnezis, A., Garcia, D., Madriles, F., Kortlever, R., Rostker, F., Brown, S., …, &
Martins, C. (2010). Selective activation of p53-mediated tumour suppression in high-grade
tumours. Nature, 468(7323), 567–571. doi:10.1038/nature09526
Jyoti, M., Matteo, M., Negri, E., Vecchia, C., & Boffetta, P. (2016). Risk Factors for Lung Cancer
Worldwide. European Respiratory Journal, 48 (3), 889-902.
32
Kadara, H., & Wistuba, I. (2012). Field cancerization in non-small cell lung cancer: implications in
disease pathogenesis. Proceedings of the American Thoracic Society, 9(2), 38–42.
doi:10.1513/pats.201201-004MS
Kadara, H., Scheet, P., Ignacio, I., & Spira, A. (2016). Early Events in the Molecular Pathogenesis of
Lung Cancer. Cancer Prevention Research, 9(7), 518-527. Doi:10.1158/1940-6270.CAPR-
15-0400
Khakwani, A., Rich, A., Powell, H., Tata, L., Stanley, R., Baldwin, D., Duffy, J., & Hubbard, R.
(2013). Lung cancer survival in England: trends in non-small-cell lung cancer survival over
the duration of the National Lung Cancer Audit. British journal of cancer, 109(8), 2058–
2065. doi:10.1038/bjc.2013.572
Kim, C., Jackson, E., Woolfenden, A., Crowley, D., Bronson, R., Jacks, T., …, Lawrence, S. (2005).
Identification of Bronchioalveolar Stem Cells in Normal Lung and Lung Cancer. Cell,
121(6), 823-835. Doi:10.1016/j.cell.2005.03.032
Kim, C., Jackson, E., Woolfenden, A., Lawrence, S., Babar, I., Vogel, S., …, Jacks, T. (2005).
Identification of Bronchioalveolar Stem Cells in Normal Lung and Lung Cancer. Cell,
121(6), 823-835. Doi:10.1016/j.cell.2005.03.032
Kim, M., Cho, J., Kim, Y., Lee, C., Lee, M., & Shin, D. (2016). Discriminating between Terminal-
and Non-Terminal Respiratory Unit-Type Lung Adenocarcinoma Based on MicroRNA
Profiles. PloS one, 11(8), e0160996. doi:10.1371/journal.pone.0160996
33
Korneeva, N., Song, A., Gram, H., Edens, M., & Rhoads, R. (2015). Inhibition of mitogen-activated
protein kinase (MAPK)-interacting kinase (MNK) preferentially affects translation of
mRNAs containing both a-5’terminal cap and hairpin*. Journal of Biological Chemistry,
291(7), 3455-3467. Doi:10.1074/jbc.m115.694190
Kyoto University Bioinformatics Center. (2019). Keggs: Pathways in Cancer. Retrieved from
https://www.genome.jp/kegg-bin/show_pathway?hsa05200
Lee, Y., & Bae, S. (2016). How do K-RAS-activated cells evade cellular defense
mechanisms?. Oncogene, 35(7), 827–832. doi:10.1038/onc.2015.153
Li, S., Liu, S., Deng, J., Akbay, E., Hai, J., Ambrogio, C., … & Wong, K. (2018). Assessing
Therapeutic Efficacy of MEK Inhibition in a KRASG12C-Driven Mouse Model of Lung
Cancer. Clinical cancer research : an official journal of the American Association for Cancer
Research, 24(19), 4854–4864. doi:10.1158/1078-0432.CCR-17-3438
Lin, C., Song, H., Huang, C., Yao, E., Gacayan, R., Xu, S., & Chuang, P. (2012). Alveolar type II
cells possess the capability of initiating lung tumor development. PloS one, 7(12), e53817.
Doi:10.1371/journal.pone.0053817
Liu, T., Jin, X., Wang, Y., & Wang, K. (2017). Role of epidermal growth factor receptor in lung
cancer and targeted therapies. American journal of cancer research, 7(2), 187–202.
PMID:PMC5336495
34
Lopez-Bergami, P., Lau, E., & Ronai, Z. (2010). Emerging roles of ATF2 and the dynamic AP1
network in cancer. Nature reviews. Cancer, 10(1), 65–76. doi:10.1038/nrc2681
Lu, T., Yang, X., Huang, Y., Zhao, M., Li, M., Ma, K., …, & Wang, Q. (2019). Trends in the
incidence, treatment, and survival of patients with lung cancer in the last four
decades. Cancer management and research, 11, 943–953. doi:10.2147/CMAR.S187317
Matson, J., & Cook, J. (2017). Cell cycle proliferation decisions: the impact of single cell
analyses. The FEBS journal, 284(3), 362–375. doi:10.1111/febs.13898
McPhail, S., Johnson, S., Greenberg, D., Peake, M., & Rous, B. (2015). Stage at diagnosis and early
mortality from cancer in England. British Journal of Cancer, 112, 108–115.
https://doi.org/10.1038/bjc.2015.49
Meng, H., Guo, X., Sun, D., Liang, Y., Lang, J., Han, Y., … , & Geng, J. (2019). Genomic profiling
of driver gene mutations in Chinese patients with non-small cell lung cancer. Frontiers
Genetics. Doi:10.3389/fgene.2019.01008
Molina, J., Yang, P., Cassivi, S., Schild, S., & Adjei, A. (2008). Non-small cell lung cancer:
epidemiology, risk factors, treatment, and survivorship. Mayo Clinic proceedings, 83(5),
584–594. doi.org/10.4065/83.5.584
Moon, Y., Lee, K., Sung, S., & Park, J. (2016). Differing histopathology and prognosis in pulmonary
adenocarcinoma at central and peripheral locations. Journal of thoracic disease, 8(1), 169–
177. https://doi.org/10.3978/j.issn.2072-1439.2016.01.15
35
Murillo, M., Rana, S., Spencer-Dene, B., Nye, E., Stamp, G., & Downward, J. (2018). Disruption of
the Interaction of RAS with PI 3-Kinase Induces Regression of EGFR-Mutant-Driven Lung
Cancer. Cell reports, 25(13), 3545–3553.e2. doi:10.1016/j.celrep.2018.12.003
National Health Service. (2020). Lung Cancer Overview. Retrieved from
https://www.nhs.uk/conditions/lung-cancer/
National Institute for Health and Care Excellence. (2019). Treating non-small-cell lung cancer.
Retrieved from https://pathways.nice.org.uk/pathways/lung-cancer/lung-cancer-
overview#content=view-info-category%3Aview-about-
menu&path=view%3A/pathways/lung-cancer/treating-non-small-cell-lung-cancer.xml
NHS. (2019). Pioneering Manchester Cancer Screening Pilot to be rolled out nationwide. Retrieved
from https://www.christie.nhs.uk/about-us/news/latest-news-stories/pioneering-manchester-
cancer-screening-pilot-to-be-rolled-out-nationwide
NICE. (2011). The Diagnosis and Treatment of Lung Cancer: Clinical Guidelines. Retrieved from
https://www.nice.org.uk/guidance/cg121/documents/lung-cancer-update-full-guideline2
NICE. (2017). Internal Guidelines upon Lung Cancer: Diagnosis and management scope for
consultation. Retrieved from https://www.nice.org.uk/guidance/ng122/documents/draft-scope
36
Nie, H. Zhou, X., Du, S., Nie, C., Zhang, X., & Huang, J. (2019). Palbociclib overcomes afatinib
resistance in non-small cell lung cancer. Biomedicine and Pharmacotherapy, 109, 1750-1757.
Dos:10.1016/j.biopha.2018.10.170
Ning, X., Sun, Z., & Wang, Y. (2011). Docetaxel plus trans-tracheal injection of adenoviral-mediated
p53 versus docetaxel alone in patients with previously treated non-small-cell lung
cancer. Cancer Gene Therapy, 18, 444–449. doi:10.1038/cgt.2011.15
Nussinov, R., Tsai, C., & Jang, H. (2019). Does Ras Activate Raf and PI3K Allosterically?. Frontiers
in oncology, 9, 1231. doi:10.3389/fonc.2019.01231
Office for National Statistics. (2019). Cancer Survival in England: adult, stage at diagnosis and
childhood - patients followed up to 2018. Retrieved from
https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsandd
iseases/bulletins/cancersurvivalinengland/stageatdiagnosisandchildhoodpatientsfollowedupto
2018
Ogoshi, Y., Shien, K., Yoshioka, T., Torigoe, H., Sato, H., Sakaguchi, M., …, & Toyooka, S. (2019).
Anti‑ tumor effect of neratinib against lung cancer cells harboring HER2 oncogene
alterations. Oncology Letters, 17, 2729-2736. doi:10.3892/ol.2019.9908
Okudela, K., Kojima, Y., Matsumara, M., Arai, H., Umeda, S., Tateishi, Y., …, Ohashi, K. (2018).
Relationsjip between non-TRU lung adenocarcinomas and bronchiolar metaplasia - potential
implication in their histogenesis. Histol Histopathology, 33, 317-326. Doi:10.14670/HH-11-
935
37
Ozenne, P., Eymin, B., Brambilla, E., & Gazzeri, S. (2010). The ARF tumour suppressor: structure,
functions and status in cancer. International Journal of Cancer, 127, 2239-2247.
Doi:10.1002/ijc.25511
Philpott, C., Tovell, H., Frayling, I., Cooper, D., & Upadhyaya, M. (2017). The NF1 somatic
mutational landscape in sporadic human cancers. Human genomics, 11(1), 13.
doi:10.1186/s40246-017-0109-3
Pillai, R., Behera, M., Berry, L., Rossi, M., Kris, M., Johnson, B., …, & Khuri, F. (2017). HER2
mutations in lung adenocarcinomas: A report from the Lung Cancer Mutation
Consortium. Cancer, 123(21), 4099–4105. doi:10.1002/cncr.30869
Pradhan, R., Singhvim G., Dubey, S., Gupta, G., & Dua, K. (2019). MAPK pathway: a potential target
for the treatment of non-small cell lung carcinoma. Future medicinal chemistry, 11,8.
Doi:10.4155/fmc-2018-0468
Rehman, S., & Dy, G. (2018). MET inhibition in non-small cell lung cancer. European Medical
Journal, 4(1), 100-111.
Reis, D., Marques, C., Dias, M., Campainha, S., Cirnes, L., & Barroso, A. (2020). Mutational profile
of non-small cell lung cancer patients: Use of next-generation sequencing. Journal of
Pulmonology, 26(1), 50-53. Doi:10.1016/j.pulmoe.2019.05.003
38
Reungwetwattana, T., Weroha, S., & Molina, J. (2012). Oncogenic Pathways, Molecularly Targeted
therapies and highlighted clinical trials in Non-small-cell lung cancer. Clinical lung Cancer,
13(4), 252-266. doi:10.1016/j.cllc.2011.09.004
Rivlin, N., Brosh, R., Oren, M., & Rotter, V. (2011). Mutations in the p53 Tumor Suppressor Gene:
Important Milestones at the Various Steps of Tumorigenesis. Genes & cancer, 2(4), 466–474.
doi:10.1177/1947601911408889
Rodriguez-Viciana, P., Warne, P., Vanhaesebroeck, B., Waterfield, M., & Downward, J. (1996).
Activation of phosphoinositide 3-kinase by interaction with Ras and by point mutation. The
EMBO journal, 15(10), 2442–2451.
Royal College of Physicians. (2018). National Lung Cancer Audit annual report for 2018. London:
Royal College of Physicians, 2019. Retrieved from
https://www.rcplondon.ac.uk/file/12841/download
Royal College of Physicians. (2020). Spotlight report on molecular testing in advanced lung cancer.
London: Royal College of Physicians, 2020. Retrieved from
https://nlcastorage.blob.core.windows.net/misc/NLCA_Spotlight-Molec-Test_2019.pdf
Shengwu, L., Shuai, L., Hai, J., Xiaoen, W., Ting, C., Quinn, M., …, & Wong, K. (2018). Targeting
HER2 Aberrations in non-small cell lung cancer with osimertinib. Clinical Cancer Res,
24(11), 2594-2604. doi:10.1158/1078-0432.ccr-17.1875
39
Siddiqui, N., & Sonenberg, N. (2015). Signalling to eIF4E in cancer. Clinical Cancer Research, 43(5),
763–772. doi:10.1042/BST20150126
Singh, S., Ramamoorthy, M., Vaughan, C., Yeudall, W., Deb, S., & Palit, S. (2013). Human
oncoprotein MDM2 activates the Akt signaling pathway through an interaction with the
repressor element-1 silencing transcription factor conferring a survival advantage to cancer
cells. Cell death and differentiation, 20(4), 558–566. doi:10.1038/cdd.2012.153
Smolle, E., & Pichler, M. (2019). Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms
of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer
Predisposition. Cancers, 11(2), 204. doi.org/10.3390/cancers11020204
Sun, Y., Liu, W., Liu, T., Feng, X., Yang, N., & Zhou, F. (2015). Signalling pathway of MAPK/ERK
in cell proliferation, differentiation, migration, senescence and apoptosis. Journal of Receptor
Signal Transduction Research, 35(6), 600-604. doi:10.3109/10799893.2015.1030412
Sutherland, K. D., & Berns, A. (2010). Cell of origin of lung cancer. Molecular oncology, 4(5), 397–
403. doi:10.1016/j.molonc.2010.05.002
Swain, S., Kim, S-B., Jungsil, R., Vladimir, S., Campone, M., Ciruelos, E., …, & Ross, G. (2015).
Pertuzumab, Trastuzumab and Docetaxel in HER2-positive metastatic breast cancer:
CLEOPATRA. New England Journal of Medicine, 372, 724-734.
doi:10.1015/NEJMoa1413513
40
Tang, Z., Zhang, J., & Lu, X. (2018). Coexistent genetic alterations
involving ALK, RET, ROS1 or MET in 15 cases of lung adenocarcinoma. Mod
Pathol, 31, 307–312. doi:10.1038/modpathol.2017.109
Tanos, T., Marinissen, J., Leskow, C., Hochbaum, D., Martinetto, H., Gutkind, S., & Coso, A. (2005).
Phosphorylation of c-Fos by members of the p38 MAPK family: Role in the AP-1 response
to UV light. Journal of Biol Chem, 280(19), 52. Dos:10.107jbc.m500620200
Thangavel, C., Boopathi, E., Liu, Y., McNair, C., Haber, A., Perepelyuk, M., … , & Den, R. (2018).
Therapeutic Challenge with a CDK4/6 inhibitor induces an RB-dependent SMAC-mediated
apoptotic response in non-small cell lung cancer. Clinical Cancer Research, 24(6).
Doi:10.1158/1078-0432.CCR-17-2074
UK Office for National Statistics. (2017). Deaths registered in England and Wales 2017. Retrieved
https://www.ons.gov.uk/peoplepopulationandcommunity/birthsdeathsandmarriages/deaths/bu
lletins/deathsregisteredinenglandandwalesseriesdr/2017
UK Office for National Statistics. (2018). Cancer Registration statistics, England.
Retrievedhttps://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/condi
tionsanddiseases/bulletins/cancerregistrationstatisticsengland/final2016
UK Office for National Statistics. (2019). Cancer Registration statistics, England. Retrieved
https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsandd
iseases/datasets/cancerregistrationstatisticscancerregistrationstatisticsengland
41
Vallejo, A., Perurena, N., Guruceaga, E., Pawel, K., Zandueta, C., Valencia, K., …, & Vincent,
S.(2017). An integrative approach unveils FOSL1 as an oncogene vulnerability in KRAS-
driven lung and pancreatic cancer. Nature Communications, 8, 14294.
doi:10.1038/ncomms14294
Veen, J., Scherzer, M., Boshuizen, J., Chu, M., Liu, A., Landman, A., …, & McMahon, M. (2019).
Mutationally-activated PI3'-kinase-α promotes de-differentiation of lung tumors initiated by
the BRAFV600E oncoprotein kinase. eLife, 8, e43668. doi:10.7554/eLife.43668
Wan, S., Wright, D., & Coveney, P. (2012). Mechanism of drug efficacy within the EGFR revealed by
microsecond molecular dynamics simulation. Molecular Cancer Therapeutics, 11(11).
Doi:10.1158/1535-7163.MCT-12-0644-T
Wang, L., Haichuan, H., Yunjian, P., Rui, W., Li, Y., Shen, L., …, & Chen, H. (2014). PI3KCA
mutations frequently coexist with EGFR/KRAS mutations in non-small cell lung cancer and
suggest poor prognosis in EGFR/KRAS wildtype group. PLoS ONE, 9(2), e88291.
doi:10.1371/journal.pone.0088291
Wilkerson, D., Yin, D., Walter, V., Zhao, N., Cabanski, C., Hayward, M., Miller, C., …, & Hayes, D.
(2012). Differential pathogenesis of lung adenocarcinoma subtypes involving sequence
mutations, copy number, chromosomal instability, and methylation. PloS one, 7(5), e36530.
https://doi.org/10.1371/journal.pone.0036530
42
Wu, Y., Hung-Chang, W., Jia-En, W., Kuo, H., Shuenn, Y., Si-Xuan, C., …, & Hong, T. (2019). The
dual PI3K/mTOR inhibitor BEZ235 restricts the growth of lung cancer tumors regardless of
EGFR status, as a potent accompanist in combined therapeutic regimens. Journal of
Experimental & Clinical Cancer Research, 38, 282. Doi:10.1186/s13046-019-1282-0
Yang, H., Liang, S., Schmid, R., & Peng, R-W. (2019). New horizons in KRAS-mutant Lung Cancer:
Dawn after Darkness. Frontiers Oncology, 9(953). doi:10.3389/fonc.2019.00953
Yang, W., Meng, L., Chen, K., Tian, C., Peng, B., Zhong, L., …, & Li, L. (2019). Preclinical
pharmacodynamic evaluation of a new Src/FOSL1 inhibitor, LY-1816, in pancreatic ductal
adenocarcinoma. Cancer science, 110(4), 1408–1419. doi:10.1111/cas.13929
Yap, Y., McPherson, J., Ong, C., Rozen, S., Teh, B., Lee, A., & Callen, D. (2014). The NF1 gene
revisited - from bench to bedside. Oncotarget, 5(15), 5873–5892.
doi:10.18632/oncotarget.2194
Yatabe, Y., Borczuk, A., & Powell, C. (2011). Do all lung adenocarcinomas follow a stepwise
progression?. Lung cancer, 74(1), 7–11. doi:10.1016/j.lungcan.2011.05.021
Yatabe, Y., Kosaka, T., Takahashi, T., & Mitsudomi, T. (2005). EGFR mutation is specific for
terminal respiratory unit type adenocarcinoma. The American Journal of Surgical Pathology,
29(5), 633-639. Doi:10.1097/01.pas.00005156935.28066.35
Yousaf-Khan, U., Aalst, C., Jong, P., Heuvelmans, M., Scholten, E., & Lammers, J., …, Koning, H.
(2017). Final screening round of the NELSON lung cancer screening trial: the effect of a 2.5-
year screening interval. BMJ Thorax, 72(6-7), 48-56. Doi:10.1136/thoraxjnl-2016-209755
43
Zappa, C., & Mousa, S. (2016). Non-small cell lung cancer: current treatment and future
advances. Translational lung cancer research, 5(3), 288–300. Doi:10.21037/tlcr.2016.06.07
Zappa, C., & Mousa, S. (2016). Non-small cell lung cancer: current treatment and future
advances. Translational lung cancer research, 5(3), 288–300. doi:10.21037/tlcr.2016.06.07
Zhang, W-W., Li, L., Li, D., Liu, J., Li, X., Li, W., … , & Lam, M-K. (2017). The first approved gene
therapy product for cancer Ad-p53 (Gendicine): 12 years in the clinic. Human Gene Therapy,
29(2). doi:10.1089/hum.2017.218
Zhang, Y., Chang, L., & Yang, Y. (2019). Intratumor heterogeneity comparison among different
subtypes of non-small-cell lung cancer through multi-region tissue and matched ctDNA
sequencing. Mol Cancer, 18, 7. doi:10.1186/s12943-019-0939-9
Zhao, H., Wang, J., & Tony, S. (2015). The phosphatidylinositol 3-kinase/Akt and c-Jun N-terminal
kinase signaling in cancer: Alliance or contradiction?. International Journal of Oncology, 47,
429-436. doi:10.3892/ijo.2015.3052
Zippo, A., Serafini, R., Rocchigiani, M., Pennacchini, S., Krepelova, A., & Oliviero, S. (2009).
Histone crosstalk between H3S10ph and H4K16ac generates a histone code that mediates
transcription elongation. Cell, 138(6), 1122-1136. doi:10.1016/j,cell.2009.07.031

