Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.

MCT-16-0793

Models and Technologies Molecular


Cancer
Therapeutics
Metformin Inhibits Cellular Proliferation and
Bioenergetics in Colorectal Cancer Patient–
Derived Xenografts
Nur-Afidah Mohamed Suhaimi1, Wai Min Phyo1, Hao Yun Yap1, Sharon Heng Yee Choy2,
Xiaona Wei1, Yukti Choudhury1, Wai Jin Tan1, Luke Anthony Peng Yee Tan1,
Roger Sik Yin Foo3,4, Suzanne Hui San Tan3, Zenia Tiang3, Chin Fong Wong5,
Poh Koon Koh6, and Min-Han Tan1,6,7

Abstract
There is increasing preclinical evidence suggesting that met- growth inhibition. Ex vivo culture of organoids generated from
formin, an antidiabetic drug, has anticancer properties against PDX demonstrates that metformin inhibits growth by executing
various malignancies, including colorectal cancer. However, the metabolic changes to decrease oxygen consumption and acti-
majority of evidence, which was derived from cancer cell lines vating AMPK-mediated pathways. In addition, we also per-
and xenografts, was likely to overestimate the benefit of met- formed genetic characterizations of serial PDX samples with
formin because these models are inadequate and require supra- corresponding parental tissues from patients using next-gener-
physiologic levels of metformin. Here, we generated patient- ation sequencing (NGS). Our pilot NGS study demonstrates
derived xenograft (PDX) lines from 2 colorectal cancer patients that PDX represents a useful platform for analysis in cancer
to assess the properties of metformin and 5-fluorouracil (5-FU), research because it demonstrates high fidelity with parental
the first-line drug treatment for colorectal cancer. Metformin tumor. Furthermore, NGS analysis of PDX may be useful to
(150 mg/kg) as a single agent inhibits the growth of both PDX determine genetic identifiers of drug response. This is the first
tumors by at least 50% (P < 0.05) when administered orally for preclinical study using PDX and PDX-derived organoids
24 days. In one of the PDX models, metformin given concur- to investigate the efficacy of metformin in colorectal cancer.
rently with 5-FU (25 mg/kg) leads to an 85% (P ¼ 0.054) Mol Cancer Ther; 16(9); 2035–44. 2017 AACR.

Introduction which patient-derived tumor fragments are directly implanted


into immunocompromised mice through serial passaging, are an
Anticancer drug development is often impeded by a lack of
increasingly recognized tool for drug candidate evaluation. These
suitable preclinical models that are highly predictive of therapeu-
PDXs are unlikely to have undergone extensive selection pressure
tic efficacy in humans. Most studies utilizing cell lines and
typically associated with cell line establishment as manifested
xenografts are useful in the laboratory setting, but may not
through genetic and molecular changes (3), and are thus valuable
translate well to the clinical setting (1). The failure of many cell
for drug studies.
line xenografts to accurately predict the clinical efficacy of anti-
We evaluated the performance of 5-fluorouracil (5-FU), the
cancer agents is due to their inability to reflect the complexity and
first-line treatment for colorectal cancer (6), along with the
heterogeneity of human tumors (2). Substantial genetic diver-
antidiabetic drug metformin. Epidemiologic studies have sug-
gence exists between primary tumor and the corresponding cell
gested the beneficial effects of metformin in cancer treatment
line derived from that tumor, as compared with a direct transplant
among diabetic patients (7, 8). This was supported by preclin-
of tumor into mice (3–5). Patient-derived xenografts (PDX), in
ical studies, which demonstrate that metformin has antineo-
plastic activity in breast, colon, lung, and prostate cancer
(9–14), spurring significant interest to repurpose metformin
1
Institute of Bioengineering and Nanotechnology Singapore, Singapore. 2Bio- as an anticancer agent. Nonetheless, preclinical studies often
logical Resource Centre Singapore, Singapore. 3Genome Institute of Singapore, involve supraphysiologic concentrations of metformin that are
Singapore. 4Cardiovascular Research Institute, National University Health Sys- in excess of the therapeutic levels achieved in patients. In
tems, Singapore. 5Tan Tock Seng Hospital Singapore, Singapore. 6Concord humans, the initial dose of metformin is 500 to 1,000 mg/
Cancer Hospital Singapore, Singapore. 7National Cancer Centre Singapore, daily, and subsequently, a maintenance dose of 2,000 mg/daily
Singapore.
is required. Studies performed on cell lines require elevated
Note: Supplementary data for this article are available at Molecular Cancer doses of metformin (2–50 mmol/L) to elicit cellular response,
Therapeutics Online (http://mct.aacrjournals.org/).
while in vivo studies require up to 4 times the maximum clinical
Corresponding Author: Min-Han Tan, Institute of Bioengineering and Nano- dose in humans of 2,550 mg/day (15). Thus, it is anticipated
technology, 31 Biopolis Way, The Nanos #09-01, Singapore 138669. Phone: 65- that good relevant models are necessary for physiologic model-
68247110; Fax: 65-64789080; E-mail: mhtan@ibn.a-star.edu.sg
ing of metformin.
doi: 10.1158/1535-7163.MCT-16-0793 To the best of our knowledge, this is the first study evaluating
2017 American Association for Cancer Research. the use of metformin on colorectal cancer PDX, a highly

www.aacrjournals.org 2035

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Mohamed Suhaimi et al.

