You are on page 1of 9

Lung Cancer 159 (2021) 1–9

Contents lists available at ScienceDirect

Lung Cancer
journal homepage: www.elsevier.com/locate/lungcan

A retrospective observational study of the natural history of advanced


non–small-cell lung cancer in patients with KRAS p.G12C mutated or
wild-type disease
Alexander I. Spira a, b, c, Huakang Tu d, Shivani Aggarwal d, Hil Hsu d, Gillis Carrigan d,
Xuena Wang e, Gataree Ngarmchamnanrith f, Victoria Chia d, Jhanelle E. Gray g, *
a
Virginia Cancer Specialists, 8503 Arlington Blvd Suite 400, Fairfax, VA, 22031, USA
b
US Oncology Research, The Woodlands, TX, USA
c
Johns Hopkins School of Medicine, Baltimore, MD, USA
d
Center for Observational Research, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA 91320-1799, USA
e
Global Biostatistical Science, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320-1799, USA
f
Clinical Development, Amgen Inc., One Amgen Center Drive, Thousand Oaks, CA, 91320-1799, USA
g
Department of Thoracic Oncology, Moffitt Cancer Center, 12902 Magnolia Dr, Tampa, FL, 33612, USA

A R T I C L E I N F O A B S T R A C T

Keywords: Introduction: The KRAS p.G12C mutation, prevalent in non–small-cell lung cancer (NSCLC), has only recently
Non–small-cell lung cancer become a viable target. Here we present results of the largest retrospective observational study analyzing KRAS
KRAS p.G12C p.G12C in patients with advanced NSCLC.
Retrospective
Materials and Methods: Adults with advanced NSCLC (All Advanced NSCLC cohort) and subcohorts with different
mutation profiles (KRAS p.G12C [G12C] and KRAS/EGFR/ALK wild type [Triple WT]) diagnosed January 2011
to March 2019 were selected from a US clinico-genomic database; treatment-related characteristics, molecular
profiles, real-world overall (rwOS) and progression-free survival (rwPFS) were analyzed.
Results: Demographics were similar across cohorts, with more smokers and nonsquamous cell carcinoma his­
tology in the G12C cohort. KRAS p.G12C was nearly mutually exclusive (≤1.2 %) with known actionable driver
mutations, but non-driver co-mutations were common (STK11, 21.5 %; KEAP1, 7.0 %; TP53, 48.0 %). Among
G12C patients, 20 % had no documentation of receiving systemic therapy. Across treated G12C patients, 67 %
received immune checkpoint inhibitors; first-line usage increased from 0% (2014) to 81 % (2019). Among G12C
patients, median (95 % CI) rwOS was 12.0 (9.6–15.3), 9.5 (8.1–13.1), and 6.7 (5.9–10.7) months after first,
second, and third line of therapy, respectively; median (95 % CI) rwPFS was 5.0 (4.4–5.8), 4.0 (2.8–5.3), and 3.1
(2.4–4.3) months. Outcomes for the G12C subcohort were similar to those for all patients (All Advanced NSCLC
cohort). Mutations in STK11/KEAP1 were associated with poorer survival across all cohorts.
Conclusion: The poor outcomes associated with KRAS p.G12C mutated advanced NSCLC indicate an unmet need
for more effective novel treatments.

Abbreviations: ALK, anaplastic lymphoma kinase; BRAF, B-Raf proto-oncogene; CGDB, clinico-genomic database; CGP, comprehensive genomic profiling; EGFR,
epidermal growth factor receptor; EHR, electronic health records; FH-FMI, Flatiron Health-Foundation Medicine; G12C, patients with KRAS p.G12C mutated NSCLC
included in study; KEAP1, Kelch-like ECH-associated protein 1 oncogene; KRAS, Kirsten rat sarcoma viral oncogene homolog; KRAS p.G12C, codon 12 glycine-to-
cysteine substitution of KRAS; MET, mesenchymal-epithelial transition; NGS, next-generation sequencing; NSCLC, non‒small-cell lung cancer; NTRK1‒3, neuro­
trophic tyrosine kinases 1–3 gene; PD-1, programmed death 1; PD-L1, programmed death ligand 1; RAS, rat sarcoma viral oncogene homolog; RECIST, Response
Evaluation Criteria in Solid Tumors; RET, rearranged-during-transfection gene; ROS1, ROS proto-oncogene 1; rwOS, real-world overall survival; rwPFS, real-world
progression-free survival; STK11/LKB1, serine/threonine kinase 11, liver kinase B1; TP53, tumor protein p53; Triple WT, KRAS/EGFR/ALK wild-type; VEGF, vascular
endothelial growth factor.
* Corresponding author.
E-mail addresses: Alexander.Spira@USOncology.com (A.I. Spira), htu01@amgen.com (H. Tu), shivania@amgen.com (S. Aggarwal), hhsu02@amgen.com
(H. Hsu), gcarriga@amgen.com (G. Carrigan), xuenaw@amgen.com (X. Wang), gatareen@amgen.com (G. Ngarmchamnanrith), vchia@amgen.com (V. Chia),
Jhanelle.gray@moffitt.org (J.E. Gray).

https://doi.org/10.1016/j.lungcan.2021.05.026
Received 15 April 2021; Received in revised form 18 May 2021; Accepted 22 May 2021
Available online 25 May 2021
0169-5002/© 2021 Published by Elsevier B.V.
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