Mais conteúdo relacionado

Mais procurados

Gene expression mining for predicting survivability of patients in earlystage...
Gene expression mining for predicting survivability of patients in earlystage...Gene expression mining for predicting survivability of patients in earlystage...
Gene expression mining for predicting survivability of patients in earlystage...
ijbbjournal
 

Mais procurados (19)

Overexpression of primary microRNA 221/222 in acute myeloid leukemia
Overexpression of primary microRNA 221/222 in acute myeloid leukemiaOverexpression of primary microRNA 221/222 in acute myeloid leukemia
Overexpression of primary microRNA 221/222 in acute myeloid leukemia
 
Stereotactic Radiotherapy of Recurrent Malignant Gliomas Clinical White Paper
Stereotactic Radiotherapy of Recurrent Malignant Gliomas Clinical White PaperStereotactic Radiotherapy of Recurrent Malignant Gliomas Clinical White Paper
Stereotactic Radiotherapy of Recurrent Malignant Gliomas Clinical White Paper
 
Hormone naive prostate cancer
Hormone naive prostate cancerHormone naive prostate cancer
Hormone naive prostate cancer
 
Personalized vs. Precision, let’s call it Medicine
Personalized vs. Precision, let’s call it MedicinePersonalized vs. Precision, let’s call it Medicine
Personalized vs. Precision, let’s call it Medicine
 
88 24-1853 051.812.955.17 folder to Tax Return
88 24-1853  051.812.955.17 folder to Tax Return88 24-1853  051.812.955.17 folder to Tax Return
88 24-1853 051.812.955.17 folder to Tax Return
 
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
hMSH2 Gly322Asp (rs4987188) Single nucleotide polymorphism and the risk of br...
 
Expanding treatment platform in m crc bayer - asyut 2018
Expanding treatment platform in m crc   bayer - asyut 2018Expanding treatment platform in m crc   bayer - asyut 2018
Expanding treatment platform in m crc bayer - asyut 2018
 
Knocking on the clinic door of precision medicine
Knocking on the clinic door of precision medicine Knocking on the clinic door of precision medicine
Knocking on the clinic door of precision medicine
 
1471 2407-13-363
1471 2407-13-3631471 2407-13-363
1471 2407-13-363
 
Ca prostata
Ca prostataCa prostata
Ca prostata
 
Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer Personalized Medicine in Diagnosis and Treatment of Cancer
Personalized Medicine in Diagnosis and Treatment of Cancer
 
Prostate cancer the androgenic fortified dogma
Prostate cancer  the androgenic fortified dogmaProstate cancer  the androgenic fortified dogma
Prostate cancer the androgenic fortified dogma
 
Case study
Case studyCase study
Case study
 
Chemotherapy of head & neck region /certified fixed orthodontic courses by In...
Chemotherapy of head & neck region /certified fixed orthodontic courses by In...Chemotherapy of head & neck region /certified fixed orthodontic courses by In...
Chemotherapy of head & neck region /certified fixed orthodontic courses by In...
 