relevant system for drug candidate testing. Using next-genera- Tumor growth inhibition (TGI) was calculated as shown: TGI ¼
tion sequencing (NGS), we interrogated the molecular profiles [(tumor volume of control on day 24  tumor volume of treated
of these PDX to evaluate selection pressures initiated during the on day 24)/(tumor volume of control on day 24  tumor volume
course of drug treatment. In addition, we aimed to identify of control on day 0)]  100.
molecular signatures that influence response to metformin. In At endpoint, tumors were harvested. A portion of each tumor
parallel, we utilized colorectal cancer organoids to elucidate the was fixed for histologic sections and hematoxylin and eosin
mechanistic effect of metformin. (H&E) staining, while the remaining portions were snap-frozen
or used for organoids derivation.
Materials and Methods
Organoids derivation and cellular respiration assay
Cell culture maintenance, proliferation, and clonogenic assays Tumors from control mice were used for ex vivo measurement of
Cell lines were obtained in 2012 from ATCC directly while cellular respiration. After removal of necrotic tissue, tumors were
isogenic lines HCT116 p53þ/þ (wild-type, WT) and p53/ washed in HBSS, minced and digested in digestion media (RPMI
(knockout, KO) were kindly provided by Dr. Bert Vogelstein media with 5X Penicillin-Streptomycin-Amphotericin B, 10
(John Hopkins University, MD, USA). Independent authentica- mmol/L HEPES, 300 U/mL Collagenase, 100 U/mL Hyaluroni-
tion of cells was not performed. Cells were cultured in DMEM with dase, and 10 mmol/L Y-27632) for 2 hours at 37 C. Digestion
10% fetal bovine serum and maintained in 37 C incubator media were removed and digested tissues were resuspended in
supplemented with 5% CO2. The MycoAlert Mycoplasma Detec- culture media (Advanced DMEM/F12 with 5 Penicillin–Strep-
tion Kit (Lonza) was used to test for Mycoplasma contamination at tomycin–Amphotericin B, 1 Glutamax, 1 B27, 1 N-2,
defined intervals, and the tests indicate that cell lines were free of 1 mmol/L N-Acetylcysteine, 50 ng/mL rhEGF, and 10 mmol/L
Mycoplasma throughout the study. Y-27632), and filtered through 100- and 40-mm filter. Organoids
The CellTiter96 AQueous One Solution Assay (Promega) was were collected from the 40- to 100-mm fraction, and the numbers
used to assess proliferation of cells incubated with metformin were estimated.
(0–20 mmol/L) and/or 5-FU (10 mmol/L) in the presence of Oxygen consumption rate (OCR) of organoids were measured
high (4,500 mg/L) or low (1,000 mg/L) glucose. For clonogenic using Seahorse Bioscience XF96 analyzer as described previously
assay, cells harvested from exponential phase cultures were (16). Five hundred organoids were seeded overnight in a 96-well
plated in six-well plates. Seeding densities varied from 100 to cartridge and pretreated with 0.1 mmol/L or 1.0 mmol/L met-
300 cells per well. After 7 days, cells were washed with PBS, formin for 24 hours. Before measurement, the cells were washed
fixed with methanol and stained with Coomassie blue. Assays and medium was replaced. Reagents from the Cell Mito Stress Kit
were done in triplicate. Colonies were counted by two inde- (Seahorse Bioscience) were injected into each well sequentially
pendent investigators. and serve as control.
Microsatellite instability (MSI) analysis
MSI was determined using the MSI Analysis Kit version 1.2 Next-generation sequencing
(Promega). Briefly, DNA from matched normal and tumor A customized Ion AmpliSeqColon Panel was designed to
specimens was amplified using a panel of markers designed amplify 513 regions covering 40 genes implicated in colorectal
to amplify targeted microsatellite regions (BAT-25, BAT-26, cancer (Supplementary Table S1). Briefly, the customized panel
NR-21, NR-24, and MONO-27). Fragment analysis was per- was designed based on the multidimensional analysis of genomic
formed to assess the size of microsatellite repeats and deter- profiles in colorectal cancer published by The Cancer Genome
mine MSI status. Atlas Network (17) and molecular subtype signatures (18–20).
Hotspot mutations in genes of interest and other genes related to
Development of PDX probable actionable drug targets were identified and ranked. Ten
Fresh colorectal tumors were obtained from consenting nanograms of DNA was used to generate libraries using the Ion
patients at Concord Cancer Hospital in accordance with pro- Ampliseq library preparation kit v2.0. The barcoded libraries were
tocols approved by the Institutional Review Board. Tumors diluted to 7 pmol/L for template preparation using the OneTouch
were transplanted into severe combined immunodeficient 2 instrument and Ion PGMTemplate OT2 200 Kit. The resulting
(SCID) female mice aged 4 to 8 weeks (InVivos Singapore) pooled libraries were quality control checked using the Ion Sphere
under the approved protocol by the Institutional Animal Care quality control kit, and were then sequenced on the Ion Torrent
and Use Committee. Tumor specimens were cut into 2 to 4 PGM using a PGM 200 sequencing kit v2 and 318 Chip v2 (Life
mm3 pieces and implanted subcutaneously in the left flank of Technologies).
the mice. PDX was maintained by passaging into subsequent
generations when tumor volumes reached approximately 1,000 NGS variant analysis
to 1,500 mm3. For treatment studies, tumors were expanded in Point mutations were identified with the Torrent Suite Software
the left flanks of 5 to 12 mice (at least 5 evaluable tumors per v3.0 and the Ion Torrent Variant Caller v4.0 Plugin using the
arm). Mice were grouped into three arms: (i) control, (ii) somatic high stringency parameters and the targeted and hotspot
metformin, or (iii) metformin and 5-FU when tumor volumes pipelines. A 5% allele frequency threshold and 500 minimum
reached 100 to 200 mm3. Mice were treated daily with met- coverage was set to report de novo mutations. All the variants
formin dissolved in drinking water (150 mg/kg, orally) and/or identified were established by visualizing the data through IGV
weekly with 5-FU (25 mg/kg, intraperitoneally) for 24 days. 2.3 (Broad Institute). Concurrently, orthogonal verification of
Mice were monitored daily for signs of toxicity and tumor size variant calls made was performed using the iCMDB (Vishuo
was evaluated twice weekly by caliper measurements using the Biomedical) platform which provides variant annotation, as well
following formula: tumor volume in mm3 ¼ [length  width2]/2. as Sanger sequencing for variant validation.

2036 Mol Cancer Ther; 16(9) September 2017 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Metformin Inhibits Growth of Colorectal Cancer PDX