1. Introduction comprehensive CGP of >300 cancer-related genes on FMI’s


next-generation sequencing (NGS) based FoundationOne® panel [23,
Lung cancer is the second most common cancer in both men and 24]. As of June 2020, over 400,000 samples had been sequenced from
women, as well as the leading cause of cancer death worldwide [1]. patients across the United States. A recent study using data from the
Non–small-cell lung cancer (NSCLC) comprises about 84 % of lung NSCLC cohort of the FH-FMI CGDB demonstrated that using a CGDB
cancer cases [1]. The majority of US patients with NSCLC are diagnosed derived from routine clinical care can be representative of the real-world
with advanced disease and have a poor prognosis [2]. For patients with patient population [25]. Furthermore, well-established genomic
advanced NSCLC and targetable driver mutations, targeted therapies markers in the CGDB correlate with clinical outcomes [25].
significantly improve treatment outcomes [3] and are the recommended
first-line therapies [4]; indeed, the benefit of targeted therapies in 2.3. Patients
earlier stage disease has also been demonstrated [5]. For patients with
advanced NSCLC without targetable driver mutations, immune check­ Patients were included in the study if they were diagnosed with
point inhibitors for programmed cell death-1 (PD-1) or programmed advanced NSCLC and were ≥18 years old at the time of diagnosis
death ligand-1 (PD-L1), used as a monotherapy or in combination with (N = 7069). Advanced NSCLC was defined as an initial diagnosis of
chemotherapy, are recommended as first-line treatment [4,6]. locally advanced (stage IIIB/IIIC) or metastatic (stage IV) disease, or an
Mutations in the rat sarcoma viral oncogene homolog (RAS) family of initial diagnosis of early-stage disease with subsequent recurrence or
proto-oncogenes are highly prevalent in NSCLC, with the Kirsten RAS progressive disease. Demographic and clinical characteristics including
(KRAS) homolog being the most frequently mutated isoform [7]. age, race, Eastern Cooperative Oncology Group performance status,
Approximately 90 % of KRAS mutations in NSCLC occur in codon 12 [8]. disease stage at initial diagnosis, metastatic sites, treatment patterns,
The most common codon 12 mutation is a single guanine-to-thymine and mutation profiles were collected from all patients in the database
substitution resulting in a glycine-to-cysteine substitution (KRAS p. meeting the inclusion criteria.
G12C) [7]. KRAS p.G12C occurs in approximately 13 % of NSCLC
adenocarcinoma cases and 41 % of KRAS mutated disease [9] and has
2.4. Molecular characteristics
been identified as a putative oncogenic driver in several types of solid
tumors including NSCLC [10]. With the recent development of KRAS p.
Mutation profiles were detected via FoundationOne tumor DNA
G12C inhibitors such as sotorasib [11,12] and adagrasib [13,14], the
sequencing (Foundation Medicine, Cambridge, MA, USA) as part of
KRAS p.G12C mutation is emerging as the next targetable driver mu­
routine, real-world care. The FoundationOne platform is used to detect
tation in NSCLC [15,16].
short-variant mutations, rearrangements, and copy number alterations
There is a lack of robust characterization of patients with KRAS p.
from cancer specimens and has been shown to have >95 % sensitivity
G12C mutated advanced NSCLC; few studies have described outcomes in
and >99 % positive predictive value [23]. Variants of known or likely
KRAS mutated NSCLC, and results have been inconsistent [17–20]. This
significance were considered to be positive.
is partly due to sparse information on large cohorts of patients with
NSCLC with comprehensive clinical and genomic alteration data and
partly due to the effect of co-mutations such as serine/threonine kinase 2.5. Treatment-related characteristics
11 (STK11/LKB1) and Kelch-like ECH-associated protein 1 oncogene
(KEAP1) [15]. The purpose of this study was to provide real-world ev­ Several treatment-related characteristics were collected, including
idence on clinico-pathologic and molecular characteristics, treatment information related to treatment patterns and lines of therapy. Available
patterns, and outcomes in patients with KRAS p.G12C mutated treatment history, including chemotherapies, targeted therapies, im­
advanced NSCLC (G12C cohort), as well as for patients with KRAS munotherapies, and other therapies, was collected from the EHRs of the
wild-type, epidermal growth factor receptor (EGFR) wild-type, and study center. Detailed information on start and end dates for each line of
anaplastic lymphoma kinase (ALK) wild-type advanced NSCLC (here­ therapy was collected, as were details of each drug regimen. The first
after referred to as Triple WT), and the overall cohort of patients with line of therapy was defined from the start of the first systemic therapy
advanced NSCLC (hereafter referred to as All Advanced NSCLC cohort) drug to the last administration of that systemic course of therapy or
using a de-identified nationwide (US-based) NSCLC clinico-genomic death. All subsequent lines of therapy were defined in a similar fashion.
database (CGDB).
2.6. Outcomes
2. Materials and methods
The endpoints of the study were real-world overall survival (rwOS)
2.1. Study design and real-world progression-free survival (rwPFS) by line of therapy. For
patients who received systemic treatment, rwOS was defined as time
An observational retrospective cohort study of patients with from start of treatment to death; rwPFS was defined as time from start of
advanced NSCLC diagnosed between January 1, 2011, and March 31, treatment to progression or death, whichever occurred first. For patients
2019, was undertaken to characterize the natural history of patients without any documentation of receiving systemic treatment, rwOS was
with advanced NSCLC. To ensure at least 6 months of follow-up for defined as time from advanced NSCLC diagnosis to death. The date of
outcomes, data were collected through September 30, 2019. death was computed using a composite real-world mortality endpoint
derived from EHR [26]. Patients who were lost to follow-up or followed
2.2. Data source up to the end of study period without an outcome were censored. Date of
death or progression was extracted from real-world data collected in the
This study used the nationwide (US-based) de-identified Flatiron EHR as part of routine clinical care, with the information about each
Health-Foundation Medicine NSCLC CGDB (FH-FMI CGDB). The de- progression event being retrospectively captured. Real-world disease
identified data originated from approximately 280 US cancer clinics progression was identified from clinic notes from visits during which a
(~800 sites of care) and excluded patients involved in clinical trials. The patient was evaluated for progression by the treating clinician and has
FH-FMI CGDB integrates FMI’s comprehensive genomic profiling (CGP) been shown to correlate with overall survival [27]. This approach is
results with de-identified patient-level structured and unstructured data conceptually different from prospective collection of progression data
derived from electronic health records (EHR) by de-identified deter­ within the context of a clinical study, commonly assessed using
ministic matching [21,22]. Genomic alterations were identified via Response Evaluation Criteria in Solid Tumors (RECIST) [28].

2
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

2.7. Statistical analyses and 3957 (56.0 %) were KRAS/EGFR/ALK wild type and included in the
Triple WT cohort.
All analyses were descriptive in nature, with no formal hypothesis Median age at advanced diagnosis was 68 years for the G12C cohort,
testing. Descriptive statistics (mean, median, standard deviation [SD], 69 years for the Triple WT cohort, and 68 years for the All Advanced
range) were presented for continuous variables (eg, age). For categorical NSCLC cohort. Female patients accounted for 61.1 % (n = 454/743) of
variables (eg, race, disease stage), the number and percentage of pa­ G12C patients, 42.1 % (n = 1665/3957) of Triple WT patients, and 50 %
tients in each group were reported. rwOS and rwPFS and corresponding (n = 3532/7069) of patients in the All Advanced NSCLC cohort. Overall,
95 % confidence intervals (CI) were calculated using Kaplan-Meier es­ ~82 % of patients were current or former smokers, with the percentage
timates. In real-world settings, patients may have NGS undertaken at the being higher in the G12C cohort (96.8 % [n = 719/743]). Furthermore,
time of their advanced NSCLC diagnosis and before starting initial most tumors were nonsquamous (76.1 % [n = 5382/7069] overall),
therapy, or after receiving their initial therapy, resulting in an immortal with a higher percentage of nonsquamous histology in the G12C cohort
time period (ie, time from the start of the line of therapy to the NGS test (90.8 % [n = 675/743]). In addition, 86.1 % (n = 640/743) of the G12C
where the patient would have been alive and not eligible for an cohort, 80.6 % (n = 3191/3957) of the Triple WT cohort, and 84.0 %
outcome). Immortal time bias was accounted for via cohort restriction (n = 5937/7069) of the All Advanced NSCLC cohort had evidence of
by selecting patients whose NGS reporting date was before or up to 21 stage IV disease at the time of their advanced NSCLC diagnosis (Table 1).
days after the start of line of therapy for outcome analyses by line of The pattern of distant metastases was generally consistent across all
therapy. For those who did not have any documentation of receiving cohorts; however, a slightly higher percentage of patients with brain
systemic treatment, rwOS and rwPFS were measured from advanced metastasis was found in the G12C cohort. Baseline characteristics for all
NSCLC diagnosis date, and a delayed entry model was used to account cohorts are summarized in Table 1; baseline characteristics for the
for immortal time bias. cohort of patients with a KRAS mutation that was not a KRAS p.G12C
mutation are summarized in Supplementary Table 1.
3. Results KRAS p.G12C was nearly mutually exclusive (≤1.2 %) with all
actionable driver mutations with likely or known significance in NSCLC,
3.1. Patient population such as ALK rearrangements, EGFR mutations, ROS proto-oncogene 1
(ROS1) rearrangements, B-Raf proto-oncogene (BRAF) mutations, neu­
There were 7069 patients who were diagnosed with advanced-stage rotrophic tyrosine kinases 1–3 gene (NTRK1–3) rearrangements,
NSCLC and had received molecular testing through the FMI NGS testing rearranged-during-transfection gene (RET) rearrangements, and
panel within the study time frame of January 1, 2011, to March 31, 2019 tyrosine-protein kinase gene mesenchymal-epithelial transition (MET)
(All Advanced NSCLC cohort). Among these patients, 743 (10.5 %) were short-variant alterations (Fig. 1; left). In comparison, the All Advanced
positive for the KRAS p.G12C mutation and included in the G12C cohort, NSCLC cohort exhibited a high prevalence of EGFR mutations (13.6 %),