Gene expression mining for predicting survivability of patients in earlystage...
Gene expression mining for predicting survivability of patients in earlystage...Gene expression mining for predicting survivability of patients in earlystage...
Gene expression mining for predicting survivability of patients in earlystage...
 
CCRT in locally advanced head & neck cancer @imammd
CCRT in locally advanced head & neck cancer @imammdCCRT in locally advanced head & neck cancer @imammd
CCRT in locally advanced head & neck cancer @imammd
 
Metanlysis adjuvant pancreatic
Metanlysis adjuvant pancreaticMetanlysis adjuvant pancreatic
Metanlysis adjuvant pancreatic
 
070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2070125 chemotherapy for hn scc2
070125 chemotherapy for hn scc2
 
Oligometastatic prostate cancer- radiation Therapy
Oligometastatic prostate cancer- radiation TherapyOligometastatic prostate cancer- radiation Therapy
Oligometastatic prostate cancer- radiation Therapy
 

Semelhante a f

2016-Crawford-BMC Pulm Med published
2016-Crawford-BMC Pulm Med published2016-Crawford-BMC Pulm Med published
2016-Crawford-BMC Pulm Med published
Ji-Youn Yeo
 
Sk microfluidics and lab on-a-chip-ch6
Sk microfluidics and lab on-a-chip-ch6Sk microfluidics and lab on-a-chip-ch6
Sk microfluidics and lab on-a-chip-ch6
stanislas547
 

Semelhante a f (20)

ACC Cancer Cell May 2016
ACC Cancer Cell May 2016ACC Cancer Cell May 2016
ACC Cancer Cell May 2016
 
408131
408131408131
408131
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
Deadenylase Expression in Small Cell Lung Cancer Related To Clinical Characte...
 
275758
275758275758
275758
 
MCR_Article_JW
MCR_Article_JWMCR_Article_JW
MCR_Article_JW
 
HDAC4 and HDAC7 Promote Breast and Ovarian Cancer Cell Migration by Regulatin...
HDAC4 and HDAC7 Promote Breast and Ovarian Cancer Cell Migration by Regulatin...HDAC4 and HDAC7 Promote Breast and Ovarian Cancer Cell Migration by Regulatin...
HDAC4 and HDAC7 Promote Breast and Ovarian Cancer Cell Migration by Regulatin...
 
CRC CHP final PDF
CRC CHP final PDFCRC CHP final PDF
CRC CHP final PDF
 
S13148 019-0757-3
S13148 019-0757-3S13148 019-0757-3
S13148 019-0757-3
 
2016-Crawford-BMC Pulm Med published
2016-Crawford-BMC Pulm Med published2016-Crawford-BMC Pulm Med published
2016-Crawford-BMC Pulm Med published
 
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
Sex-Based Difference in Gene Alterations and Biomarkers in Anal Squamous Cell...
 
Secondary Malignancy after Treatment of Prostate Cancer. Radical Prostatectom...
Secondary Malignancy after Treatment of Prostate Cancer. Radical Prostatectom...Secondary Malignancy after Treatment of Prostate Cancer. Radical Prostatectom...
Secondary Malignancy after Treatment of Prostate Cancer. Radical Prostatectom...
 
An Overview: Treatment of Lung Cancer on Researcher Point of View
An Overview: Treatment of Lung Cancer on Researcher Point of ViewAn Overview: Treatment of Lung Cancer on Researcher Point of View
An Overview: Treatment of Lung Cancer on Researcher Point of View
 
Genomic oncology and personalized medicine
Genomic oncology and personalized medicine Genomic oncology and personalized medicine
Genomic oncology and personalized medicine
 
Sk microfluidics and lab on-a-chip-ch6
Sk microfluidics and lab on-a-chip-ch6Sk microfluidics and lab on-a-chip-ch6
Sk microfluidics and lab on-a-chip-ch6
 

Mais de AdamBoulger (20)

1
11
1
 
d
dd
d
 
d
dd
d
 
s
ss
s
 
d
dd
d
 
d
dd
d
 
h
hh
h
 
Jffj
JffjJffj
Jffj
 
Presentation1
Presentation1Presentation1
Presentation1
 
Employable
EmployableEmployable
Employable
 
a
aa
a
 
a
aa
a
 
G
GG
G
 
f
ff
f
 
a
aa
a
 
e
ee
e
 
P
PP
P
 
L
LL
L
 
Pp
PpPp
Pp
 
s
ss
s
 

Último

Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
drmarathore
 
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
ehyxf
 
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
gajnagarg
 
Abortion pills in Dammam +966572737505 Buy Cytotec
Abortion pills in Dammam +966572737505 Buy CytotecAbortion pills in Dammam +966572737505 Buy Cytotec
Abortion pills in Dammam +966572737505 Buy Cytotec
Abortion pills in Riyadh +966572737505 get cytotec
 
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pillsIn Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
Abortion pills in Riyadh +966572737505 get cytotec
 
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
wpkuukw
 
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
uodye
 
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
ehyxf
 
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
wpkuukw
 
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in DammamAbortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
ahmedjiabur940
 
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
ehyxf
 
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
wpkuukw
 
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
tufbav
 
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
uodye
 
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
uodye
 

Último (20)

Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
Abort pregnancy in research centre+966_505195917 abortion pills in Kuwait cyt...
 
Critical Commentary Social Work Ethics.pptx
Critical Commentary Social Work Ethics.pptxCritical Commentary Social Work Ethics.pptx
Critical Commentary Social Work Ethics.pptx
 
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
怎样办理圣芭芭拉分校毕业证(UCSB毕业证书)成绩单留信认证
 
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
Top profile Call Girls In Udgir [ 7014168258 ] Call Me For Genuine Models We ...
 
Abortion pills in Dammam +966572737505 Buy Cytotec
Abortion pills in Dammam +966572737505 Buy CytotecAbortion pills in Dammam +966572737505 Buy Cytotec
Abortion pills in Dammam +966572737505 Buy Cytotec
 
Vashi Affordable Call Girls ,07506202331,Vasai Virar Charming Call Girl
Vashi Affordable Call Girls ,07506202331,Vasai Virar Charming Call GirlVashi Affordable Call Girls ,07506202331,Vasai Virar Charming Call Girl
Vashi Affordable Call Girls ,07506202331,Vasai Virar Charming Call Girl
 
Guwahati Escorts Service Girl ^ 9332606886, WhatsApp Anytime Guwahati
Guwahati Escorts Service Girl ^ 9332606886, WhatsApp Anytime GuwahatiGuwahati Escorts Service Girl ^ 9332606886, WhatsApp Anytime Guwahati
Guwahati Escorts Service Girl ^ 9332606886, WhatsApp Anytime Guwahati
 
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pillsIn Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
In Riyadh Saudi Arabia |+966572737505 | Buy Cytotec| Get Abortion pills
 
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
一比一定(购)新西兰林肯大学毕业证(Lincoln毕业证)成绩单学位证
 
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
一比一原版(Otago毕业证书)奥塔哥理工学院毕业证成绩单学位证靠谱定制
 
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
怎样办理昆士兰大学毕业证(UQ毕业证书)成绩单留信认证
 
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
一比一定(购)国立南方理工学院毕业证(Southern毕业证)成绩单学位证
 
Low Cost Patna Call Girls Service Just Call 🍑👄6378878445 🍑👄 Top Class Call Gi...
Low Cost Patna Call Girls Service Just Call 🍑👄6378878445 🍑👄 Top Class Call Gi...Low Cost Patna Call Girls Service Just Call 🍑👄6378878445 🍑👄 Top Class Call Gi...
Low Cost Patna Call Girls Service Just Call 🍑👄6378878445 🍑👄 Top Class Call Gi...
 
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in DammamAbortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
Abortion Pill for sale in Riyadh ((+918761049707) Get Cytotec in Dammam
 
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
怎样办理伍伦贡大学毕业证(UOW毕业证书)成绩单留信认证
 
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
一比一定(购)UNITEC理工学院毕业证(UNITEC毕业证)成绩单学位证
 
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
怎样办理维多利亚大学毕业证(UVic毕业证书)成绩单留信认证
 
Call Girls Amethi 9332606886 HOT & SEXY Models beautiful and charming call g...
Call Girls Amethi  9332606886 HOT & SEXY Models beautiful and charming call g...Call Girls Amethi  9332606886 HOT & SEXY Models beautiful and charming call g...
Call Girls Amethi 9332606886 HOT & SEXY Models beautiful and charming call g...
 
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
一比一维多利亚大学毕业证(victoria毕业证)成绩单学位证如何办理
 
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
在线制作(UQ毕业证书)昆士兰大学毕业证成绩单原版一比一
 