Western blotting Table 1. p53 and microsatellite status of cell lines and PDX
One hundred milligrams of snap-frozen tissues was lysed in Sample p53 status MSI/MSS
RIPA buffer containing protease and phosphatase inhibitors. DLD-1 Mutant MSI
Protein extracts (50 mg) were electrophoresed on 4% to 12% HCT116 Wild-type MSS
SK-CO-1 Wild-type MSS
Bis-Tris gels and transferred to PVDF membranes. Membranes SW-480 Mutant MSS
were blocked with 5% bovine serum albumin in Tris-buffered PDX FIT-CRC-086 Mutant MSS
saline and incubated overnight at 4 C with 1:1,000 dilutions of PDX FIT-CRC-104 Wild-type MSI
anti-phospho-mTOR (Ser2448), anti-mTOR, anti-phospho-
p70S6K (Thr389), anti-p70S6K, anti-phospho-AMPKa (Thr172),
and anti-AMPK antibodies (Thermofisher). Membranes were then PDX generation
washed and incubated with a 1:2,000 dilution of horseradish Two primary colorectal cancer tumors, FIT-CRC-086 and
peroxidase-conjugated goat anti-rabbit secondary antibody. FIT-CRC-104, with different clinicopathologic characteristics
Immunoreactive bands were detected by chemiluminescence (Table 2), were engrafted into 4 to 6 weeks old female SCID
using the ECL Prime Western Blotting Kit (GE Healthcare). mice. Growth rates ranged from 8 to 15 weeks for the initial
Intensity of each immunoreactive band was quantified by den- transplantation to reach a tumor volume of 1,000 to 1,500
sitometry using Image J software, and expressed relative to the mm3, and 3 to 8 weeks for subsequent passages. Histopathol-
control mice after normalization to ß-actin. ogy of patient's primary tumors and various xenograft passages
showed that the PDX models retained the original morphology
of the human primary tumor (Fig. 2A).
Statistical analysis
Sample size determination for in vivo studies was performed
Non-differential inhibition of metformin on PDX
using preliminary data generated from cell line xenografts a priori.
To evaluate tumor response in vivo, we utilized two PDX models
Briefly, the number of samples required per treatment group is 4,
with differing p53 and MSI/MSS status (Table 1). Mice treated with
at 95% confidence level and 80% power, for a desired effect size of
metformin had a lower tumor burden as compared with control
at least 1,000 mm3. For all experiments, Mann–Whitney and
group, regardless of p53 nor microsatellite status (Fig. 2B–D).
Kruskal–Wallis tests were used to determine the differences
Metformin was able to slow tumor growth initially, but at 12 days
between two groups, or three and more groups, respectively. P
posttreatment initiation, PDX tumors eventually progressed. After
values <0.05 were recognized as statistically significant.
24 days of drug treatment, the average tumor volume of the
metformin-treated group was reduced by 50% (P ¼ 0.056) and
Results 65% (P < 0.05) for p53-wild-type and p53-mutant PDX, respec-
Growth inhibition of 5-FU and metformin on cancer cell lines tively. A systemic effect of metformin on cell growth was observed
Metformin has been reported to preferentially inhibit the in the PDXs independent of tumor p53 status, with more pro-
growth of p53-mutant cells by forcing a metabolic burden that nounced growth inhibition being observed in the PDXs with
result in selective toxicity (9, 21, 22). However, recent evidence mutant-p53.
suggests that metformin requires p53 activity for metformin- When metformin was administered concurrently with a clin-
induced growth inhibition (23, 24). To investigate whether met- ically acceptable dose of 5-FU, only an additional 4% (P ¼ 0.767)
formin affects tumor proliferation in a p53-dependent manner, we tumor inhibition was observed in PDX-FIT-CRC-104; in contrast,
screened a panel of colorectal cancer cell lines with known p53 a pronounced tumor growth inhibition of 85% (P ¼ 0.054) was
statuses (Table 1), including a pair of isogenic cell lines HCT-116 observed in PDX-FIT-CRC-086.
(p53-WT and p53-KO). Increasing concentration of 5-FU at micro-
molar levels results in a growth inhibition of all colorectal cancer Metformin activates the AMPK pathway
cells (Fig. 1A) while supraphysiologic concentrations of metfor- To determine whether metformin influenced the AMPK and
min were required to achieve 50% cell growth inhibition (relative mTOR signaling pathway in the PDX materials, we evaluated the
to untreated cells) at 48 hours (Fig. 1B). phosphorylation of AMPK, mTOR, and p70S6K (Fig. 2E). Met-
Under glucose-deprived conditions, the degree of growth inhi- formin treatment activated AMPK as determined by phosphory-
bition by metformin is higher in colorectal cancer lines (Fig. 1C). lation at Thr172, with a corresponding inhibition of mTOR
There was no differential growth inhibition in response to 5-FU signaling and downstream target phosphorylation of p70S6K. In
(Fig. 1D) and metformin between the paired isogenic p53 lines tumors treated with metformin and 5-FU, AMPK was activated;
(P ¼ 0.206) as was previously reported, although cells with p53- interestingly, mTOR signaling appears to be activated and not
WT were inhibited to a greater extent under low glucose condi- suppressed, although phosphorylation of p70S6K was inhibited
tions (P ¼ 0.06; Fig. 1E). The clonogenic ability of p53-WT and (Fig. 2F).
p53-KO cells were similar upon metformin treatment (P ¼ 0.743);
nonetheless, cell growth for both lines were severely impaired Metformin inhibits oxygen consumption rate (OCR)
under glucose-deprived conditions (Fig. 1C and D). For ex vivo measurement of oxygen consumption, organoids
Approximately 15% of colorectal cancers display microsatellite derived from PDX were treated with metformin (Fig. 3A). Drugs
instability and are classified as microsatellite instable (MSI) or affecting mitochondrial functions were used in parallel to dem-
microsatellite stable (MSS). Characterization of colorectal cancer onstrate normal mitochondrial function. Metformin inhibited
cells for MSI/MSS status indicates that there was no difference in OCR of both FIT-CRC-086 and FIT-CRC-104 (Fig. 3B and C,
response to metformin between MSI and MSS cell lines. Essen- respectively) in a concentration-dependent manner. Consequent
tially, our results indicate that there is non-differential growth injection of oligomycin did not suppress the OCR further in these
inhibition to metformin, regardless of microsatellite or p53 status. metformin-treated organoids, while injection of FCCP, the

www.aacrjournals.org Mol Cancer Ther; 16(9) September 2017 2037

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Mohamed Suhaimi et al.

Figure 1.
In vitro effects of 5-fluorouracil (5-FU) and metformin
on colorectal cancer cells. 5-FU, the first-line
treatment for colorectal cancer, inhibits cell
proliferation at micromolar levels (A). Millimolar levels
of metformin are required to inhibit cell proliferation
at 48 hours, with pronounced inhibition observed in
combination with 10 mmol/L 5-FU (B). Glucose
deprivation accentuates growth inhibition by
metformin (C). Inhibition of colorectal cancer cell
proliferation to 5-FU (D) and metformin is
independent of p53 status, in both HCT116 p53
knockout (p53 KO) and HCT116 p53 wild-type (p53
WT) cell lines, but with stronger degree of growth
inhibition observed under glucose-deprived
conditions in the presence of metformin (E). The
absolute number of colonies is presented as an
average of triplicates (F).