Table 1
Baseline characteristics.
Characteristic G12C (n = 743) Triple WT (n = 3957) All Advanced NSCLC (n = 7069)

Age at advanced diagnosis, years, median (range) 68 (29–85) 69 (26–85) 68 (24–85)


Female, n (%) 454 (61.1) 1665 (42.1) 3532 (50.0)
Race, n (%)
Asian 7 (0.9) 71 (1.8) 223 (3.2)
Black 38 (5.1) 258 (6.5) 401 (5.7)
Hispanic or Latino 2 (0.3) 2 (0.1) 4 (0.1)
White 550 (74.0) 2800 (70.8) 4951 (70.0)
Other 80 (10.8) 489 (12.4) 882 (12.5)
Not available 66 (8.9) 337 (8.5) 608 (8.6)
Current or former smoker, n (%) 719 (96.8) 3430 (86.7) 5786 (81.9)
Histology of NSCLC, n (%)
Nonsquamous 675 (90.8) 2506 (63.3) 5382 (76.1)
Squamous 31 (4.2) 1252 (31.6) 1387 (19.6)
Not otherwise specified 37 (5.0) 199 (5.0) 300 (4.2)
Stage at initial diagnosis, n (%)
Stage ≤ IIIA 213 (28.7) 1078 (27.2) 1825 (25.8)
Stage IIIB–IVB 513 (69.0) 2776 (70.2) 5079 (71.8)
Not reported 17 (2.3) 103 (2.6) 165 (2.3)
Stage IV disease at index date, n (%)
Any metastatic disease 640 (86.1) 3191 (80.6) 5937 (84.0)
Bone 231 (31.1) 1079 (27.3) 2132 (30.2)
Brain 174 (23.4) 577 (14.6) 1243 (17.6)
Distant lymph node 73 (9.8) 461 (11.7) 787 (11.1)
Lung 206 (27.7) 1091 (27.6) 2063 (29.2)
Advanced disease diagnosed in 2015 or later, n (%)* 611 (82.2) 3307 (83.6) 5810 (82.2)
Practice type, n (%)
Academic 46 (6.2) 245 (6.2) 537 (7.6)
Community 697 (93.8) 3712 (93.8) 6532 (92.4)
Number of total lines of therapy in advanced setting, n (%)
0 149 (20.1) 681 (17.2) 1206 (17.1)
1 293 (39.4) 1381 (34.9) 2479 (35.1)
2 150 (20.2) 1015 (25.7) 1755 (24.8)
3 83 (11.2) 491 (12.4) 871 (12.3)
≥4 68 (9.2) 389 (9.8) 758 (10.7)

ALK=anaplastic lymphoma kinase; EGFR=epidermal growth factor receptor oncogene; KRAS=Kirsten rat sarcoma viral oncogene homolog; NSCLC = non–small-cell
lung cancer; Triple WT=KRAS/EGFR/ALK wild type.
*
Checkpoint inhibitor therapy gained its first approval in NSCLC in March 2015.

3
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

Fig. 1. Co-mutation patterns in the three


advanced NSCLC cohorts.* Left: Mutually
exclusive co-mutations. Right: Frequent co-
mutations with KRAS mutations.
*Percentages based on the number of patients
tested for the co-mutation. ALK=anaplastic
lymphoma kinase; BRAF=B-Raf proto-
oncogene; EGFR=epidermal growth factor re­
ceptor oncogene; KEAP1=Kelch-like ECH-
associated protein 1 gene; KRAS=Kirsten rat
sarcoma viral oncogene homolog;
MET = mesenchymal-epithelial transition;
NSCLC = non–small-cell lung cancer;
NTRK=neurotrophic tyrosine kinase gene;
RET=rearranged-during-transfection gene; ROS1=ROS proto-oncogene 1; STK11=serine-threonine kinase 11 gene; TP53=tumor protein p53 gene; Triple
WT=KRAS/EGFR/ALK wild type.