f

  • 1. 1 Bioscience & Chemistry Programme Professional & Scientific Practice 3 Evaluating the pathogenesis of Lung Adenocarcinoma in relation to NICE treatment guidelines and futuristic treatment developments By Adam Boulger B7040016 Submitted Date: 21/02/2020
  • 2. 2 Table of Contents Abstract ………………………………………………………………………………..3 1. Introduction……………………………………………………………………… 3-5 2. Genetic analysis to identify underlying pathological mechanisms…………. 5-9 2.1 MAPK & PI3K/mTOR pathway dysregulation drives pathogenesis…… ..10-15 2.2 Cell Cycle propelled by MAPK/PI3K effectors……………………………...16-17 3 Current treatment approach to NSCLC Adenocarcinoma…………………...18-19 3.1 Futuristic treatment approach: Personalised Pharmacogenomics……… 19-23 4.0 Conclusion…………………………………………………………………….. 23-24 Bibliography……………………………………………………………………….. 25-43
  • 3. 3 Evaluating the pathogenesis of Lung Adenocarcinoma in relation to NICE treatment guidelines and futuristic treatment developments. Abstract Non-small cell lung cancer (NSCLC) is the deadliest form of cancer, accentuating the clinical demand for more effective treatments in addition to the introduction of early detection programs. Genomic advancements are driving the understanding around the underlying pathogenesis by identifying recurrent MAPK/PI3K and cell cycle pathway dysregulation. Current targeted treatment approach against epidermal growth factor receptor (EGFR) and programmed death-ligand 1 (PD-L1) enable median survival of 216 days against non-targeted 203 days. An underappreciation towards copy number alterations could be a contributing factor and in recent years, advancements are focusing upon human epidermal growth factor receptor-2 (Her-2), Kirsten rat sarcoma (K-Ras) and cyclin-dependent kinases 4/6 (CDK-4/6) inhibitors
  • 4. 4 present the potential to increase disease survival rates. Although, rapid and considerable progress are required due to the poor survival and a lack of specific patient cohorts, hindering developments through a bottleneck in clinical trials. 1. Introduction to Lung Adenocarcinoma Lung cancer is the second most prevalent type of cancer for both males and females across the UK (UK Office for National Statistics, 2018). UK government figures from 2017 outlined 38,888 cancer registrations, consisting of 12.7% of the total cancer burden (UK Office for National Statistics, 2019) forming the deadliest form of cancer and attributed as the fifth leading causation of death with around 30570 deaths during 2017 (UK Office for National Statistics, 2017). This figure, which is greater than breast, colon and pancreatic cancers combined (Zappa, & Mousa, 2016), exemplifies the importance of reviewing and researching lung cancer to improve the approach of diagnosis and treatment of lung cancers. Lung cancer is divided into 13% patients with small-cell and 87% NSCLC (National Health Service, 2020). Although, this is potentially simplified as a major flaw was identified by NHS statistics stating only 72% of lung cancer patients are confirmed by phenotypic or molecular genotyping (NICE, 2017). It could be suggested that this is in relation to around 70% of individuals being diagnosed at an advanced disease state, commonly stated as terminal (Molina, Yang, Cassivi, Schild, & Adjei, 2008). However, research concurs that the major histological form of NSCLC is adenocarcinoma and is accepted as the leading cause of cancer deaths worldwide (Collison et al, 2014), with Reis et al identifying the relative frequency being 90.9% of cases (Reis et al, 2020). Although, comparative analysis with a 2019 publication commissioned and implemented by NHS identified adenocarcinoma at 60% (Crosbie et al, 2019). This agreement outlined the most prevalent sub-type, whilst highlighting clear issues with data standards and a lack of consistency. Until recently, it was thought major sub-types could be characterised by their antithetical anatomical tumour locations, with
  • 5. 5 adenocarcinoma being peripheral (Kadara, Scheet, Ignacio, & Spira, 2016), but this is no longer assumed following Moon et al’s publication highlighting 13.3% of cases being central bound (Moon, Lee, Sung, & Park, 2016). This could suggest reasoning behind the discrepancy identified in the data, therefore, these statistics may not be comparable or accurate. A current focus is upon early diagnosis, with a Manchester research screening pilot across 1384 high risk individuals produced diagnostic results showing 80.4% patients diagnosed at stages I/II compared to the standard of 31% and advanced stages III/IV at 19.6% compared to standard of 69%, with the biggest decrease being for stage IV from 48% to 11% (Crosbie et al, 2019). Furthermore, another screening trial reduced mortality by 26% in men and up to 61% in females (Yousaf-Khan et al, 2017). This directly correlates to findings by McPhail et al, stating that an earlier diagnosis directly correlates to an increased rate of survival (McPhail, Johnson, Greenberg, Peake, & Rous, 2015). This review will evaluate the current understanding of pathogenesis in relation to current and futuristic treatment approaches. 2. Genetic analysis to identify underlying pathological mechanisms Across adenocarcinoma, difficulties identifying the cell of origin have historically hampered the understanding of initiation and progression pathogenesis studies (Devarakonda, Morgensztern, & Govindan, 2015), with alveolar type II cells now accepted as the origin of adenocarcinoma (Kim et al, 2005., Lin et al, 2012). Critically, the issues with cell of origin still exist with central adenocarcinoma. To elucidate the pathogenesis of adenocarcinoma, The Cancer Genome Atlas Research Network (TCGARN) published a large-scale genome project (Collison et al, 2014) to identify prevalent recurrent genetic alterations to be identified in those with adenocarcinoma. This data can be mapped in accordance to affected pathways to suggest the basis of underlying pathological mechanisms. Critically, a major issue with
  • 6. 6 TCGARN’s publication was the focus upon point mutations rather than copy number alterations, although the copy number alteration data was provided. Figure 1: 2-D Bar Chart presenting percentage (%) Lung adenocarcinoma patients with somatic point mutation (missense, nonsense, in-frame deletion, frameshift) against correlating gene. Selected all >10%. Created using The Cancer Genome Atlas Research Network’s publication of data (Collison etal,2014).Loss of function (LOF) mutation to TP53, KEAP1, STK11 and NF1. Gain of function (GOF) mutation to KRAS, EGFR, BRAF. 0 5 10 15 20 25 30 35 40 45 50 BRAF NF1 EGFR SKT11 KEAP1 KRAS TP53 % Patients Geneaffected RecurrentLung adenocarcinomapatient point mutations
  • 7. 7 Figure 2: 2-D Bar Chart presenting % patients presenting with low-medium copy number amplification to a gene. Selected all >10%. Constructed using raw genetic data from supplementary data from The Cancer Genome Atlas Research Network 2014 publication (Collison etal,2014). 0 10 20 30 40 50 60 70 RBM10 HRAS AKT1 CCNE1 U2AF1 MDM2 CDK4 NRAS PI3KCA ARID1A KRAS TERC CCND1 NKX2-1 MET NF1 BRAF ERBB2 EGFR TERT MYC RIT1 % Patients GeneAffected Recurrentlow-mediumcopy number amplifications inLung adenocarcinomapatients
  • 8. 8 0 2 4 6 8 10 12 14 16 18 20 TERT RIT1 NKX2-1 MYC MDM2 EGFR CDK4 KRAS % Patients GeneAffected Recurrenthighcopy number amplificationinlung adenocarcinomapatients Figure 3: 2-D Bar Chart presenting % patients presenting with a specific high copy number amplification to a gene. Selected all >5%. Constructed using raw genetic data from supplementary data from The Cancer Genome Atlas Research Network 2014 publication (Collison etal,2014).
  • 9. 9 The underlying disease mechanisms are yet to be understood (Chen et al, 2018) with Zhang et al highlighting this difficulty due to the complex heterogeneity (Zhang, Chang, & Yang, 2019). However, research has a collective acceptance that the MAPK and PI3K pathways are essential drivers (Wu et al 2019., Pradhan et al 2019). This in relation to 75% of patients presenting with direct oncogenic alterations driving Ras activity (Inamura, 2017) and 62% of patients presenting direct mutations to KRAS and EGFR (Kadara et al, 2016., Yap et al, 2014) with an average of 8.9 mutations per million bases (Collison et al, 2014). 0 10 20 30 40 50 60 CDKN2A SETD2 RB1 KEAP1 SMARCA44 STK11 MGA TP53 % patients GeneAffected Recurrent gene deletions in lung adenocarcinoma patients Figure 4 : 2-D Bar Chart presenting % patients presenting with genetic absence of a gene. Heterozygous loss is coded as blue and homozygous loss (2L) is orange. Heterozygous loss selected top 8 most common, majority of Homozygous loss only present as affecting CDKN2A, second most common was gene MGA with 3.04% patients affected. Constructed using raw genetic data from supplementary data from The Cancer Genome Atlas Research Network 2014 publication (Collison etal,2014). CDKN2A
  • 10. 10 2.1 MAPK & PI3K/mTOR pathway dysregulation drives pathogenesis Figure 5: Created using data outlined in Figures 1, 2, 3 and 4 to review the major pathway modifications to signalling pathways involved in propagation and pathogenesis by identifying recurrent pathogenic drivers to suggest potential treatment approaches to lung adenocarcinoma. Diagram created to review and collate the plethora of recent advancements and publications in the field of adenocarcinoma utilising information from (Collison etal,2014.,Singh et al,2013., Reis et al,2020., Cheung, & Nguyen, 2015., Cheng, Alexander, & MacLennan, 2012., Greulich,2010., Berger, Imielinski,& Duke, 2014., Kadara et al,2016., Korneeva, Song, Gram, Edens, & Rhoads, 2015.,Siddiqui,& Sonenberg, 2015., Jahangiri,& Weiss,2013.,Gul,Leyland-Jones, Dey, & De, 2018., Hemmings, & Restuccia,2012.,Ozenne, Eymin, Brambilla,& Gazzeri, 2010).Key: green
  • 11. 11 arrows represent activation, red arrows represent inhibition, orange boxes highlight oncogenes and grey boxes highlight tumour suppressor genes. Abbreviations: GOF, gain of function, LOF, loss of function, LMA, low medium amplification, HA, high amplification, -1hl heterozygous loss, -2hl, homozygous loss. Chen et al highlighted that although the underlying disease mechanisms have yet to be elucidated, their research analysing KEGG pathway enrichment highlights the importance of the cell cycle and cancer pathways (Chen et al, 2018). Recent publications by Pradhan et al and Wu et al highlight the importance of MAPK and PI3K cancer pathways as essential for pathogenesis by driving the cell cycle (Wu et al 2019., Pradhan et al 2019). Across the genomic landscape of adenocarcinoma, Figure 5 presents the recurrent receptor alterations to MET, ERBB1and ERBB2 which directly induce MAPK and PI3K signalling (Collison et al, 2014) leading to proliferation, evasion of apoptosis and angiogenesis (Sun et al, 2015., Liu, Jin, Wang, & Wang, 2017). The most prevalent is ERBB1, presenting two recurrent gain of function (GOF) aberrations which consist for 90% total mutation burden with exon 21 p.L858R (Wan, Wright, & Coveney, 2012) and micro-deletion p.del19 which directly increase pro-oncogenic downstream signalling (Imielinks et al, 2012., Jakobsen, Santoni-Rugiu, Grauslund, Melchior, & Sørensen, 2018) in addition to 47% of patients presenting copy number amplifications (Collison et al, 2014). This is supported by Kadara et al and Reungwetwattana et al indicating the involvement of ERBB1 and Ras family alterations as the most prominent driver mutations (Kadara et al, 2016., Reungwetwattana, Weroha, & Molina, 2012), with Lee et Bae highlighting K-ras as an important event in the early initiation of adenocarcinoma (Lee, & Bae, 2016). As illustrated in Figure 5, the intercalation between MAPK and PI3K can be driven by K-ras, presenting 33% and 33.5% of patients with GOF and amplifications respectively (Collison et al, 2014). Jordan et al identified the most prevalent somatic alteration as p.G12C across both primary and