2038 Mol Cancer Ther; 16(9) September 2017 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Metformin Inhibits Growth of Colorectal Cancer PDX

Table 2. Clinicopathologic information of patients with xenografts generated


Tumor Tumor Tumor Tumor size AJCC Lymph node
Patient Histology site grade type (largest dimension in cm) stage pT pN pM invasion
FIT-CRC-086 Adenocarcinoma Rectum 1 Ulcerated 4 IVA 3 0 1a 0/7
FIT-CRC-104 Mucinous adenocarcinoma Ascending colon 2 Localized 5 IIA 3 0 0 0/39

mitochondrial uncoupler, rescues the respiration inhibition, albe- indicates that there are no key colorectal cancer genes that are
it to a lesser extent in the metformin-treated organoids as com- associated with metformin's activity.
pared with control.
Correspondingly, metformin inhibition on cellular respiration
resulted in an increase in glycolysis, as measured by the extracel- Discussion
lular acidification rate (ECAR; Fig. 3D and E). It was evident that Metformin has been extensively reported as a promising anti-
metformin inhibits mitochondrial function as early as 4 hours cancer agent in cancer prevention and therapy (9–12, 21, 22,
(Fig. 3F and G). 28, 29). In the present study, we evaluated the effects of metfor-
min using preclinical models. Similar to reported studies, we
observed that the antiproliferative properties of metformin man-
Molecular profiling of circulating biomarkers in mice
ifest at supraphysiologic conditions in cell lines (30). A recurrent
To determine if tumor burden in vivo can be assessed using
feature is that these cells are typically maintained in non-phys-
alternative approaches that are viable in the clinical setting, we
iologic conditions that are optimized only for in vitro growth and
investigate the use of cfDNA in our models (25). CfDNA is a
proliferation. The excessive concentration of growth factors, insu-
heterogeneous mixture of normal and tumor-associated cell
lin and glucose may account for the elevated doses of metformin
populations (26, 27) which makes cfDNA analysis technically
required to elicit cellular responses and trigger metabolic stress. As
challenging. In xenografts, both human and mouse-derived cell
such, it is virtually impossible to translate these supraphysiologic
populations are present, and the ability to distinguish these two
conditions in the clinical setting, especially when the pharmaco-
cell populations is critical in order to assess the PDX response to
kinetics of the drug is different in culture settings and in the
treatment using cfDNA. Here, using a limited amount of
human body.
cfDNA, our assay can distinguish human and mice-derived
Consequently, we observed that low glucose conditions accen-
DNA efficiently (Supplementary Fig. S1), with limit of detec-
tuated the growth inhibition by metformin. This is not surpris-
tion of 10% human DNA in a background of mice DNA. No
ing—glucose deprivation is a distinctive feature of the tumor
cfDNA was detected at week 0, at the point of tumor implan-
microenvironment caused by the imbalance between nutrient
tation. Subsequent evaluation of the cfDNA at different time
supply and oxygen consumption rate (31). In a study exploring
points in the course of treatment indicates that human-derived
the role of the tumor microenvironment in potentiating the effect
DNA could be detected in the mice blood, suggesting that
of metformin, metformin is synthetically lethal with glucose
cfDNA could be a viable noninvasive biomarker for tumor
withdrawal in cancer cells (32). Essentially, this suggests that the
burden.
clinically irrelevant, supraphysiologic levels needed to observe
metformin's anticancer effects as reported by many in vitro studies
Mutation profile of primary tumor and corresponding merely underlie the artefactual experimental conditions that
xenografts poorly reflect actual glucose-starved in vivo conditions.
Human samples and PDX were analyzed for variants/muta- Most preclinical in vivo models have generally proven to be sub-
tions by NGS using a panel of 40 genes commonly implicated in optimal for directing clinical application of new anticancer ther-
colorectal cancer. The mean sequencing depth across all experi- apies, largely due to their inability to reflect the complexity and
ments was 1,385X for FIT-CRC-086 and 2,534X for FIT-CRC-104. heterogeneity of human tumors. PDX, where surgically resected
A summary of annotated variants is shown in Fig. 4A. tumor samples are engrafted directly into immunocompromised
For FIT-CRC-086, mutations in APC (K1350 ), KRAS (G12V), mice, offer several advantages (33, 34). In our study, we have
TP53 (R248Q), and SMAD4 (L495R) were observed for parental shown that PDX tumors maintained the molecular, genetic, and
tumor and xenografts, but were not present in germline or normal histologic heterogeneity typical of tumors of origin (35–39). PDX
colon. Likewise, in FIT-CRC-104, mutations were observed in provides an excellent platform to study cancer biology, analyze
KRAS (A146T) and MLH1 (R100 ) in parental tumor. Essentially, cancer therapeutics, and novel drug combinations. In our study,
mutations remain conserved across PDX passages. An example of we demonstrated metformin efficacy in our PDX models. To the
a variant conserved in tumor and PDX as depicted by NGS are best of our knowledge, ours represents the first study reported for
corroborated by Sanger sequencing in Fig. 4B. Interestingly, we colorectal cancer PDX using metformin. Administering metfor-
also observed the presence of variants that are not reported in min at physiologic levels of 150 mg/kg per day in our mice, which
COSMIC database. Using Sanger sequencing, we verified the non- is equivalent to initial clinical dose of 500 to 1,000 mg/daily in
COSMIC variants that were detected (Fig. 4C). Nonetheless, non- human, is sufficient to inhibit tumor growth in PDX. Furthermore,
COSMIC variants were not present in every PDX. Within the same we observed that the response to metformin may be independent
treatment subset, individuals exhibit heterogeneity. Whether of p53 status and is inconsistent with the observations reported
these new variants were a function of clonal selection of a subset earlier (9, 24). Conversely, studies conducted in breast cancer
of tumor cells warrants further investigation. indicate that metformin's effects are independent of p53 status
One of the key objectives of performing NGS is to identify (40, 41). This contraindication suggests that p53 may not be a
biomarkers that may reflect response to metformin. Our analysis reliable companion biomarker to predict response to metformin.

www.aacrjournals.org Mol Cancer Ther; 16(9) September 2017 2039

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Mohamed Suhaimi et al.