followed by BRAF mutations (4.2 %), ALK rearrangements (2.7 %), MET received systemic treatment, 67 % (n = 396/594) received PD-1/PD-L1
short-variant alterations (2.0 %), and RET (0.9 %), ROS1 (0.7 %), and inhibitors at some point in their treatment history. Year-on-year analysis
NTRK rearrangements (0.2 %). For non-actionable drivers, patients with demonstrates a steady increase in the use of PD-1/PD-L1 inhibitors from
the KRAS p.G12C mutation had a 21.5 % co-mutation rate of STK11 the time of first approval in 2014. None of the patients received first-line
(also known as LKB1) versus only 10.7 % for the Triple WT cohort and PD1/PD-L1 inhibitors in 2014, and only 7.5 % (n = 3/40) of patients in
12.4 % for the All Advanced NSCLC cohort. KEAP1 mutations were also the G12C cohort, 7.9 % (n = 10/126) of patients in the Triple WT
prevalent in the G12C cohort (7.0 %) and occurred at a similar per­ cohort, and 6.2 % (n = 17/275) in the All Advanced NSCLC cohort
centage in the other two cohorts (7.5 % for the Triple WT cohort, 6.4 % received first-line PD-1/PD-L1 inhibitors in 2015. In 2019, these
for the All Advanced NSCLC cohort). Lastly, tumor protein p53 gene numbers increased to 81.0 % (n = 17/21) of patients in the G12C
(TP53) short-variant alterations were present in approximately half of cohort, 74.4 % (n = 96/129) in the Triple WT cohort, and 64.6 %
patients in the G12C cohort (48.0 %), 73.1 % of the Triple WT cohort, (n = 157/243) in the All Advanced NSCLC cohort (Fig. 2).
and 63.2 % of the All Advanced NSCLC cohort (Fig. 1; right). Co-
mutation patterns for patients with KRAS mutations that were not
3.3. Outcomes
KRAS p.G12C are shown in Supplementary Fig. 1.
For the first line of therapy, median rwOS (95 % CI) was 12.0
3.2. Treatment characteristics (9.6–15.3) months for the G12C cohort, 10.8 (9.9–11.8) months for the
Triple WT cohort, and 12.9 (11.9–14.2) months for the All Advanced
A total of 594 (79.9 %) patients from the G12C cohort, 3276 (82.8 %) NSCLC cohort (Fig. 3A). For the second line of therapy, median rwOS
patients from the Triple WT cohort, and 5863 (82.9 %) from the All (95 % CI) was 9.5 (8.1–13.1) months for the G12C cohort, 8.8 (7.8–9.8)
Advanced NSCLC cohort had documentation of having received sys­ months for the Triple WT cohort, and 10.2 (9.5–11.3) months for the All
temic treatment after diagnosis. The main therapies used were platinum- Advanced NSCLC cohort. Kaplan-Meier estimates for rwOS for the first
based chemotherapy with or without a PD-1/PD-L1 inhibitor, PD-1/PD- and second line of therapy showed a similar survival probability across
L1 monotherapy, and other therapies, including targeted therapies such time for the three cohorts, with equally poor survival rates at the end of
as combinations of immune checkpoint inhibitors, single-agent chemo­ database tracking (Fig. 4A, B). Similar patterns can be observed for the
therapies (eg, docetaxel, gemcitabine, pemetrexed), and vascular third and fourth lines of therapy (Supplementary Fig. 2).
endothelial growth factor (VEGF) and VEGF receptor inhibitors (eg, The median rwPFS in the G12C cohort was similar to that in the
bevacizumab and ramucirumab). Of all patients in the G12C cohort who Triple WT and All Advanced NSCLC cohorts across all lines of therapy

Fig. 2. Use of PD-1/PD-L1 inhibitors as a first-line therapy by year of first-line initiation in each cohort. ALK=anaplastic lymphoma kinase; EGFR=epidermal growth
factor receptor oncogene; KRAS=Kirsten rat sarcoma viral oncogene homolog; NSCLC = non–small-cell lung cancer; PD-1/L1=programmed cell death-1/
programmed death ligand-1; Triple WT=KRAS/EGFR/ALK wild type.

4
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

Fig. 3. Median (95 % CI) (A) real-world overall survival (rwOS) and (B) real-world progression-free survival (rwPFS) for each line of treatment in each cohort.
ALK=anaplastic lymphoma kinase; EGFR=epidermal growth factor receptor oncogene; KRAS=Kirsten rat sarcoma viral oncogene homolog; NSCLC = non–small-cell
lung cancer; Triple WT=KRAS/EGFR/ALK wild type.

(Fig. 3B). For the first line of therapy, median rwPFS (95 % CI) was 5.0 (G12C cohort, n = 52, 9.0 [6.9–19.8] months; Triple WT, n = 114, 8.6
(4.4–5.8) months for the G12C cohort, 4.9 (4.5–5.2) months for the [5.2–10.0] months; All Advanced NSCLC, n = 252, 8.9 [7.3–10.2]
Triple WT cohort, and 5.6 (5.3–5.8) months for the All Advanced NSCLC months; Fig. 5A). For the second line of therapy, one of the lowest rwOS
cohort. For the second line, median rwPFS (95 % CI) was 4.0 (2.8–5.3) (95 % CI) was observed among patients in the G12C cohort who were
months for the G12C cohort, 3.5 (3.2–4.1) months for the Triple WT STK11 wild type and KEAP1 mutated (n = 5, 2.5 [1.0–14.9] months;
cohort, and 4.0 (3.7–4.4) months for the All Advanced NSCLC cohort. Supplementary Fig. 5A).
Kaplan-Meier estimates for rwPFS for the first and second line of therapy Median rwPFS (95 % CI) for the first line of therapy was slightly
show a similar PFS probability across time for the three cohorts. In all higher for patients in the G12C cohort wild type for STK11 and KEAP1
three cohorts, almost all patients show progression by the end of data­ (6.5 [5.1–8.4] months) than for patients in the Triple WT (5.1 [4.7–5.5]
base tracking (Fig. 4C, D). Findings were similar across cohorts for the months) and All Advanced NSCLC cohorts (6.1 [5.7–6.5] months) with
third and fourth lines of therapy (Supplementary Fig. 3). Median rwOS the same co-mutation status. Median rwPFS (95 % CI) was lower for
and rwPFS and Kaplan-Meier estimates across lines of therapy for pa­ G12C patients in the STK11 wild type and KEAP1 mutated subcohort
tients with KRAS mutations that were not KRAS p.G12C are shown in (2.5 [1.1–5.7] months) than for patients in the Triple WT (4.4 [2.8–5.8]
Supplementary Figs. 1 and 4. months) and All Advanced NSCLC cohorts (4.3 [2.8–5.6] months) with
Due to the high co-occurrence of KRAS p.G12C with STK11 and/or the same co-mutation status. In patients with STK11 mutated and KEAP1
KEAP1 mutations (Fig. 1), outcomes for all cohorts were also stratified wild type, median rwPFS was similar for patients in the G12C (4.1
by STK11/KEAP1 co-mutation status for the first and second lines of [3.4–4.8] months), Triple WT (3.9 [2.8–5.0] months), and All Advanced
therapy to assess for potential confounding (Fig. 5 and Supplementary NSCLC cohorts (4.0 [3.5–4.6] months; Fig. 5B). Similar patterns were
Fig. 5; outcomes for patients with KRAS mutations that were not KRAS p. observed for the second line of therapy (Supplementary Fig. 5B).
G12C stratified by STK11/KEAP1 co-mutation status are shown in
Supplementary Fig. 6). Overall, those wild type for both STK11 and 4. Discussion
KEAP1 had numerically higher rwOS and rwPFS among all cohorts for
first line therapy (Fig. 5) than those with STK11 or KEAP1 mutations. To our knowledge, this study uses the largest dataset to date con­
Although formal statistical comparisons were not undertaken, the taining information on outcomes and molecular characteristics of pa­
observed rwOS was slightly higher for patients in the G12C cohort tients with advanced NSCLC harboring the KRAS p.G12C mutation. With
(n = 197, 15.2 [12.0–18.3] months) than for patients in the Triple WT the possibility that the KRAS p.G12C mutation will become a targetable
(n = 1023, 11.7 [10.6–12.9] months) and All Advanced NSCLC cohorts driver mutation first in advanced NSCLC and then, if proven effective, in
(n = 1905, 14.8 [13.5–16.3] months) who were wild type for both earlier stage disease, we analyzed real-world evidence on clinical and
genes. For patients who were STK11 wild type and KEAP1 mutated, pathologic characteristics, treatment patterns, and outcomes in patients
median rwOS (95 % CI) was lower for patients in the G12C cohort with KRAS p.G12C mutation-positive advanced NSCLC (G12C cohort)
(n = 10, 4.5 [1.4–21.5] months) than for patients in the Triple WT from a de-identified nationwide, US-based NSCLC CGDB. Additionally,
(n = 68, 7.2 [5.8–18.4] months) and the All Advanced NSCLC cohorts these characteristics were also described in patients without the three
(n = 93, 7.1 [5.8–12.7] months). In patients who were STK11 mutated main actionable drivers (Triple WT cohort) and the overall cohort of
and KEAP1 wild type, median rwOS was similar among all cohorts patients with advanced NSCLC (All Advanced NSCLC cohort).