  • 12. 12 metastatic patients (Jordan et al, 2017), which links to alternative research highlighting this mutation leads to greater oncogenic activity by higher extracellular signal-related kinase 1/2 (ERK) downstream activatory phosphorylation in comparison to p.G12D (Yang, Liang, Schmid, & Peng, 2019., Li et al, 2018) which drives downstream MAPK activity. This suggests that analysing gene heterogeneity could lead to an improved pharmacogenomic approach. The Kegg publication identified the key effectors of the MAPK pathway as c-Jun, c- Fos, Ets1, MSK1 and c-MYC (Kyoto University Bioinformatics Centre, 2019). ERK’s phosphorylate and activate ETS-Like-1 protein (Elk-1) and Ets-1 transcription factors which rapidly upregulate the Jun and Fos proto-oncogene families, with Kegg highlighting the up-regulation of c-Fos and c-Jun proteins as key which form activator protein-1 (AP-1) heterodimers (Kyoto University Bioinformatics Centre, 2019., Atsaves, Leventaki, Rassidakis, & Claret, 2019) . A landmark paper identified the c- Fos:c-Jun heterodimer exhibits higher transactivation in comparison to alternatives such as JunB or JunD (Chiu, Angel, & Karin, 1989). This activity is activated upon the transcriptional activation domain of c-Jun at sites p.Ser63 and p.Ser73 being phosphorylated by c-Jun N-terminal terminal kinases (JNK) (Zhao, Wang, & Tony, 2015), with Tanos et al identifying c-Fos is activated by P38 MAPK phosphorylation (Tanos et al, 2005). A key finding, highlighted by Alonso et al, is c-Jun promoter and AP-1 exhibiting a high affinity, therefore, it could be suggested that a positive feedback loop exists with this perpetual and self-activatory action exacerbating pathogenesis by increasing AP-1 levels (Alonso et al, 2018), with Figure 5 outlining AP-1 upregulating cyclins, CDK’s and activating E2F transcription factors.
  • 13. 13 The identification of an underlying driver could lead to rapid progression in the treatment of NSCLC adenocarcinoma. Elangovan et al’s breakthrough research discovered Fos family, Fos-like antigen-1 (FOSL-1) as a major effector of RAS-MAPK signalling, presenting FOSL-1 expression as a critical determinant of the tumourigenesis of NSCLC adenocarcinoma, highlighting that emerging data is indicating that FOSL-1 expression and poor survival are directly correlated (Elangovan et al, 2018). These findings were developed upon Vallejo et al’s publication, with data identifying FOSL-1 up regulation in mutant K-ras mutant (p.G12D) cell lines by multiple downstream kinases via an autonomous mechanism (Vallejo et al, 2017). With the key transcriptional regulators being ATF-2, MYC and AP-1 (Lopez-Bergami, Lau, & Ronai, 2010), with a finding identifying a key role of MSK1, by phosphorylating histone H3S10 to induce the transcriptional elongation of FOSL-1, C-Jun and C-Fos (Zippo et al, 2009). This links directly back to Kegg publication highlighting c-Jun, c-Fos, Ets1, MSK1 and c-MYC as the major factors (Kyoto University Bioinformatics Centre, 2019) whilst portraying the symbiotic self-activatory activities of the constituents. Furthermore, as indicated by Figure 5, Warne et al identified the MAPK/PI3K intercalation by Ras activation of PI3K (Rodriguez-Viciana, Warne, Vanhaesebroeck, Waterfield, & Downward, 1996), with Castellano et Downard’s publication presenting the PI3K pathway as essential in RAS mutant in-vivo tumourigenesis studies with PI3KCA’s p.Lys227 being essential in this intercalation activation (Castellano, & Downward, 2011), with 24% of patients presenting amplifications (Collison et al, 2014). However, Nussinov et al recently highlighted the difficulty in identifying the mechanism by which this occurs (Nussinov, Tsai, & Jang, 2019). An investigation into whether Ras heterogeneity (such as K-ras p.G12C>p.G12D discussed above) can
  • 14. 14 induce variable levels of PI3K activity could lead to breakthrough findings. This essential Ras:PI3KCA interaction was disrupted by Castellano et al in Ras driven adenocarcinoma and this led to regression by an autonomous mechanism (Castellano et al, 2013). This was also found by Murillo et al in mutant ERBB1 cells, where disrupting Ras:PI3KCA inhibited tumourigenesis and also induced regression in established adenocarcinoma (Murillo et al, 2018). Furthermore, Veen et al recently published findings with mutant BRAF (p.V600E) cells unable to initiate adenocarcinoma tumourigenesis unless co-expressed with PI3KCA pathway mutant (p.H1047R), compared to mutant K-ras (p.G12D) being able to solely induce tumourigenesis, which is directly associated to RAS:PI3K cross-interaction (Veen et al, 2019). Upon this, it could be suggested that this interaction is a major underlying factor in the pathogenesis, even with a critical flaw in Veen et al’s methodology using PI3KCA mutant not present in TCGARN’s data (Collison et al, 2014). Wang et al highlighted across their study into PI3KCA mutant patients that 86% had coexisting mutations in ERBB1 or K-Ras (Wang et al, 2014). However, Tang et al presented findings which suggested all patients present with co-existing mutations (Tang, Zhang, & Lu, 2018). Although, it could be suggested that a key flaw across papers is the incomparable selection of individuals, therefore, this could lead to inaccurate comparative analysis between papers. Meng et al presents an example of this by focusing only upon Chinese patients (Meng et al, 2019). Furthermore, TCGARN highlight unknown mechanisms of MAPK/PI3K activation exist (Collison et al, 2014), which could link to the under appreciation of copy number alterations mostly under appreciating the alterations to HRAS, NRAS and RIT1 genes. TCGARN data presents 1% of patients present with somatic mutations to these genes
  • 15. 15 (Collison et al, 2014). However, including analysis of copy number alterations identifies high levels of amplifications, with 16%, 23% and 74% patients presenting amplifications respectively. Critically, Figure 5 shows patients can present with loss of function (11%) or amplifications (37%) to NF1, which Yap et al outline is a tumour suppressor by suppressing Ras activity (Yap et al, 2014). This would indicate NF1 reduces MAPK/PI3K activity, although, questioning the understanding of the role of NF1 could lead to novel findings around the functionality as recurrent amplifications to a tumour suppressor with current understanding would not assume to induce adenocarcinoma. This is yet to be analysed in relation to adenocarcinoma, but breast cancer patients present 17% NF1 amplification, with Philpott et al suggesting it is important for the pathogenesis (Philpott, Tovell, Frayling, Cooper, & Upadhyaya, 2017). It could be suggested that NF1 presents oncogenic properties with certain genomic profiles, potentially an investigation into whether NF1 repressing RAS: MAPK leads to an increase of RAS:PI3K interaction.
  • 16. 16 2.2 Cell Cycle propelled by MAPK/PI3Keffectors Figure 6: Constructed upon Figures 1, 2, 3, 4 and 5 to review the major pathway modifications to the cell cycle pathway. Key: green arrows represent activation, red arrows represent inhibition, orange boxes highlight oncogenes, pink highlight transcription factors and grey boxes highlight tumour suppressor genes. References; (Collison etal,2014., Singh et al,2013.,Reis et al,2020., Cheung, & Nguyen, 2015., Cheng et al,2012., Greulich,2010., Berger et al,2014., Kadara et al,2016). The role of MAPK/PI3K outlined in Figure 5, in addition to Figure 6, highlights the recurrent cell cycle alterations, which is a clear presentation of the correlation and symbiotic nature of
  • 17. 17 pathway alterations leading to R-point passage for S-phase which is understood as the ‘point of no return’ for cellular proliferation (Matson, & Cook, 2017). As presented by Figure 6, PI3K effector ATK1 inhibits P53 with ATK1 and MDM2 both present with recurrent amplifications, highlighting the essential inhibition of P53 (Collison et al, 2014). This is also evident by 61% patients presenting a form of CDK2NA loss and this prevents the inhibition of MDM2. (Collison et al, 2014). Furthermore, this is also evident by 46% and 55% of patients presenting with TP53 LOF or heterozygous deletion respectively. This finding is key as in cases of heterozygosity, mutant P53 exhibits the ability to antagonise wild-type P53 in a dominant-negative manner by inhibiting tetramer formation (Rivlin, Brosh, Oren, & Rotter, 2011). Alexandrova et al suggests this could underestimate true P53 loss of function (Alexandrova, Mirza, & Xu, 2017). Although, Lee et Bae stated that it is currently unclear whether TP53 inactivation is the progression or initiation of pathogenesis (Lee, & Bae, 2016). Investigations by Junttila et al & Feldser et al induced adenocarcinoma by mutating Ras/TP53 and reversing this led to regression (Junttila et al, 2010., Feldser et al, 2010). Furthermore, Junttila et al identified Ras mutants are a potent trigger of P53 and the oncogenic driving action of Ras mutants is selective for the acquisition of mutant TP53 genes (Junttila et al, 2010), whilst Feldser et al highlighted MAPK signalling as a critical determinant of adenocarcinoma (Feldser et al, 2010). This is the basis which enables the extreme accumulation of mutations.
  • 18. 18 3.0 Current Treatment Approach to NSCLC Adenocarcinoma Figure 7: Adapted from NICE guidelines illustrating current clinical algorithm (National Institutefor Health and Care Excellence, 2019).Altered to remove ALK analysis following TCGARN data (34) which presented <1% adenocarcinoma patients with ALK mutant. This is a flaw in the current practice due to grouping into ‘squamous’ and ‘non-squamous’ forms.
  • 19. 19 Khakwani analysed UK NSCLC survival trends identifying a slight survival improvement due to increased surgery rate, even with an increasing proportion of NSCLC adenocarcinoma (Khakwani et al, 2013). The current treatment algorithm is fundamentally flawed with the NHS 2019 Lung Cancer audit stating 67.3% and 37% of patients achieved three-month survival and one-year survival respectively with only 18.4% of patients undergoing surgical cancer removal and 40% of stage IIIB patients not receiving treatment (Royal College of Physicians, 2018). Furthermore, a 2020 audit on molecular testing across 1157 advanced ‘non-squamous’ patients 83% of patients analysed following guidelines for EGFR, PD-L1 and ALK alterations (Royal College of Physicians, 2020). Although, clinical data identifies 3.5% of non-squamous patients present with ALK, which does not appear in adenocarcinoma patients (Collison et al, 2014), questioning the rationale behind this. Across the 16.5% EGFR+ patients, 75% received treatment. Median survival was 216 days (Royal College of Physicians, 2020). This is a slight improvement from the median survival of 203 days (2011) (NICE, 2011). It could, therefore, be suggested novel therapeutics are an essential clinical demand. 3.1 Futuristic treatmentapproaches: Personalised Pharmacogenomics The current outdated and generalised histopathological and anatomical classification into squamous or non-squamous should be updated in line with advancements of genomic understanding and to introduce a personalised targeted approach for adenocarcinoma patients. A logical solution suggested by TCGARN’s publication was to characterise by point mutations (Collison et al, 2014), with additions of NF1, STK11 and B-Raf (Collison et al, 2014) to Wilkerson et al’s TP53, K-Ras and ERBB1 suggestions (Wilkerson et al, 2012). Although, it could be suggested this is an