Figure 2.
Histomorphologic features of parental tumor are retained in patient-derived xenografts after serial passaging. Representative tumor cells are indicated by solid
arrows; mucins are indicated by diamond arrowheads (A). Gross morphology of PDX-FIT-CRC-086 tumors treated with metformin and 5-FU (B). Tumor growth
profile of PDX-FIT-CRC-086 (C) and PDX-FIT-CRC-104 (D). Metformin triggers AMPK activation and inhibits mTOR signaling in PDX (E). Protein levels were
normalized to ß-actin and fold changes are quantified relative to vehicle control (F).

Studies from a recent phase II clinical trial evaluating metfor- refractory colorectal cancer cases and that the benefits may be
min and 5-FU in patients with refractory colorectal cancer showed significant in nonrefractory colorectal cancer cases.
only a mild median progression-free survival (PFS) of 1.8 months Interestingly, in our study, we observed that combination of
and median overall survival (OS) of 7.9 months, with a majority 5-FU with metformin leads to a pronounced growth inhibition in
of patients (78%) having disease progression before 8 weeks (42). the p53-mutant PDX but not in p53-wild-type PDX. It may be
For 11 out of 50 patients (22%) having stable disease at the end of possible that p53 status plays a role in differential response to
8 weeks, their median PFS and OS were 5.6 months and 16.2 metformin and 5-FU; however, it is also likely that 5-FU is non-
months, respectively. Likely, the modest benefits of metformin beneficial in the p53-wild-type PDX because this PDX has MSI.
and 5-FU observed in this trial may be due to the nature of the Indeed, clinical observations have suggested that patients having a

2040 Mol Cancer Ther; 16(9) September 2017 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Metformin Inhibits Growth of Colorectal Cancer PDX

OXYGEN CONSUMPTION RATE OXYGEN CONSUMPTION RATE

B OCR
73
OCR vs TIME (Avg)

OLIGOMYCIN 1.0 µmol/L FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L


C OCR
417 OLIGOMYCIN 1.0 µmol/L
OCR vs TIME (Avg)

FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L

500 ORGANOIDS CONTROL (24 HRS) 500 ORGANOIDS CONTROL (24 HRS)
64 500 ORGANOIDS + 1.0 METFORMIN (24 HRS) 370 500 ORGANOIDS + 1.0 METFORMIN (24 HRS)
500 ORGANOIDS + 0.1 METFORMIN (24 HRS) 500 ORGANOIDS + 0.1 METFORMIN (24 HRS)

56 322

48 275
OCR pMoles/min

OCR pMoles/min
39 227

31 180

22 132
Control
14 85
0.1 mmol/L Met
1.0 mmol/L Met Control
5 37 0.1 mmol/L Met
1.0 mmol/L Met
–3 –10

–11 –58
0 15 30 45 60 75 91 106 121 136 151 0 15 30 45 60 75 90 105 120 135 151
TIME (min) TIME (min)

OXYGEN CONSUMPTION RATE OXYGEN CONSUMPTION RATE

D ECAR
ECAR vs TIME (Avg)
E ECAR
46
ECAR vs TIME (Avg)

30 OLIGOMYCIN 1.0 µmol/L FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L OLIGOMYCIN 1.0 µmol/L FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L
500 ORGANOIDS CONTROL (24 HRS)
500 ORGANOIDS CONTROL (24 HRS)
27 500 ORGANOIDS + 1.0 METFORMIN (24 HRS) 41 500 ORGANOIDS + 1.0 METFORMIN (24 HRS)
500 ORGANOIDS + 0.1 METFORMIN (24 HRS)
500 ORGANOIDS + 0.1 METFORMIN (24 HRS)

23 35

20 Control 30
1.0 mmol/L Met Control
ECAR mpH/min

24
ECAR mpH/min

16
0.1 mmol/L Met 0.1 mmol/L Met
13 19 1.0 mmol/L Met

9 13

6 8

2 2

–1 –3

–5 –9
0 15 30 45 60 75 91 106 121 136 151 0 15 30 45 60 75 90 105 120 135 151
TIME (min) TIME (min)

OXYGEN CONSUMPTION RATE OXYGEN CONSUMPTION RATE

F OCR
73
OCR vs TIME (Avg)
G OCR
196
OCR vs TIME (Avg)

OLIGOMYCIN 1.0 µmol/L FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L OLIGOMYCIN 1.0 µmol/L FCCP 1.0 µmol/L ROT\AA 0.5 µmol/L
500 ORGANOIDS CONTROL (4 HRS)
500 ORGANOIDS CONTROL (4 HRS)
64 500 ORGANOIDS + 1 METFORMIN (4 HRS) 174 500 ORGANOIDS + 1 METFORMIN (4 HRS)
500 ORGANOIDS + 0.1 METFORMIN (4 HRS)
500 ORGANOIDS + 0.1 METFORMIN (4 HRS)

56 152

48 130
OCR pMoles/min

OCR pMoles/min

39 108

31 86

22 64

14 Control 41 Control
1.0 mmol/L Met 0.1 mmol/L Met
5 0.1 mmol/L Met 19
1.0 mmol/L Met

–3 –3

–11 –25
0 15 30 45 60 75 91 106 121 136 151 0 15 30 45 60 75 90 105 120 135 151
TIME (min) TIME (min)

Figure 3.
Organoids derived from PDX under 10 (left image) and 40 (right image) (A). Pretreatment with 0.1 mmol/L metformin or 1.0 mmol/L metformin for 24 hours
inhibits oxygen consumption rate (OCR) relative to vehicle control for both FIT-CRC-086 (B) and FIT-CRC-104 (C), with corresponding increase in glycolysis
for FIT-CRC-086 (D) and FIT-CRC-104 (E). Pretreatment with 0.1 mmol/L metformin and 1.0 mmol/L metformin inhibits OCR as early as 4 hours in both
FIT-CRC-086 (F) and FIT-CRC-104 (G). 1.0 mmol/L oligomycin, 1.0 mmol/L carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP), and 0.5 mmol/L
rotenone/antimycin A were used as mitochondrial stress controls.

www.aacrjournals.org Mol Cancer Ther; 16(9) September 2017 2041

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Mohamed Suhaimi et al.