5
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

Fig. 4. Kaplan-Meier analyses for real-world overall survival (rwOS) after (A) first and (B) second lines of therapy, and for real-world progression-free survival
(rwPFS) after (C) first and (D) second lines of therapy. ALK=anaplastic lymphoma kinase; EGFR=epidermal growth factor receptor oncogene; KRAS=Kirsten rat
sarcoma viral oncogene homolog; NSCLC = non–small-cell lung cancer; Triple WT=KRAS/EGFR/ALK wild type.

Patients in the G12C cohort had demographic and clinical character­ The KRAS p.G12C mutation was nearly mutually exclusive (≤1.2 %)
istics similar to those in the overall group of patients with advanced with all other actionable driver mutations (EGFR and BRAF mutations;
NSCLC; however, higher proportions of past or present smokers and MET short-variant alterations; and ALK, ROS1, NTRK1–3, and RET
nonsquamous cell carcinoma histology were observed in the G12C cohort, rearrangements). This is consistent with the current understanding that
as has recently been reported based on data from the CRISP registry of KRAS p.G12C is mutually exclusive with other actionable mutations
German patients [29]. These findings are consistent with earlier studies in [35–38]. Therefore, patients with NSCLC harboring a KRAS p.G12C
smaller populations showing that KRAS mutations, specifically the p.G12C mutation are unlikely to be candidates for therapy targeted at these
mutation, are highly prevalent among current or former smokers, and that actionable driver mutations. As such, there is a need for additional
this mutation is associated with a high rate of brain metastases [18,20,30, novel, biomarker-driven, anticancer therapies with tolerable safety
31]. Similarly, the KRAS p.G12C mutation was found to be higher in profiles to address KRAS p.G12C mutated advanced NSCLC.
current and former smokers in a large European data set in patients with Furthermore, the KRAS p.G12C mutation was observed in the pres­
earlier stage disease (stage Ia to IIIb NSCLC) [32]. Other studies have also ence of high STK11 mutation levels in this study, as has recently been
demonstrated that nonsquamous cell carcinoma is the most common reported by others in a single treatment center [39]. STK11 mutations
histology type associated with KRAS mutations in general, as well as KRAS have been observed at a high rate in NSCLC and even higher in KRAS
p.G12C specifically [30,33]. Although KRAS mutations may be relatively mutated NSCLC; the KRAS p.G12C/STK11 co-mutation pattern has been
uncommon in some phenotypes, with a recent study finding a prevalence associated with lower overall survival and resistance to immune
of only 4.4 % in squamous cell carcinoma [33], KRAS p.G12C mutations checkpoint inhibitors [38,40–44]. KEAP1 mutations were prevalent in
can be found across patient types (eg, in nonsmokers [34]); thus, broad the G12C cohort, as well as the other cohorts in this study; although
molecular testing is needed to identify KRAS p.G12C status in all patients others have reported even higher levels of KEAP1 co-mutation [39].
with NSCLC. Pathogenic KEAP1 mutations are associated with chemoresistance in

6
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

Fig. 5. Median (95 % CI) (A) real-world overall


survival (rwOS) and (B) real-world progression-
free survival (rwPFS) for the first line of therapy
in subcohorts of patients based on STK11 and
KEAP1 co-mutation patterns. ALK=anaplastic
lymphoma kinase; EGFR=epidermal growth
factor receptor oncogene; KEAP1=Kelch-like
ECH-associated protein 1 gene; KRAS=Kirsten
rat sarcoma viral oncogene homolog;
NSCLC = non–small-cell lung cancer;
STK11=serine-threonine kinase 11 gene; Triple
WT=KRAS/EGFR/ALK wild type.

preclinical models of NSCLC, and lower overall survival in NSCLC, effective treatments for patients with KRAS p.G12C mutation in both
whereas patients with NSCLC harboring KEAP1 mutations are less sen­ early and advanced NSCLC. Furthermore, approximately 20 % of pa­
sitive to platinum-based treatments [45–50]. In our study, rwOS and tients in the G12C cohort in our study did not have any record of
rwPFS were generally longer for patients who had KRAS p.G12C receiving any systemic therapies, further highlighting the need for
mutated advanced NSCLC that was wild type for both STK11 and KEAP1. effective, safer, and more targeted therapies.
Therefore, the evidence points to a potential association of KRAS p.G12C To our knowledge, this is the largest real-world dataset to compre­
with other mutations that negatively affect survival outcomes in hensively characterize the clinico-pathologic and molecular characteris­
advanced NSCLC and may lead to unresponsiveness to immune check­ tics, treatment patterns, and outcomes in patients with KRAS p.G12C
point inhibitors [41]. mutated advanced NSCLC, compared with patients with Triple WT
Treatment patterns were generally similar among patients in the advanced NSCLC and All Advanced NSCLC cohorts. In addition, this study
G12C, Triple WT, and All Advanced NSCLC cohorts. The most common included patients from a large number of community oncology practices
regimens in the first and second lines of therapy after diagnosis of and academic centers across the United States, representing a true real-
advanced disease were platinum-based chemotherapy regimens and world population. Furthermore, the mortality data in the CGDB is well
regimens including immune checkpoint inhibitors; however, since their validated, with high sensitivity and specificity [26,51]. Finally, this study
approval in 2014, PD1/PD-L1 inhibitors are increasingly used, as has included comprehensive genomic profiling results with de-identified
been reported by others [29]. In our study, they were the most common clinical data derived from EHR, including key variables such as histol­
first-line treatment in the G12C, Triple WT, and All Advanced NSCLC ogy, dates of diagnosis, stage, treatment, and real-world progression.
cohorts. For example, 81 % of patients in 2019 in the G12C cohort This study has some limitations. First, the CGDB is limited to US pa­
received PD-1/PD-L1 inhibitors as part of their first-line therapy (Fig. 2). tients, primarily those in community health centers, and those who
Once patients, including those with KRAS p.G12C mutated NSCLC, received molecular testing through the FMI NGS testing panel. Thus, re­
progressed on PD-1/PD-L1 inhibitors and first-line chemotherapy, sults may not be generalizable to all patients with advanced NSCLC,
limited options are available [4]. particularly those treated in academic centers or in other countries, or
In this study, the Triple WT cohort was included to illustrate out­ patients who did not receive NGS testing as part of their care, or who were
comes in patients for whom there are no or few approved targeted identified using different testing methodologies, or those with earlier stage
therapies. In the real-world setting, patients in the G12C cohort were disease. Nonetheless, it has been demonstrated that information obtained
found to have poor survival outcomes after different lines of therapy, from the database is reflective of the real-world patient population [25].
including immune checkpoint inhibitors; their prognosis was as poor as Second, the definition of rwPFS used in this study may lead to potential
that observed in patients in the Triple WT and All Advanced NSCLC misclassification of outcomes because the information about progression
cohorts. This similarity in outcomes is consistent with other studies is collected retrospectively rather than prospectively based on RECIST
showing that in Western populations, the KRAS p.G12C mutation is not criteria and relied on interpretation by individual physicians without
predictive of outcomes in patients with advanced NSCLC [17–20,39]. In standardization. In addition, the subgroups analyzed in this study have
earlier stage NSCLC, KRAS p.G12C mutations were associated with a relatively small sample sizes, particularly subgroups with STK11 and/or
trend toward shorter overall survival than those without KRAS muta­ KEAP1 co-mutations; therefore, results should be interpreted with caution.
tions [32]. Taken together, these findings reflect an unmet need for Finally, there are some limitations associated with most retrospective