  • 20. 20 incomplete solution with TCGARN stating copy number alterations, presented by Figures 5 and 6, were not considered as pathogenesis driver alterations in their publication (Collison et al, 2014). Potential futuristic approaches based upon pharmacogenomic analysis of Figures 1-6 could include; Her-2, K-RAS, CDK-4/6, PI3KCA and MET. Reviewing Figure 7 suggests a clear issue with the treatment algorithm such as PD- L1 negative patients treated with Atelozimub (anti-PD1 agent). It could be suggested that directing this individual towards clinical trial testing could lead to a better outcome. Furthermore, osimertinib is currently available towards EGFR+ patients and this drug targets Her-2+ (National Institute for Health and Care Excellence, 2019). A critical and fundamental flaw across this protocol is the lack of personalised genomic analysis, with Her-2 not possible to request by oncologists (Royal College of Physicians, 2020). Shengwu et al suggest that Her-2 is the single direct oncogenic driver for 6% of adenocarcinoma patients, but their breakthrough finding was the Her-2 amplification acquisition as an underlying mechanism in the resistance towards EGFR inhibitors (Shengwu et al, 2018). Mouse models indicated monotherapy-osimertinib presented robust anti-tumour efficacy, suggesting a potential alternative to the current “unmet clinical demand” (Shengwu et al, 2018). Although, a previous phase II clinical trial by Gatzemeier et al indicated a poor response to trastuzumab, with only 1.5 month increase in progression-free survival (Gatzemeier et al, 2004). A potential flaw in the methodology was the requirement for ‘untreated patients’ upon EGFR resistance findings (Shengwu et al, 2018). Recently, Pillai et al’s trial stated patient group administered Her-2 targeted therapies had median survival of 2.1 years against non-Her-2 therapies 1.4 year survival,
  • 21. 21 suggesting further investigations (Pilla et al, 2017). Upon this, Ogoshi et al identified that cells with co-contaminant Her-2 and K-Ras mutants do not respond to Her-2 targeted therapies, but results using neratinib against cell lines H2170 and Calu-3 cell lines led inhibition of proliferation, whilst , H1781 cell line indicated a strong cytotoxic affect (Ogoshi et al, 2019). The current focus is upon phase II clinical trial (NCT03845270) following prior CLEOPATRA breast cancer methodology using trastuzumab, pertuzumab and docetaxel (Clinical Trials, 2019) could lead to promising findings, with CLEOPATRA presenting 37% of patients alive after 8 years (Swain et al, 2015). Similar to Her-2 amplification, Rehman et al state MET amplification is another method by which EGFR resistance is acquired, highlighting a key issue with MET progression by a severe lack of patients leading to a bottleneck of trials (Rehman, & Dy, 2018). However, a potential solution, as highlighted prior, is adenocarcinoma patients being grouped into ‘non-squamous’ and analysed for ALK alterations (National Institute for Health and Care Excellence, 2019., Royal College of Physicians, 2020 ). If positive, Crizotinib, a ALK/MET inhibitor is used (Chen, Zhao, & Zhang, 2018), back-tracking adenocarcinoma patients receiving crizotinib could produce relevant data. Although, critically, less than 1% of adenocarcinoma patients possess ALK alterations (Collison et al, 2014). Gopalan et al’s Phase II clinical trial (NCT01291017) using CDK-4/6 inhibitor palbociclib in previously treated advanced patients (CDK2NA loss and wild-type RB1) achieved stable disease in 50% of patients but did not affect overall progression-free survival (Gopalan et al, 2017., Clinical Trials, 2016). Nie et al suggested a similar finding to Her-2 findings, with palbociclib treated cells overcoming EGFR resistance to afatinib, this could suggest combination therapy as
  • 22. 22 a method of treatment whilst results are indicating this also reduces the risk of relapse (Nie et al, 2019). Whilst, Thangavel et al identified novel findings with palbociclib treated RB1-proficient cells suggested RB1-induced apoptosis, suggesting the basis for a clinical trial (Thangavel et al, 2018). However, even with this accumulating support for palbociclib, only one current trial (NCT02664935) for lung adenocarcinoma is ongoing with 2021 completion (Clinical Trials, 2019-2). Combination with, Gendicine, a recombinant adenovirus which expresses wild-type P53 could prove beneficial, as off-label monotherapy has not yielded major results (Zhang et al, 2017,. Chen et al, 2014., Ning, Sun, & Wang, 2011). A similar issue with a lack of analysis can be seen with recent breakthrough treatment of a first generation PI3KCA inhibitor, Alpelisib, which passed phase III breast cancer trials presenting a 2-fold increase in progression-free survival (Andre et al, 2019). This is currently being investigated by phase II clinical trial (NCT02276027) in NSCLC adenocarcinoma patients but the clinical team are not publicly publishing results (Clinical Trials, 2020). In relation to 86% of PI3KCA mutant patients presenting with K-Ras or EGFR mutations, therefore, combination therapy could generate promising results (Wang et al, 2014). K-Ras mutants are highlighted clearly as a recurrent factor in the underlying pathogenesis throughout Section 2. A critical issue arises with this common alteration as no current targeted therapy is available. Recent breakthroughs by Amgen produced AMG-510, the first K-Ras p.G12C mutant irreversible inhibitor in clinical development, in pre-clinical testing (NCT03600883) patients remain on AMG- 510 after 42 weeks of use, presenting positive factors such as regression and synergy with PD-L1 treatments (Canon, Rex, & Saiki, 2019). Upon these findings,
  • 23. 23 AMG-510 has received fast tracking, with two clinical trials (NCT04185883 and NCT03600883) utilising combination PD-L1 inhibitor:AMG-510 treatment and monotherapy AMG-510 respectively (Clinical Trials, 2020-2., Clinical Trials, 2020-3). This focus upon p.G12C links back to Jordan et al’s finding, supporting p.G12C as the most common K-Ras mutant across both primary and metastatic patients (Jordan et al, 2017). The promising initial results presenting greater survival in comparison against the current, suggest AMG-510 as the most promising treatment in clinical development. Although, in addition to the research areas suggested throughout, further research into ATK-1 (novel MK-2206), FOSL-1 (novel LY-1816), and the development of c-MYC inhibitors could lead to alternative promising treatments in line with pharmacogenomic development (Jansen, Mayer, & Arteaga, 2016., Carabet, Rennie, & Cherkasov, 2018., Yang et al, 2019). 4.0 Conclusion to Review Analysing copy number and point mutations across NSCLC adenocarcinoma patients supports findings supporting the basis of adenocarcinoma as MAPK/PI3K/Cell cycle pathway dysregulation, highlighting previously unappreciated alterations. The development and introduction of patient personalised analysis is rapidly advancing to provide targeted therapies to improve patient outcomes, with current EGFR and PD- L1 analysis enabling targeted treatments leading to a median survival of 216 days against 203 days without. Forthcoming developments correlate directly to MAPK/PI3K/Cell cycles with K-ras, HER-2 and CDK-4/6 inhibitors presenting promising findings. Suggested futuristic scope to improve early diagnosis, personalised analysis and targeted treatments will lead to major increases in survival rates.
  • 24. 24
  • 25. 25 Bibliography Alexandrova, E., Mirza, S., & Xu, S. (2017). p53 loss-of-heterozygosity is a necessary prerequisite for mutant p53 stabilization and gain-of-function in vivo. Cell Death Dis, 8, e2661. doi:10.1038/cddis.2017.80 Alonso, I., Liang, H., Turner, S., Lagger, S., Merkel, O., & Kenner, L. (2018). The Role of Activator Protein-1 (AP-1) Family Members in CD30-Positive Lymphomas. Cancers, 10(4), 93. doi:10.3390/cancers10040093 Andre, F., Ciruelos, E., Rubovszky, G., Campone, M., Loibl, S., Hope, S., …, & Lu, Y. (2019). Alpelisib for PI3KCA-mutated, hormone receptor positive advanced breast cancer. New England Journal of Medicine, 380, 1929-1940. doi:10.1056/negmoa1813904 Atsaves, V., Leventaki, V., Rassidakis, G., & Claret, F. (2019). AP-1 transcription factors as regulators of immune responses in cancer. Cancers, 11(7), 1037. doi:10.3390/cancers11071037 Auerbach, O., Stout, A., Hammond, C., & Garfinkel, L. (1961). Changes in Bronchial Epithelium in relation to cigarette smoking and in relation to Lung Cancer. The New England Journal of Medicine, 265, 253-267. Doi:10.10156/NEJM196108102650601 Berger, A., Imielinski, M., & Duke, F. (2014). Oncogenic RIT1 mutations in lung adenocarcinoma. Oncogene,33, 4418–4423. doi:10.1038/onc.2013.581
  • 26. 26 Canon, J., Rex, K., & Saiki, A. (2019). The clinical KRAS(G12C) inhibitor AMG 510 drives anti- tumour immunity. Nature, 575, 217–223. doi:10.1038/s41586-019-1694-1 Carabet, L., Rennie, P., & Cherkasov, A. (2018). Therapeutic Inhibition of Myc in Cancer. Structural Bases and Computer-Aided Drug Discovery Approaches. International journal of molecular sciences, 20(1), 120. doi:10.3390/ijms20010120 Castellano, E., & Downward, J. (2011). RAS Interaction with PI3K: More Than Just Another Effector Pathway. Genes & cancer, 2(3), 261–274. Doi:10.117/1947601911408079 Castellano, E., Sheridan, C., Thin, M., Nye, E., Spencer-Dene, B., Diefenbacher, M., …, & Downward, J. (2013). Requirement for interaction of PI3-kinase p110α with RAS in lung tumor maintenance. Cancer cell, 24(5), 617–630. https://doi.org/10.1016/j.ccr.2013.09.012 Chen, G., Zhang, S., He, X., Liu, S., Ma, C., & Zou, X. (2014). Clinical utility of recombinant adenoviral human p53 gene therapy: current perspectives. OncoTargets and therapy, 7, 1901– 1909. doi:10.2147/OTT.S50483 Chen, J., Dong, X., Lei, X., Xia, Y., Zeng, Q., Que, P., … , & Peng, B. (2018). Non-small-cell lung cancer pathological subtype-related gene selection and bioinformatics analysis based on gene expression profiles. Molecular and clinical oncology, 8(2), 356–361. DOI:10.3892/MCO.2017.1516
  • 27. 27 Chen, R., Zhao, J., & Zhang, X. (2018). Crizotinib in advanced non-small-cell lung cancer with concomitant ALK rearrangement and c-Met overexpression. BMC Cancer 18, 1171. doi:10.1186/s12885-018-5078-y Cheng, L., Alexander, R., & MacLennan, G. (2012). Molecular pathology of lung cancer: key to personalized medicine. Mod Pathology, 25, 347–369. doi:10.1038/modpathol.2011.215 Cheung, W., & Nguyen, D. (2015). Lineage factors and differentiation states in lung cancer progression. Oncogene 34, 5771–5780. doi:10.1038/onc.2015.85 Chiu, R., Angel, P., & Karin, M. (1989). Jun-B differs in its biological properties form, and is a negative regulator of, c-Jun. Cell, 59(6), 979-986. doi:10.1016/0092-8674(89)90754-X Clinical Trials. (2016). Cyclin Dependent Kinase (CDK) 4/6 inhibitor, PD0332991 in advanced non- small cell lung cancer NSCLC. Retrieved from https://clinicaltrials.gov/ct2/show/results/NCT01291017#base Clinical Trials. (2019-2). National Lung matrix trial: Multi-drug phase II trial in non-small cell lung cancer. Retrieved from https://clinicaltrials.gov/ct2/show/NCT02664935?term=palbociclib&cond=Adenocarcinoma +of+Lung&draw=2&rank=1 Clinical Trials. (2019). Her2-positive lung cancer treated with dedicated drug R2D2. Retrieved from https://clinicaltrials.gov/ct2/show/NCT03845270