Figure 4.
Molecular characterization of germline DNA, parental tumor, serial passages of xenografts and xenografts treated with drugs. Variant allelic frequency is
reflected on the scale of 0 to 1 (A). An example of a COSMIC variant detected by next-generation sequencing (NGS) and independently verified by Sanger sequencing
(B), two variants that have not been reported in COSMIC are detected by NGS and Sanger sequencing (C).

deficient mismatch repair system or MSI do not benefit from ticated micro-imaging techniques are required to visualize the
5-FU–based adjuvant therapy in colorectal cancer (43, 44). Thus, tumor. In anticipation of this limitation (should the need arise
our observations using PDX are in line with observations to monitor tumor burden in orthotopic models), we assessed
obtained in the clinic, thus, emphasizing the crucial role PDXs noninvasive circulating biomarkers (cfDNA) in our subcutane-
play as a clinically relevant model. ous PDX models. Using established techniques that we have
To demonstrate the integrity of PDX as suitable models, we utilized previously for clinical cancer patients (48, 49), we
performed high-throughput NGS to characterize the genetic pro- demonstrated that it is feasible to prove the presence of
files of parental tumor and PDX. We demonstrate that genes such human-derived DNA circulating in plasma.
as TP53, APC, KRAS, and SMAD4, were mutated in the parental Utilizing NGS approach could aid in predictive biomarker(s)
tumor and conserved throughout serial passaging of the xeno- identification as a result of drug selection. Because this is a pilot
grafts. More importantly, in an effort to characterize biomarkers study involving metformin and PDX, banking on molecular
that can serve as companion marker to predict metformin profiles derived from 2 colorectal cancer patients may be inade-
response, we compared the genetic differences between the two quate for biomarker discovery process. Expanding the repertoire
PDX lines. Our NGS analysis suggests that there is lack of novel of PDX lines with different genetic make-up is likely to shed more
actionable genes that may reflect metformin's activity based on information in future. While the creation of PDX is of clear interest
genotype. Nonetheless, the key limitation in this particular anal- as a means to better define optimal therapy for colorectal cancer, it
ysis is likely due to limited variation in genotype between the two is undeniable that the high-throughput data are challenging to
PDX lines. manage and visualize. Thoughtful interpretation is necessary to
To gain insight into possible mechanisms of metformin convert the knowledge derived from existing data in order to
action, we evaluated the frequently implicated AMPK signaling understand treatment outcomes.
pathway (45). Indeed, our data showed that metformin trig- In essence, our study demonstrated the utility of PDX in
gered phosphorylation of Thr172 on AMPKa, which conse- assessing drug efficacy via molecular analysis, metabolic profiling,
quently led to downregulation of mTOR and its target p70S6K, and bioinformatics approaches. This is the first study that
whose phosphorylation at Thr389 is necessary for protein describes the use of metformin in colorectal cancer PDX and
synthesis (46). Moreover, we observed that metformin inhibits organoids, providing direct evidence of metformin as an antican-
oxygen consumption in organoids derived from PDX, as early cer agent in colorectal cancer.
as 4 hours, with a concomitant increase in glycolysis. Taken
together, it is likely that treatment with metformin activates the Disclosure of Potential Conflicts of Interest
AMPK pathway, which alters the mTOR signaling, and executes M.-H. Tan has ownership interest in a patent filed for method for KRAS
a metabolic change in the cells, which ultimately affects cell mutation detection, owned by his employer, and licensed to industry. No
growth in the PDX. potential conflicts of interest were disclosed.
We acknowledged that there are several limitations to our
current study. Firstly, we restricted the use of PDX to subcuta- Authors' Contributions
neous tumor models. Subcutaneous models are inferior to Conception and design: N.-A. Mohamed Suhaimi, W.M. Phyo, H.Y. Yap,
orthotopic models because the former do not represent appro- M.-H. Tan
Development of methodology: N.-A. Mohamed Suhaimi, W.M. Phyo, W.J. Tan
priate sites for human tumors, and may not be accurately
Acquisition of data (provided animals, acquired and managed patients,
predictive for drug evaluation (47). However, orthotopic tumor provided facilities, etc.): N.-A. Mohamed Suhaimi, W.M. Phyo, H.Y. Yap,
models are time-consuming and technically challenging. Fur- S.H.Y. Choy, Y. Choudhury, W.J. Tan, L.A.P.Y. Tan, R.S.Y. Foo, S.H.S. Tan,
thermore, it is difficult to monitor tumor growth, and sophis- P.K. Koh, M.-H. Tan

2042 Mol Cancer Ther; 16(9) September 2017 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Metformin Inhibits Growth of Colorectal Cancer PDX

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, like to thank Chit Fang Cheok for the helpful discussion on the in vitro work and
computational analysis): N.-A. Mohamed Suhaimi, W.M. Phyo, X. Wei, Shengyong Ng for the assistance in organoids derivation and culture.
L.A.P.Y. Tan, M.-H. Tan
Writing, review, and/or revision of the manuscript: N.-A. Mohamed Suhaimi,
W.M. Phyo, Y. Choudhury, W.J. Tan, L.A.P.Y. Tan, P.K. Koh, M.-H. Tan Grant Support
Administrative, technical, or material support (i.e., reporting or organizing This work is supported by the Institute of Bioengineering and Nanotech-
data, constructing databases): W.J. Tan, L.A.P.Y. Tan, Z. Tiang, C.F. Wong, nology and Agency for Science, Technology and Research, Singapore. M.H. Tan
M.-H. Tan received grants from Biomedical Research Council (Diagnostics Grant & Stra-
Study supervision: M.-H. Tan tegic Positioning Fund SPF 2012/003 and SPF2015/002).
The costs of publication of this article were defrayed in part by the
Acknowledgments payment of page charges. This article must therefore be hereby marked
We would like to thank Concord Cancer Hospital for the provision of the advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate
tissue samples, and Vishuo Biomedical for the support in the bioinformatics this fact.
analysis. We would like to thank Chua Teck Chiew Timothy, Ma Janise Ann
Kabiling Lalic, Mukta Pathak, and Hannes Martin Hentze from A STAR Bio- Received November 22, 2016; revised March 29, 2017; accepted May 17,
logical Resource Centre for their help and support in PDX work. We would also 2017; published OnlineFirst May 22, 2017.

References
1. Voskoglou-Nomikos T, Pater JL, Seymour L. Clinical predictive value of the 17. Muzny DM, Bainbridge MN, Chang K, Dinh HH, Drummond JA., Fowler
in vitro cell line, human xenograft, and mouse allograft preclinical cancer G, et al. Comprehensive molecular characterization of human colon and
models. Clin Cancer Res 2003;9:4227–39. rectal cancer. Nature 2012;487:330–7.