7
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

observational studies, such as the possibility of unmeasured confounding Validation, Visualization, Writing - original draft, Writing - review &
or information bias [52] and potential missing data. editing. Shivani Aggarwal: Data curation, Formal analysis, Funding
acquisition, Investigation, Methodology, Software, Visualization,
5. Conclusions Writing - review & editing. Hil Hsu: Data curation, Formal analysis,
Funding acquisition, Investigation, Methodology, Software, Visuali­
This analysis of clinico-pathologic and molecular characteristics, zation, Writing - review & editing. Gillis Carrigan: Data curation,
treatment patterns, and outcomes of advanced NSCLC from a de- Formal analysis, Funding acquisition, Investigation, Methodology,
identified nationwide (US-based) NSCLC CGDB helps increase under­ Software, Visualization, Writing - review & editing. Xuena Wang:
standing of KRAS p.G12C mutation-positive advanced NSCLC. A higher Data curation, Formal analysis, Funding acquisition, Investigation,
percentage of patients with KRAS p.G12C mutation-positive advanced Methodology, Software, Visualization, Writing - review & editing.
NSCLC were current or former smokers and had nonsquamous histology. Gataree Ngarmchamnanrith: Data curation, Formal analysis, Fund­
The KRAS p.G12C mutation was virtually mutually exclusive with ing acquisition, Investigation, Methodology, Software, Visualization,
actionable driver mutations; however, co-mutation with STK11 and Writing - review & editing. Victoria Chia: Conceptualization, Data
KEAP1 occurred in approximately 20 % and 7% patients with KRAS p. curation, Formal analysis, Funding acquisition, Investigation, Meth­
G12C mutation-positive advanced NSCLC, respectively. Approximately odology, Resources, Software, Supervision, Validation, Visualization,
20 % of patients KRAS p.G12C mutation-positive advanced NSCLC had Writing - review & editing. Jhanelle E. Gray: Conceptualization, Data
no documented history of receiving systemic treatment; the majority of curation, Formal analysis, Investigation, Methodology, Project
those who received systemic treatment received PD-1/PD-L1 inhibitors. administration, Resources, Software, Supervision, Validation, Visual­
The poor rwOS and rwPFS in patients with KRAS p.G12C mutation- ization, Writing - original draft, Writing - review & editing.
positive advanced NSCLC from a de-identified nationwide (US-based)
NSCLC CGDB are consistent with outcomes from other real-world evi­ Acknowledgments
dence studies and indicate an unmet need for more effective and safer
novel treatment options in patients with this type of advanced NSCLC. The authors thank Yang Li, PhD (Amgen Inc., Thousand Oaks, CA)
and Vicky Kanta, PhD and Lee Hohaia, PharmD (ICON, North Wales,
Funding statement PA), whose work was funded by Amgen Inc., for medical writing assis­
tance in the preparation of this manuscript.
This work was funded by Amgen Inc. Except for the role of Amgen
personnel as authors as identified or as outlined in the acknowledg­ Appendix A. Supplementary data
ments, Amgen Inc. had no involvement in the study design, collection,
analysis and interpretation of data, the writing of this report, or the Supplementary material related to this article can be found, in the
decision to publish. online version, at doi:https://doi.org/10.1016/j.lungcan.2021.05.026.

Disclosures References

Alexander I. Spira reports grants from Amgen and personal fees from [1] American Cancer Society, About Lung Cancer, 2020 (accessed October 20 2020),
https://www.cancer.org/cancer/lung-cancer/about.html.
AstraZeneca, Bristol Myers Squibb, Mirati, Novartis, and Sanofi. Hua­ [2] R.L. Siegel, K.D. Miller, A. Jemal, Cancer statistics, 2020, CA Cancer J. Clin. 70
kang Tu, Shivani Aggarwal, Gillis Carrigan, Xuena Wang, Gataree (2020) 7–30.
Ngarmchamnanrith, and Victoria Chia report employment by and stock [3] N. Duma, R. Santana-Davila, J.R. Molina, Non-small cell lung cancer:
epidemiology, screening, diagnosis, and treatment, Mayo Clin. Proc. 94 (2019)
ownership of Amgen Inc. Hil Hsu reports employment by Skye Biologics 1623–1640.
and Amgen and stock ownership of Amgen Inc. Jhanelle E. Gray reports [4] National Comprehensive Cancer Network, Clinical Practice Guidelines in Oncology
personal fees from AstraZeneca, Blueprint Medicines, Bristol Myers (NCCN Guidelines): Non-Small Cell Lung Cancer (Version 8.2020), National
Comprehensive Cancer Network, 2020 (accessed February 17 2021), https://www.
Squibb, EMD Serono – Merck KGaA, Inivata, Merck, Novartis, and nccn.org/store/login/login.aspx?ReturnURL=https://www.nccn.org/professiona
Janssen Scientific Affairs, LLC; research support from AstraZeneca, ls/physician_gls/pdf/nscl.pdf.
Boehringer Ingelheim, Bristol Myers Squibb, Genentech, G 1 Thera­ [5] Y.L. Wu, M. Tsuboi, J. He, T. John, C. Grohe, M. Majem, et al., Osimertinib in
resected EGFR-Mutated non-small-Cell lung Cancer, N. Engl. J. Med. 383 (2020)
peutics, Merck, Novartis, Pfizer, and Ludwig Institute of Cancer
1711–1723.
Research; and unpaid consultancy for Daiichi Sankyo, Inc. [6] D. Planchard, S. Popat, K. Kerr, S. Novello, E.F. Smit, C. Faivre-Finn, et al.,
Metastatic non-small cell lung cancer: ESMO Clinical Practice Guidelines for
diagnosis, treatment and follow-up, Ann. Oncol. 29 (2018) iv192–iv237.
Data sharing
[7] I.A. Prior, P.D. Lewis, C. Mattos, A comprehensive survey of Ras mutations in
cancer, Cancer Res. 72 (2012) 2457–2467.
Qualified researchers may request data from Amgen clinical studies. [8] R.P. Jones, P.A. Sutton, J.P. Evans, R. Clifford, A. McAvoy, J. Lewis, et al., Specific
Complete details are available at the following: http://www.amgen.com mutations in KRAS codon 12 are associated with worse overall survival in patients
with advanced and recurrent colorectal cancer, Br. J. Cancer 116 (2017) 923–929.
/datasharing. [9] A. Biernacka, P.D. Tsongalis, J.D. Peterson, F.B. de Abreu, C.C. Black, E.
J. Gutmann, et al., The potential utility of re-mining results of somatic mutation
Transparency document testing: KRAS status in lung adenocarcinoma, Cancer Genet. 209 (2016) 195–198.
[10] A. Fernández-Medarde, E. Santos, Ras in cancer and developmental diseases, Genes
Cancer 2 (2011) 344–358.
The Transparency document associated with this article can be found [11] J. Canon, K. Rex, A.Y. Saiki, C. Mohr, K. Cooke, D. Bagal, et al., The clinical KRAS
in the online version. (G12C) inhibitor AMG 510 drives anti-tumour immunity, Nature 575 (2019)
217–223.
[12] D.S. Hong, M.G. Fakih, J.H. Strickler, J. Desai, G.A. Durm, G.I. Shapiro, et al.,
CRediT authorship contribution statement KRASG12C inhibition with sotorasib in advanced solid tumors, N. Engl. J. Med. 383
(2020) 1207–1217.
[13] J. Hallin, L.D. Engstrom, L. Hargis, A. Calinisan, R. Aranda, D.M. Briere, et al., The
Alexander I. Spira: Conceptualization, Data curation, Formal KRASG12C inhibitor MRTX849 provides insight toward therapeutic susceptibility of
analysis, Investigation, Methodology, Project administration, Re­ KRAS-mutant cancers in mouse models and patients, Cancer Discov. 10 (2020)
sources, Software, Supervision, Validation, Visualization, Writing - 54–71.
[14] Another KRAS inhibitor holds its own, Cancer Discov. 10 (2020), https://doi.org/
original draft, Writing - review & editing. Huakang Tu: Conceptual­
10.1158/2159-8290.Cd-nb2020-1098.
ization, Data curation, Formal analysis, Funding acquisition, Investi­
gation, Methodology, Project administration, Resources, Software,