  • 28. 28 Clinical Trials. (2020-2). A Phase 1/2, Study Evaluating the Safety, Tolerability, PK, and Efficacy of AMG 510 in Subjects With Solid Tumors With a Specific KRAS Mutation (CodeBreak 100). Retrieved from https://clinicaltrials.gov/ct2/show/NCT03600883?term=amg- 510&draw=2&rank=2 Clinical Trials. (2020-3). AMG 510 Activity in Subjects With Advanced Solid Tumors With KRAS p.G12C Mutation (CodeBreak 101). Retrieved from https://clinicaltrials.gov/ct2/show/NCT04185883?term=amg-510&draw=2&rank=1 Clinical Trials. (2020). A Phase II, Open Label, Multiple Arm Study of AUY922, BYL719, INC280, LDK378 and MEK162 in Chinese Patients With Advanced Non-small Cell Lung Cancer. Retrieved from https://clinicaltrials.gov/ct2/show/NCT02276027?term=alpelisib&cond=Lung+Adenocarcino ma&draw=2&rank=1 Collison, E., Campbell, J., Brooks, A., Berger, A., William, L., Chmielecki, J., …, Meyerson, M. (2014). Comprehensive molecular profiling of Lung Adenocarcinoma. Nature, 511, 543–550. doi:10.1038/nature13385 Crosbie, A., Balata, H., Evison, M., Atack, M., Colligan, D., Duerden, R., …, Booton, R. (2019). Implementing Lung Cancer Screening: baseline results from a community-based ‘Lung Health Check’ pilot in deprived areas of Manchester. BMJ Thorax, 74(4), 405-409. Doi:10.1136/thoraxjnl-2017-211377
  • 29. 29 Desai, T., Brownfield, D., & Krasnow, M. (2014). Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature, 507(7491), 190–194. Doi:10.1038/nature12930 Devarakonda, S., Morgensztern, D., & Govindan, R. (2015). Genomic Alterations in Lung Adenocarcinoma. The Lancet Oncology, 16(7), 342,351. Doi:10.1016/s1470-2045(15)00077- 7 Elangovan, I., Vaz, M., Tamatam, C., Potteti, H., Reddy, N., & Reddy, S.(2018). FOSL1 Promotes Kras-induced Lung Cancer through Amphiregulin and Cell Survival Gene Regulation. American journal of respiratory cell and molecular biology, 58(5), 625–635. doi:10.1165/rcmb.2017-0164OC Feldser, D., Kostova, K., Winslow, M., Taylor, S., Cashman, C., Whittaker, C., …, & Jacks, T. (2010). Stage-specific sensitivity to p53 restoration during lung cancer progression. Nature, 468(7323), 572–575. doi:10.1038/nature09535 Gainor, J., Varghese, A., Ignatius, S., Kabraji, S., Awad, M., Katayama, R., …, & Shaw, A. (2013). ALK rearrangements are mutually exclusive mutations in EGFR and KRAS: An analysis of 1683 patients with non-small cell lung cancer. American Association for Cancer Research, 19(15). doi:10.1158/1078-0432.ccr-13-0318 Gatzemeier, U., Groth, G., Butts, C., Zandwijk, N., Shepherd, F., Ardizzoni, A., …, & Hirsh, V. (2004). Randomized phase II trial of gemcitabine-cisplatin with or without trastuzumab in HER2-positive non-small cell lung cancer. Oncology, 15(1), 19-27. doi:10.1093/annonc/mdh031
  • 30. 30 Gopalan, P., Pinder, M., Chiappori, A., Ivey, A., Villegas, A., & Kaye, F. (2017). A phase II clinical trial of the CDK 4/6 inhibitor palbociclib (PD 0332991) in previously treated advanced non- small cell lung cancer patients with inactivated CDKN2A. Journal of Clinical Oncology, published before print. doi:10.1200/jco.2014.32.15_suppl.8077 Greulich, H. (2010). The genomics of lung adenocarcinoma: opportunities for targeted therapies. Genes & cancer, 1(12), 1200–1210. doi:10.1177/1947601911407324 Gul, A., Leyland-Jones, B., Dey, N., & De, P. (2018). A combination of the PI3K pathway inhibitor plus cell cycle pathway inhibitor to combat endocrine resistance in hormone receptor-positive breast cancer: a genomic algorithm-based treatment approach. American journal of cancer research, 8(12), 2359–2376. PMCID:PMC6325472 Hart, J., & Vogt, P. (2011). Phosphorylation of AKT: a mutational analysis. Oncotarget, 2(6), 467– 476. doi:10.18632/oncotarget.293 Hemmings, B., & Restuccia, D. (2012). PI3K-PKB/Akt pathway. Cold Spring Harbor perspectives in biology, 4(9), a011189. doi:10.1101/cshperspect.a011189 Imielinksi, M., Berger, A., Hammerman, P., Hernandez, B., Pugh, T., Hodis, E., …, & Meyerson, M. (2012). Mapping the Hallmarks of Lung Adenocarcinoma with Massively Parallel Sequencing. Cell, 150(6), 1107-1120. Doi:10.1016/j.cell.2012.08.029 Inamura, K. (2017). Lung Cancer: Understanding Its Molecular Pathology and the 2015 WHO Classification. Frontiers in oncology, 7, 193. doi:10.3389/fonc.2017.00193
  • 31. 31 Jahangiri, A., & Weiss, W. (2013). It takes two to tango: Dual inhibition of PI3K and MAPK in Rhabdomyosarcoma. Clinical Cancer Research, 19(21). Doi:10.1158/1078-0432.ccr.13-2177 Jakobsen, J., Santoni-Rugiu, E., Grauslund, M., Melchior, L., & Sørensen, J. (2018). Concomitant driver mutations in advanced EGFR-mutated non-small-cell lung cancer and their impact on erlotinib treatment. Oncotarget, 9(40), 26195–26208. doi:10.18632/oncotarget.25490 Jansen, V., Mayer, I., & Arteaga, C. (2016). Is there a future for AKT inhibitors in the treatment of Cancer?. Clinical Cancer Research, 22(11), 2599-2601. doi:10.1158/1078-0432.CCR-16- 0100 Jordan, E., Kim, H., Arcila, M., Barron, D., Chakravarty, D., Gao, J., … & Riely, G.(2017). Prospective Comprehensive Molecular Characterization of Lung Adenocarcinomas for Efficient Patient Matching to Approved and Emerging Therapies. Cancer discovery, 7(6), 596–609. doi:10.1158/2159-8290.CD-16-1337 Junttila, M., Karnezis, A., Garcia, D., Madriles, F., Kortlever, R., Rostker, F., Brown, S., …, & Martins, C. (2010). Selective activation of p53-mediated tumour suppression in high-grade tumours. Nature, 468(7323), 567–571. doi:10.1038/nature09526 Jyoti, M., Matteo, M., Negri, E., Vecchia, C., & Boffetta, P. (2016). Risk Factors for Lung Cancer Worldwide. European Respiratory Journal, 48 (3), 889-902.
  • 32. 32 Kadara, H., & Wistuba, I. (2012). Field cancerization in non-small cell lung cancer: implications in disease pathogenesis. Proceedings of the American Thoracic Society, 9(2), 38–42. doi:10.1513/pats.201201-004MS Kadara, H., Scheet, P., Ignacio, I., & Spira, A. (2016). Early Events in the Molecular Pathogenesis of Lung Cancer. Cancer Prevention Research, 9(7), 518-527. Doi:10.1158/1940-6270.CAPR- 15-0400 Khakwani, A., Rich, A., Powell, H., Tata, L., Stanley, R., Baldwin, D., Duffy, J., & Hubbard, R. (2013). Lung cancer survival in England: trends in non-small-cell lung cancer survival over the duration of the National Lung Cancer Audit. British journal of cancer, 109(8), 2058– 2065. doi:10.1038/bjc.2013.572 Kim, C., Jackson, E., Woolfenden, A., Crowley, D., Bronson, R., Jacks, T., …, Lawrence, S. (2005). Identification of Bronchioalveolar Stem Cells in Normal Lung and Lung Cancer. Cell, 121(6), 823-835. Doi:10.1016/j.cell.2005.03.032 Kim, C., Jackson, E., Woolfenden, A., Lawrence, S., Babar, I., Vogel, S., …, Jacks, T. (2005). Identification of Bronchioalveolar Stem Cells in Normal Lung and Lung Cancer. Cell, 121(6), 823-835. Doi:10.1016/j.cell.2005.03.032 Kim, M., Cho, J., Kim, Y., Lee, C., Lee, M., & Shin, D. (2016). Discriminating between Terminal- and Non-Terminal Respiratory Unit-Type Lung Adenocarcinoma Based on MicroRNA Profiles. PloS one, 11(8), e0160996. doi:10.1371/journal.pone.0160996
  • 33. 33 Korneeva, N., Song, A., Gram, H., Edens, M., & Rhoads, R. (2015). Inhibition of mitogen-activated protein kinase (MAPK)-interacting kinase (MNK) preferentially affects translation of mRNAs containing both a-5’terminal cap and hairpin*. Journal of Biological Chemistry, 291(7), 3455-3467. Doi:10.1074/jbc.m115.694190 Kyoto University Bioinformatics Center. (2019). Keggs: Pathways in Cancer. Retrieved from https://www.genome.jp/kegg-bin/show_pathway?hsa05200 Lee, Y., & Bae, S. (2016). How do K-RAS-activated cells evade cellular defense mechanisms?. Oncogene, 35(7), 827–832. doi:10.1038/onc.2015.153 Li, S., Liu, S., Deng, J., Akbay, E., Hai, J., Ambrogio, C., … & Wong, K. (2018). Assessing Therapeutic Efficacy of MEK Inhibition in a KRASG12C-Driven Mouse Model of Lung Cancer. Clinical cancer research : an official journal of the American Association for Cancer Research, 24(19), 4854–4864. doi:10.1158/1078-0432.CCR-17-3438 Lin, C., Song, H., Huang, C., Yao, E., Gacayan, R., Xu, S., & Chuang, P. (2012). Alveolar type II cells possess the capability of initiating lung tumor development. PloS one, 7(12), e53817. Doi:10.1371/journal.pone.0053817 Liu, T., Jin, X., Wang, Y., & Wang, K. (2017). Role of epidermal growth factor receptor in lung cancer and targeted therapies. American journal of cancer research, 7(2), 187–202. PMID:PMC5336495
  • 34. 34 Lopez-Bergami, P., Lau, E., & Ronai, Z. (2010). Emerging roles of ATF2 and the dynamic AP1 network in cancer. Nature reviews. Cancer, 10(1), 65–76. doi:10.1038/nrc2681 Lu, T., Yang, X., Huang, Y., Zhao, M., Li, M., Ma, K., …, & Wang, Q. (2019). Trends in the incidence, treatment, and survival of patients with lung cancer in the last four decades. Cancer management and research, 11, 943–953. doi:10.2147/CMAR.S187317 Matson, J., & Cook, J. (2017). Cell cycle proliferation decisions: the impact of single cell analyses. The FEBS journal, 284(3), 362–375. doi:10.1111/febs.13898 McPhail, S., Johnson, S., Greenberg, D., Peake, M., & Rous, B. (2015). Stage at diagnosis and early mortality from cancer in England. British Journal of Cancer, 112, 108–115. https://doi.org/10.1038/bjc.2015.49 Meng, H., Guo, X., Sun, D., Liang, Y., Lang, J., Han, Y., … , & Geng, J. (2019). Genomic profiling of driver gene mutations in Chinese patients with non-small cell lung cancer. Frontiers Genetics. Doi:10.3389/fgene.2019.01008 Molina, J., Yang, P., Cassivi, S., Schild, S., & Adjei, A. (2008). Non-small cell lung cancer: epidemiology, risk factors, treatment, and survivorship. Mayo Clinic proceedings, 83(5), 584–594. doi.org/10.4065/83.5.584 Moon, Y., Lee, K., Sung, S., & Park, J. (2016). Differing histopathology and prognosis in pulmonary adenocarcinoma at central and peripheral locations. Journal of thoracic disease, 8(1), 169– 177. https://doi.org/10.3978/j.issn.2072-1439.2016.01.15
  • 35. 35 Murillo, M., Rana, S., Spencer-Dene, B., Nye, E., Stamp, G., & Downward, J. (2018). Disruption of the Interaction of RAS with PI 3-Kinase Induces Regression of EGFR-Mutant-Driven Lung Cancer. Cell reports, 25(13), 3545–3553.e2. doi:10.1016/j.celrep.2018.12.003 National Health Service. (2020). Lung Cancer Overview. Retrieved from https://www.nhs.uk/conditions/lung-cancer/ National Institute for Health and Care Excellence. (2019). Treating non-small-cell lung cancer. Retrieved from https://pathways.nice.org.uk/pathways/lung-cancer/lung-cancer- overview#content=view-info-category%3Aview-about- menu&path=view%3A/pathways/lung-cancer/treating-non-small-cell-lung-cancer.xml NHS. (2019). Pioneering Manchester Cancer Screening Pilot to be rolled out nationwide. Retrieved from https://www.christie.nhs.uk/about-us/news/latest-news-stories/pioneering-manchester- cancer-screening-pilot-to-be-rolled-out-nationwide NICE. (2011). The Diagnosis and Treatment of Lung Cancer: Clinical Guidelines. Retrieved from https://www.nice.org.uk/guidance/cg121/documents/lung-cancer-update-full-guideline2 NICE. (2017). Internal Guidelines upon Lung Cancer: Diagnosis and management scope for consultation. Retrieved from https://www.nice.org.uk/guidance/ng122/documents/draft-scope