2. Ostman A, Augsten M. Cancer-associated fibroblasts and tumor growth – 18. De Sousa E Melo F, Wang X, Jansen M, Fessler E, Trinh A, de Rooij
bystanders turning into key players. Curr Opin Genet Dev 2009;19:67–73. LPMH. Poor-prognosis colon cancer is defined by a molecularly distinct
3. Ertel A, Verghese A, Byers S, Ochs M, Tozeren A. Pathway-specific differ- subtype and develops from serrated precursor lesions. Nat Med 2013;
ences between tumor cell lines and normal and tumor tissue cells. Mol 19:614–8.
Cancer 2006;5:55. 19. Sadanandam A, Lyssiotis CA, Homicsko K, Collisson EA, Gibb WJ,
4. Stein WD, Litman T, Fojo T, Bates SE. A Serial analysis of gene expression Wullschleger S, et al. A colorectal cancer classification system that
(SAGE) database analysis of chemosensitivity: comparing solid tumors associates cellular phenotype and responses to therapy. Nat Med
with cell lines and comparing solid tumors from different tissue origins. 2013;19:619–25.
Cancer Res 2004;64:2805–16. 20. Budinska E, Popovici V, Tejpar S, D'Ario G, Lapique N, Sikora KO, et al.
5. Leupin N, Kuhn A, H€ ugli B, Grob TJ, Jaggi R, Tobler A, et al. Gene Gene expression patterns unveil a new level of molecular heterogeneity in
expression profiling reveals consistent differences between clinical colorectal cancer. J Pathol 2013;231:63–76.
samples of human leukaemias and their model cell lines. Br J Haematol 21. Sarfstein R, Friedman Y, Attias-Geva Z, Fishman A, Bruchim I, Werner H.
2006;135:520–3. Metformin downregulates the insulin/IGF-I signaling pathway and inhi-
6. de Gramont A, Figer A, Seymour M, Homerin M, Hmissi A, Cassidy J, et al. bits different uterine serous carcinoma (USC) cells proliferation and
Leucovorin and fluorouracil with or without oxaliplatin as first-line treat- migration in p53-dependent or -independent manners. PLoS One
ment in advanced colorectal cancer. J Clin Oncol 2000;18:2938–47. 2013;8:e61537.
7. Evans JM, Donnelly LA, Emslie-Smith AM, Alessi DR, Morris AD. Metfor- 22. Ben Sahra I, Laurent K, Giuliano S, Larbret F, Ponzio G, Gounon P, et al.
min and reduced risk of cancer in diabetic patients. BMJ 2005;330: Targeting cancer cell metabolism: the combination of metformin and 2-
1304–5. deoxyglucose Induces p53-dependent apoptosis in prostate cancer cells.
8. Morales DR, Morris AD. Metformin in cancer treatment and prevention. Cancer Res 2010;70:2465–75.
Annu Rev Med 2014;1–13. 23. Li P, Zhao M, Parris AB, Feng X, Yang X. p53 is required for metformin-
9. Buzzai M, Jones RG, Amaravadi RK, Lum JJ, DeBerardinis RJ, Zhao F, et al. induced growth inhibition, senescence and apoptosis in breast cancer cells.
Systemic treatment with the antidiabetic drug metformin selectively Biochem Biophys Res Commun 2015;464:1267–74.
impairs p53-deficient tumor cell growth. Cancer Res 2007;67:6745–52. 24. Yi G, He Z, Zhou X, Xian L, Yuan T, Jia X, et al. Low concentration of
10. Zi FM, He JS, Li Y, Wu C, Yang L, Yang Y, et al. Metformin displays anti- metformin induces a p53-dependent senescence in hepatoma cells via
myeloma activity and synergistic effect with dexamethasone in in vitro and activation of the AMPK pathway. Int J Oncol 2013;43:5.
in vivo xenograft models. Cancer Lett 2015;356:443–53. 25. Mohamed Suhaimi NA, Tan MH. Circulating tumor cells and circulating
11. Memmott RM, Mercado JR, Maier CR, Kawabata S, Fox SD, Dennis PA. tumor DNA in colorectal cancer. Expert Rev Precis Med Drug Dev
Metformin prevents tobacco carcinogen–induced lung tumorigenesis. 2016;1:181–94.
Cancer Prev Res 2010;3:1066–76. 26. Jahr S, Hentze H, Englisch S, Hardt D, Fackelmayer FO, Hesch RD, et al.
12. Lengyel E, Litchfield LM, Mitra AK, Nieman KM, Mukherjee A, Zhang Y, DNA fragments in the blood plasma of cancer patients: quantitations and
et al. Metformin inhibits ovarian cancer growth and increases sensitivity to evidence for their origin from apoptotic and necrotic cells. Cancer Res
paclitaxel in mouse models. Am J Obstet Gynecol 2015;212:479.e1-479. 2001;61:1659–65.
e10. 27. Tomochika S, Iizuka N, Watanabe Y, Tsutsui M, Takeda S, Yoshino S, et al.
13. Ben Sahra I, Laurent K, Loubat A, Giorgetti-Peraldi S, Colosetti P, Auberger Increased serum cell-free DNA levels in relation to inflammation are
P, et al. The antidiabetic drug metformin exerts an antitumoral effect predictive of distant metastasis of esophageal squamous cell carcinoma.
in vitro and in vivo through a decrease of cyclin D1 level. Oncogene Exp Ther Med 2010;1:89–92.
2008;27:3576–86. 28. Hirsch HA, Iliopoulos D, Tsichlis PN, Struhl K. Metformin selectively
14. Zordoky BNM, Bark D, Soltys CL, Sung MM, Dyck JRB. The anti-prolifer- targets cancer stem cells, and acts together with chemotherapy to block
ative effect of metformin in triple-negative MDA-MB-231 breast cancer tumor growth and prolong remission. Cancer Res 2009;69:7507–11.
cells is highly dependent on glucose concentration: implications for cancer 29. Quinn BJ, Kitagawa H, Memmott RM, Gills JJ, Dennis PA. Repositioning
therapy and prevention. Biochim Biophys Acta 2014;1840:1943–57. metformin for cancer prevention and treatment. Trends Endocrinol Metab
15. Kinaan M, Ding H, Triggle CR. Metformin: an old drug for the treatment of 2013;24:469–80.
diabetes but a new drug for the protection of the endothelium. Med Princ 30. Martin-Castillo B, Vazquez-Martin A, Oliveras-Ferraros C, Menendez JA.
Pract 2015;24:401–15. Metformin and cancer: doses, mechanisms and the dandelion and hor-
16. Krishnamurthy S, Ng VW, Gao S, Tan MH, Yang YY. Phenformin-loaded metic phenomena. Cell Cycle 2010;9:1057–64.
polymeric micelles for targeting both cancer cells and cancer stem cells in 31. Gatenby RA, Gillies RJ. Why do cancers have high aerobic glycolysis? Nat
vitro and in vivo. Biomaterials 2014;35:9177–86. Rev Cancer 2004;4:891–9.

www.aacrjournals.org Mol Cancer Ther; 16(9) September 2017 2043

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Mohamed Suhaimi et al.