8
A.I. Spira et al. Lung Cancer 159 (2021) 1–9

[15] T.F. Burns, H. Borghaei, S.S. Ramalingam, T.S. Mok, S. Peters, Targeting KRAS- [33] S.V. Liu, A.M. Vanderwalde, H. Mamdani, L.E. Raez, Y. Baca, J. Xiu, et al.,
mutant non-small-cell lung cancer: one mutation at a time, with a focus on KRAS Characterization of KRAS mutations (mt) in non-small cell lung cancer (NSCLC,
G12C mutations, J. Clin. Oncol. 38 (2020) 4208–4218. J. Clin. Oncol. 38 (2020), 9544-9544.
[16] D. Kim, J.Y. Xue, P. Lito, Targeting KRAS(G12C): from inhibitory mechanism to [34] M.L. Forsythe, A. Alwithenani, D. Bethune, M. Castonguay, A. Drucker,
modulation of antitumor effects in patients, Cell 183 (2020) 850–859. G. Flowerdew, et al., Molecular profiling of non-small cell lung cancer, PLoS One
[17] W. Cui, F. Franchini, M. Alexander, A. Officer, H.L. Wong, M.J. IJzerman, et al., 15 (2020), e0236580.
Assessing the significance of KRAS G12C mutation: clinicopathologic features, [35] R. Scharpf, G. Riely, M. Awad, M. Lenoue-Newton, B. Ricciuti, J. Rudolph, et al.,
treatments, and survival outcomes in a real-world KRAS mutant non-small cell lung Abstract 1095: comprehensive pan-cancer analyses of RAS genomic diversity,
cancer cohort, J. Clin. Oncol. 38 (2020), https://doi.org/10.1200/ Cancer Res. 80 (2020), 1095-1095.
JCO.2020.1238.1215_suppl.e19324. [36] P. Martín Martorell, M. Huerta, A. Compañ Quilis, R. Abellán, E. Seda, S. Blesa, et
[18] W. Cui, F. Franchini, M. Alexander, A. Officer, H.L. Wong, I.J. M, et al., Real world al., Coexistence of EGFR, KRAS, BRAF, and PIK3CA mutations and ALK
outcomes in KRAS G12C mutation positive non-small cell lung cancer, Lung Cancer rearrangement in a comprehensive cohort of 326 consecutive Spanish
146 (2020) 310–317. nonsquamous NSCLC patients, Clin. Lung Cancer 18 (2017) e395–e402.
[19] F. Griesinger, W.E.E. Eberhardt, P. Hoffknecht, M. Metzenmacher, T. Wehler, [37] J.F. Gainor, A.M. Varghese, S.H. Ou, S. Kabraji, M.M. Awad, R. Katayama, et al.,
K. Kokowski, et al., 1364P Treatment and outcome of a real-world cohort of ALK rearrangements are mutually exclusive with mutations in EGFR or KRAS: an
patients with advanced, non-squamous NSCLC and KRAS mutations with a special analysis of 1,683 patients with non-small cell lung cancer, Clin. Cancer Res. 19
focus on KRAS G12C, Ann. Oncol. 31 (suppl 4) (2020) S872. (2013) 4273–4281.
[20] K.C. Arbour, H. Rizvi, A.J. Plodkowski, D. Halpenny, M.D. Hellmann, G. Heller, et [38] M. Scheffler, M.A. Ihle, R. Hein, S. Merkelbach-Bruse, A.H. Scheel, J. Siemanowski,
al., Clinical characteristics and anti-PD-(L)1 treatment outcomes of KRAS-G12C et al., K-ras mutation subtypes in NSCLC and associated co-occuring mutations in
mutant lung cancer compared to other molecular subtypes of KRAS-mutant lung other oncogenic pathways, J. Thorac. Oncol. 14 (2019) 606–616.
cancer, J. Clin. Oncol. 38 (15 suppl) (2020), https://doi.org/10.1200/ [39] K.C. Arbour, H. Rizvi, A.J. Plodkowski, M.D. Hellmann, A. Knezevic, G. Heller, et
JCO.2020.1238.1215_suppl.9596. al., Treatment outcomes and clinical characteristics of patients with KRAS-G12C
[21] B. Birnbaum, N. Nussbaum, K. Seidl-Rathkopf, M. Agrawal, M. Estevez, E. Estola, et mutant non-small cell lung Cancer, Clin. Cancer Res. (2021).
al., Model-assisted Cohort Selection With Bias Analysis for Generating Large-scale [40] B. Ricciuti, K.C. Arbour, J.J. Lin, N. Vokes, A.V. Hoojghan, Y.Y. Li, et al., Effect of
Cohorts From the EHR for Oncology Research, 2020 (accessed February 18 2021), STK11 mutations on efficacy of PD-1 inhibition in non-small cell lung cancer
https://arxiv.org/ftp/arxiv/papers/2001/2001.09765.pdf. (NSCLC) and dependence on KRAS mutation status, J. Clin. Oncol. 38 (2020)
[22] X. Ma, L. Long, S. Moon, B.J.S. Adamson, S.S. Baxi, Comparison of population e15113-e15113.
characteristics in real-world clinical oncology databases in the US: flatiron Health, [41] A. Pavan, E. Zulato, L. Calvetti, A. Ferro, G. Nardo, A. Boscolo, et al., Plasma next-
SEER, and NPCR, medRxiv (2020), https://doi.org/10.1101/ generation sequencing (NGS) in advanced non-small cell lung cancer (aNSCLC)
2020.1103.1116.20037143. patients (pts) treated with immune checkpoint inhibitors (ICIs): impact of STK11
[23] G.M. Frampton, A. Fichtenholtz, G.A. Otto, K. Wang, S.R. Downing, J. He, et al., and TP53 mutations on outcome, J. Clin. Oncol. 38 (2020), 3046-3046.
Development and validation of a clinical cancer genomic profiling test based on [42] Y. Tamiya, Y. Zenke, S. Matsumoto, N. Furuya, T. Sakamoto, T. Kato, et al.,