  • 36. 36 Nie, H. Zhou, X., Du, S., Nie, C., Zhang, X., & Huang, J. (2019). Palbociclib overcomes afatinib resistance in non-small cell lung cancer. Biomedicine and Pharmacotherapy, 109, 1750-1757. Dos:10.1016/j.biopha.2018.10.170 Ning, X., Sun, Z., & Wang, Y. (2011). Docetaxel plus trans-tracheal injection of adenoviral-mediated p53 versus docetaxel alone in patients with previously treated non-small-cell lung cancer. Cancer Gene Therapy, 18, 444–449. doi:10.1038/cgt.2011.15 Nussinov, R., Tsai, C., & Jang, H. (2019). Does Ras Activate Raf and PI3K Allosterically?. Frontiers in oncology, 9, 1231. doi:10.3389/fonc.2019.01231 Office for National Statistics. (2019). Cancer Survival in England: adult, stage at diagnosis and childhood - patients followed up to 2018. Retrieved from https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsandd iseases/bulletins/cancersurvivalinengland/stageatdiagnosisandchildhoodpatientsfollowedupto 2018 Ogoshi, Y., Shien, K., Yoshioka, T., Torigoe, H., Sato, H., Sakaguchi, M., …, & Toyooka, S. (2019). Anti‑ tumor effect of neratinib against lung cancer cells harboring HER2 oncogene alterations. Oncology Letters, 17, 2729-2736. doi:10.3892/ol.2019.9908 Okudela, K., Kojima, Y., Matsumara, M., Arai, H., Umeda, S., Tateishi, Y., …, Ohashi, K. (2018). Relationsjip between non-TRU lung adenocarcinomas and bronchiolar metaplasia - potential implication in their histogenesis. Histol Histopathology, 33, 317-326. Doi:10.14670/HH-11- 935
  • 37. 37 Ozenne, P., Eymin, B., Brambilla, E., & Gazzeri, S. (2010). The ARF tumour suppressor: structure, functions and status in cancer. International Journal of Cancer, 127, 2239-2247. Doi:10.1002/ijc.25511 Philpott, C., Tovell, H., Frayling, I., Cooper, D., & Upadhyaya, M. (2017). The NF1 somatic mutational landscape in sporadic human cancers. Human genomics, 11(1), 13. doi:10.1186/s40246-017-0109-3 Pillai, R., Behera, M., Berry, L., Rossi, M., Kris, M., Johnson, B., …, & Khuri, F. (2017). HER2 mutations in lung adenocarcinomas: A report from the Lung Cancer Mutation Consortium. Cancer, 123(21), 4099–4105. doi:10.1002/cncr.30869 Pradhan, R., Singhvim G., Dubey, S., Gupta, G., & Dua, K. (2019). MAPK pathway: a potential target for the treatment of non-small cell lung carcinoma. Future medicinal chemistry, 11,8. Doi:10.4155/fmc-2018-0468 Rehman, S., & Dy, G. (2018). MET inhibition in non-small cell lung cancer. European Medical Journal, 4(1), 100-111. Reis, D., Marques, C., Dias, M., Campainha, S., Cirnes, L., & Barroso, A. (2020). Mutational profile of non-small cell lung cancer patients: Use of next-generation sequencing. Journal of Pulmonology, 26(1), 50-53. Doi:10.1016/j.pulmoe.2019.05.003
  • 38. 38 Reungwetwattana, T., Weroha, S., & Molina, J. (2012). Oncogenic Pathways, Molecularly Targeted therapies and highlighted clinical trials in Non-small-cell lung cancer. Clinical lung Cancer, 13(4), 252-266. doi:10.1016/j.cllc.2011.09.004 Rivlin, N., Brosh, R., Oren, M., & Rotter, V. (2011). Mutations in the p53 Tumor Suppressor Gene: Important Milestones at the Various Steps of Tumorigenesis. Genes & cancer, 2(4), 466–474. doi:10.1177/1947601911408889 Rodriguez-Viciana, P., Warne, P., Vanhaesebroeck, B., Waterfield, M., & Downward, J. (1996). Activation of phosphoinositide 3-kinase by interaction with Ras and by point mutation. The EMBO journal, 15(10), 2442–2451. Royal College of Physicians. (2018). National Lung Cancer Audit annual report for 2018. London: Royal College of Physicians, 2019. Retrieved from https://www.rcplondon.ac.uk/file/12841/download Royal College of Physicians. (2020). Spotlight report on molecular testing in advanced lung cancer. London: Royal College of Physicians, 2020. Retrieved from https://nlcastorage.blob.core.windows.net/misc/NLCA_Spotlight-Molec-Test_2019.pdf Shengwu, L., Shuai, L., Hai, J., Xiaoen, W., Ting, C., Quinn, M., …, & Wong, K. (2018). Targeting HER2 Aberrations in non-small cell lung cancer with osimertinib. Clinical Cancer Res, 24(11), 2594-2604. doi:10.1158/1078-0432.ccr-17.1875
  • 39. 39 Siddiqui, N., & Sonenberg, N. (2015). Signalling to eIF4E in cancer. Clinical Cancer Research, 43(5), 763–772. doi:10.1042/BST20150126 Singh, S., Ramamoorthy, M., Vaughan, C., Yeudall, W., Deb, S., & Palit, S. (2013). Human oncoprotein MDM2 activates the Akt signaling pathway through an interaction with the repressor element-1 silencing transcription factor conferring a survival advantage to cancer cells. Cell death and differentiation, 20(4), 558–566. doi:10.1038/cdd.2012.153 Smolle, E., & Pichler, M. (2019). Non-Smoking-Associated Lung Cancer: A distinct Entity in Terms of Tumor Biology, Patient Characteristics and Impact of Hereditary Cancer Predisposition. Cancers, 11(2), 204. doi.org/10.3390/cancers11020204 Sun, Y., Liu, W., Liu, T., Feng, X., Yang, N., & Zhou, F. (2015). Signalling pathway of MAPK/ERK in cell proliferation, differentiation, migration, senescence and apoptosis. Journal of Receptor Signal Transduction Research, 35(6), 600-604. doi:10.3109/10799893.2015.1030412 Sutherland, K. D., & Berns, A. (2010). Cell of origin of lung cancer. Molecular oncology, 4(5), 397– 403. doi:10.1016/j.molonc.2010.05.002 Swain, S., Kim, S-B., Jungsil, R., Vladimir, S., Campone, M., Ciruelos, E., …, & Ross, G. (2015). Pertuzumab, Trastuzumab and Docetaxel in HER2-positive metastatic breast cancer: CLEOPATRA. New England Journal of Medicine, 372, 724-734. doi:10.1015/NEJMoa1413513
  • 40. 40 Tang, Z., Zhang, J., & Lu, X. (2018). Coexistent genetic alterations involving ALK, RET, ROS1 or MET in 15 cases of lung adenocarcinoma. Mod Pathol, 31, 307–312. doi:10.1038/modpathol.2017.109 Tanos, T., Marinissen, J., Leskow, C., Hochbaum, D., Martinetto, H., Gutkind, S., & Coso, A. (2005). Phosphorylation of c-Fos by members of the p38 MAPK family: Role in the AP-1 response to UV light. Journal of Biol Chem, 280(19), 52. Dos:10.107jbc.m500620200 Thangavel, C., Boopathi, E., Liu, Y., McNair, C., Haber, A., Perepelyuk, M., … , & Den, R. (2018). Therapeutic Challenge with a CDK4/6 inhibitor induces an RB-dependent SMAC-mediated apoptotic response in non-small cell lung cancer. Clinical Cancer Research, 24(6). Doi:10.1158/1078-0432.CCR-17-2074 UK Office for National Statistics. (2017). Deaths registered in England and Wales 2017. Retrieved https://www.ons.gov.uk/peoplepopulationandcommunity/birthsdeathsandmarriages/deaths/bu lletins/deathsregisteredinenglandandwalesseriesdr/2017 UK Office for National Statistics. (2018). Cancer Registration statistics, England. Retrievedhttps://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/condi tionsanddiseases/bulletins/cancerregistrationstatisticsengland/final2016 UK Office for National Statistics. (2019). Cancer Registration statistics, England. Retrieved https://www.ons.gov.uk/peoplepopulationandcommunity/healthandsocialcare/conditionsandd iseases/datasets/cancerregistrationstatisticscancerregistrationstatisticsengland
  • 41. 41 Vallejo, A., Perurena, N., Guruceaga, E., Pawel, K., Zandueta, C., Valencia, K., …, & Vincent, S.(2017). An integrative approach unveils FOSL1 as an oncogene vulnerability in KRAS- driven lung and pancreatic cancer. Nature Communications, 8, 14294. doi:10.1038/ncomms14294 Veen, J., Scherzer, M., Boshuizen, J., Chu, M., Liu, A., Landman, A., …, & McMahon, M. (2019). Mutationally-activated PI3'-kinase-α promotes de-differentiation of lung tumors initiated by the BRAFV600E oncoprotein kinase. eLife, 8, e43668. doi:10.7554/eLife.43668 Wan, S., Wright, D., & Coveney, P. (2012). Mechanism of drug efficacy within the EGFR revealed by microsecond molecular dynamics simulation. Molecular Cancer Therapeutics, 11(11). Doi:10.1158/1535-7163.MCT-12-0644-T Wang, L., Haichuan, H., Yunjian, P., Rui, W., Li, Y., Shen, L., …, & Chen, H. (2014). PI3KCA mutations frequently coexist with EGFR/KRAS mutations in non-small cell lung cancer and suggest poor prognosis in EGFR/KRAS wildtype group. PLoS ONE, 9(2), e88291. doi:10.1371/journal.pone.0088291 Wilkerson, D., Yin, D., Walter, V., Zhao, N., Cabanski, C., Hayward, M., Miller, C., …, & Hayes, D. (2012). Differential pathogenesis of lung adenocarcinoma subtypes involving sequence mutations, copy number, chromosomal instability, and methylation. PloS one, 7(5), e36530. https://doi.org/10.1371/journal.pone.0036530
  • 42. 42 Wu, Y., Hung-Chang, W., Jia-En, W., Kuo, H., Shuenn, Y., Si-Xuan, C., …, & Hong, T. (2019). The dual PI3K/mTOR inhibitor BEZ235 restricts the growth of lung cancer tumors regardless of EGFR status, as a potent accompanist in combined therapeutic regimens. Journal of Experimental & Clinical Cancer Research, 38, 282. Doi:10.1186/s13046-019-1282-0 Yang, H., Liang, S., Schmid, R., & Peng, R-W. (2019). New horizons in KRAS-mutant Lung Cancer: Dawn after Darkness. Frontiers Oncology, 9(953). doi:10.3389/fonc.2019.00953 Yang, W., Meng, L., Chen, K., Tian, C., Peng, B., Zhong, L., …, & Li, L. (2019). Preclinical pharmacodynamic evaluation of a new Src/FOSL1 inhibitor, LY-1816, in pancreatic ductal adenocarcinoma. Cancer science, 110(4), 1408–1419. doi:10.1111/cas.13929 Yap, Y., McPherson, J., Ong, C., Rozen, S., Teh, B., Lee, A., & Callen, D. (2014). The NF1 gene revisited - from bench to bedside. Oncotarget, 5(15), 5873–5892. doi:10.18632/oncotarget.2194 Yatabe, Y., Borczuk, A., & Powell, C. (2011). Do all lung adenocarcinomas follow a stepwise progression?. Lung cancer, 74(1), 7–11. doi:10.1016/j.lungcan.2011.05.021 Yatabe, Y., Kosaka, T., Takahashi, T., & Mitsudomi, T. (2005). EGFR mutation is specific for terminal respiratory unit type adenocarcinoma. The American Journal of Surgical Pathology, 29(5), 633-639. Doi:10.1097/01.pas.00005156935.28066.35 Yousaf-Khan, U., Aalst, C., Jong, P., Heuvelmans, M., Scholten, E., & Lammers, J., …, Koning, H. (2017). Final screening round of the NELSON lung cancer screening trial: the effect of a 2.5- year screening interval. BMJ Thorax, 72(6-7), 48-56. Doi:10.1136/thoraxjnl-2016-209755
  • 43. 43 Zappa, C., & Mousa, S. (2016). Non-small cell lung cancer: current treatment and future advances. Translational lung cancer research, 5(3), 288–300. Doi:10.21037/tlcr.2016.06.07 Zappa, C., & Mousa, S. (2016). Non-small cell lung cancer: current treatment and future advances. Translational lung cancer research, 5(3), 288–300. doi:10.21037/tlcr.2016.06.07 Zhang, W-W., Li, L., Li, D., Liu, J., Li, X., Li, W., … , & Lam, M-K. (2017). The first approved gene therapy product for cancer Ad-p53 (Gendicine): 12 years in the clinic. Human Gene Therapy, 29(2). doi:10.1089/hum.2017.218 Zhang, Y., Chang, L., & Yang, Y. (2019). Intratumor heterogeneity comparison among different subtypes of non-small-cell lung cancer through multi-region tissue and matched ctDNA sequencing. Mol Cancer, 18, 7. doi:10.1186/s12943-019-0939-9 Zhao, H., Wang, J., & Tony, S. (2015). The phosphatidylinositol 3-kinase/Akt and c-Jun N-terminal kinase signaling in cancer: Alliance or contradiction?. International Journal of Oncology, 47, 429-436. doi:10.3892/ijo.2015.3052 Zippo, A., Serafini, R., Rocchigiani, M., Pennacchini, S., Krepelova, A., & Oliviero, S. (2009). Histone crosstalk between H3S10ph and H4K16ac generates a histone code that mediates transcription elongation. Cell, 138(6), 1122-1136. doi:10.1016/j,cell.2009.07.031