32. Menendez JA, Oliveras-Ferraros C, Cufí S, Corominas-Faja B, Joven J, 41. Pandiri I, Chen Y, Joe Y, Kim HJ, Park J, Chung HT, et al. Tristetraprolin
Martin-Castillo B, et al. Metformin is synthetically lethal with glucose mediates the anti-proliferative effects of metformin in breast cancer cells.
withdrawal in cancer cells. Cell Cycle 2012;11:2782–92. Breast Cancer Res Treat 2016;156:57–64.
33. Tentler JJ, Tan AC, Weekes CD, Jimeno A, Leong S, Pitts TM, et al. Patient- 42. Miranda VC, Braghiroli MI, Faria LD, Bariani G, Alex A, Evangelista J, et al.
derived tumour xenografts as models for oncology drug development. Phase 2 trial of metformin combined with 5-fluorouracil in patients with
Nat Rev Clin Oncol 2012;9:338–50. refractory metastatic colorectal cancer. Clin Colorectal Cancer 2016;15:
34. Hidalgo M, Amant F, Biankin A V, Budinska E, Byrne AT, Caldas C, et al. 321–328.e1.
Patient-derived xenograft models: an emerging platform for translational 43. Sargent DJ, Marsoni S, Monges G, Thibodeau SN, Labianca R, Hamilton SR,
cancer research. Cancer Discov 2014;4:998–1013. et al. Defective mismatch repair as a predictive marker for lack of efficacy of
35. Garralda E, Paz K, L opez-Casas PP, Jones S, Katz A, Kann LM, et al. fluorouracil-based adjuvant therapy in colon cancer. J Clin Oncol
Integrated next-generation sequencing and avatar mouse models for per- 2010;28:3219–26.
sonalized cancer treatment. Clin Cancer Res 2014;20:2476–84. 44. Ribic CM, Sargent DJ, Moore MJ, Thibodeau SN, French AJ, Goldberg RM,
36. Mattie M, Christensen A, Chang MS, Yeh W, Said S, Shostak Y, et al. et al. Tumor microsatellite-instability status as a predictor of benefit from
Molecular characterization of patient-derived human pancreatic tumor fluorouracil-based adjuvant chemotherapy for colon cancer. N Engl J Med
xenograft models for preclinical and translational development of cancer 2003;349:247–57.
therapeutics. Neoplasia 2013;15:1138–50. 45. Viollet B, Guigas B, Sanz Garcia N, Leclerc J, Foretz M, Andreelli F. Cellular
37. Klinghammer K, Raguse JD, Plath T, Albers AE, Joehrens K, Zakar- and molecular mechanisms of metformin: an overview. Clin Sci (Lond)
neh A, et al. A comprehensively characterized large panel of head 2012;122:253–70.
and neck cancer patient-derived xenografts identifies the mTOR 46. Zakikhani M, Blouin MJ, Piura E, Pollak MN. Metformin and rapamycin
inhibitor everolimus as potential new treatment option. Int J have distinct effects on the AKT pathway and proliferation in breast cancer
Cancer 2014;2948. cells. Breast Cancer Res Treat 2010;123:271–9.
38. Seol HS, Kang H, Lee SI, Kim NE, Kim TI, Chun SM, et al. Development and 47. Killion J, Radinsky R, Fidler I. Orthotopic models are necessary to predict
characterization of a colon PDX model that reproduces drug responsive- therapy of transplantable tumors in mice. Cancer Metastasis Rev
ness and the mutation profiles of its original tumor. Cancer Lett 1998;17:279–84.
2014;345:56–64. 48. Lim LS, Hu M, Huang MC, Cheong WC, Gan ATL, Looi XL, et al. Microsieve
39. Hao C, Wang L, Peng S, Cao M, Li H, Hu J, et al. Gene mutations in primary lab-chip device for rapid enumeration and fluorescence in situ hybridiza-
tumors and corresponding patient-derived xenografts derived from non- tion of circulating tumor cells. Lab Chip 2012;12:4388.
small cell lung cancer. Cancer Lett 2015;357:179–85. 49. Mohamed Suhaimi NA, Foong YM, Lee DYS, Phyo WM, Cima I, Lee EX,
40. Rattan R, Giri S, Hartmann LC, Shridhar V. Metformin attenuates ovarian et al. Non-invasive sensitive detection of KRAS and BRAF mutation in
cancer cell growth in an AMP-kinase dispensable manner. J Cell Mol Med circulating tumor cells of colorectal cancer patients. Mol Oncol 2015;
2011;15:166–78. 9:850–60.

2044 Mol Cancer Ther; 16(9) September 2017 Molecular Cancer Therapeutics

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.
Published OnlineFirst May 22, 2017; DOI: 10.1158/1535-7163.MCT-16-0793

Metformin Inhibits Cellular Proliferation and Bioenergetics in


Colorectal Cancer Patient−Derived Xenografts
Nur-Afidah Mohamed Suhaimi, Wai Min Phyo, Hao Yun Yap, et al.

Mol Cancer Ther 2017;16:2035-2044. Published OnlineFirst May 22, 2017.

Updated version Access the most recent version of this article at:
doi:10.1158/1535-7163.MCT-16-0793

Supplementary Access the most recent supplemental material at:


Material http://mct.aacrjournals.org/content/suppl/2017/05/22/1535-7163.MCT-16-0793.DC1

Cited articles This article cites 47 articles, 12 of which you can access for free at:
http://mct.aacrjournals.org/content/16/9/2035.full#ref-list-1

Citing articles This article has been cited by 2 HighWire-hosted articles. Access the articles at:
http://mct.aacrjournals.org/content/16/9/2035.full#related-urls

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at
Subscriptions pubs@aacr.org.

Permissions To request permission to re-use all or part of this article, use this link
http://mct.aacrjournals.org/content/16/9/2035.
Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)
Rightslink site.

Downloaded from mct.aacrjournals.org on May 27, 2021. © 2017 American Association for Cancer Research.

You might also like