massively parallel DNA sequencing, Nat. Biotechnol. 31 (2013) 1023–1031. Therapeutic impact of mutation subtypes and concomitant STK11 mutations in
[24] FoundationOne® CDx: Technical Information, Foundation Medicine, Inc., KRAS–mutated non-small cell lung cancer (NSCLC): a result of nationwide genomic
Cambridge, MA, 2020. screening project (LC-SCRUM-Japan), J. Clin. Oncol. 38 (2020), 9589-9589.
[25] G. Singal, P.G. Miller, V. Agarwala, G. Li, G. Kaushik, D. Backenroth, et al., [43] J. An, M. Yan, N. Yu, A. Chennamadhavuni, M. Furqan, T. Kruser, et al., Outcomes
Association of patient characteristics and tumor genomics with clinical outcomes of patients with stage III non-small cell lung cancer (NSCLC) that harbor a STK11
among patients with non-small cell lung cancer using a clinicogenomic database, mutation, J. Clin. Oncol. 38 (2020), 9033-9033.
JAMA 321 (2019) 1391–1399. [44] R. Uba, L.E. Raez, K. Dumais, F. Gentile, H.W. Powery, G.C. Domingo, et al.,
[26] M.D. Curtis, S.D. Griffith, M. Tucker, M.D. Taylor, W.B. Capra, G. Carrigan, et al., Serine/threonine kinase 11 (STK11) mutations and immunotherapy resistance in
Development and validation of a high-quality composite real-world mortality patients with non-small cell lung cancer, J. Clin. Oncol. 38 (2020) e15055-e15055.
endpoint, Health Serv. Res. 53 (2018) 4460–4476. [45] Y. Jeong, J.A. Hellyer, H. Stehr, N.T. Hoang, X. Niu, M. Das, et al., Role of KEAP1/
[27] S.D. Griffith, R.A. Miksad, G. Calkins, P. You, N.G. Lipitz, A.B. Bourla, et al., NFE2L2 mutations in the chemotherapeutic response of patients with non-small
Characterizing the feasibility and performance of real-world tumor progression end cell lung cancer, Clin. Cancer Res. 26 (2020) 274–281.
points and their association with overall survival in a large advanced non–small- [46] Y. Tian, K. Wu, Q. Liu, N. Han, L. Zhang, Q. Chu, et al., Modification of platinum
cell lung cancer data set, Jco Clin. Cancer Inform. (2019) 1–13. sensitivity by KEAP1/NRF2 signals in non-small cell lung cancer, J. Hematol.
[28] J.E. Dancey, L.E. Dodd, R. Ford, R. Kaplan, M. Mooney, L. Rubinstein, et al., Oncol. 9 (2016) 83.
Recommendations for the assessment of progression in randomised cancer [47] L.M. Solis, C. Behrens, W. Dong, M. Suraokar, N.C. Ozburn, C.A. Moran, et al., Nrf2
treatment trials, Eur. J. Cancer 45 (2009) 281–289. and KEAP1 abnormalities in non-small cell lung carcinoma and association with
[29] M. Sebastian, W.E.E. Eberhardt, P. Hoffknecht, M. Metzenmacher, T. Wehler, clinicopathologic features, Clin. Cancer Res. 16 (2010) 3743–3753.
K. Kokowski, et al., KRAS G12C-mutated advanced non-small cell lung cancer: a [48] K.C. Arbour, E. Jordan, H.R. Kim, J. Dienstag, H.A. Yu, F. Sanchez-Vega, et al.,
real-world cohort from the German prospective, observational, nation-wide CRISP Effects of co-occurring genomic alterations on outcomes in patients with KRAS-
Registry (AIO-TRK-0315), Lung Cancer (2021). In press. mutant non-small cell lung cancer, Clin. Cancer Res. 24 (2018) 334–340.
[30] A. Ghimessy, P. Radeczky, V. Laszlo, B. Hegedus, F. Renyi-Vamos, J. Fillinger, et [49] F. Goeman, F. De Nicola, S. Scalera, F. Sperati, E. Gallo, L. Ciuffreda, et al.,
al., Current therapy of KRAS-mutant lung cancer, Cancer Metastasis Rev. 39 (2020) Mutations in the KEAP1-NFE2L2 pathway define a molecular subset of rapidly
1159–1177. progressing lung adenocarcinoma, J. Thorac. Oncol. 14 (2019) 1924–1934.
[31] S. Dogan, R. Shen, D.C. Ang, M.L. Johnson, S.P. D’Angelo, P.K. Paik, et al., [50] C.A. Wohlhieter, A.L. Richards, F. Uddin, C.H. Hulton, À. Quintanal-Villalonga,
Molecular epidemiology of EGFR and KRAS mutations in 3,026 lung A. Martin, et al., Concurrent mutations in STK11 and KEAP1 promote ferroptosis
adenocarcinomas: higher susceptibility of women to smoking-related KRAS-mutant protection and SCD1 dependence in lung cancer, Cell Rep. 33 (2020), 108444.
cancers, Clin. Cancer Res. 18 (2012) 6169–6177. [51] Q. Zhang, A. Gossai, S. Monroe, N.C. Nussbaum, C.M. Parrinello, Abstract 5772:
[32] S.P. Finn, A. Addeo, U. Dafni, E. Thunnissen, L. Bubendorf, L.B. Madsen, et al., validation analysis of a composite real-world mortality endpoint for US cancer
Prognostic impact of KRAS G12C mutation in patients with NSCLC: results from the patients, Cancer Res. 80 (2020), 5772-5772.
European Thoracic Oncology Platform Lungscape Project, J. Thorac. Oncol. [52] E.J. Boyko, Observational research–opportunities and limitations, J. Diabetes
(2021). February 26) [Epub ahead of print]. Complications 27 (2013) 642–648.

You might also like