throbber
Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Integrated Systems and Technologies
`
`Cancer
`Research
`
`Free Somatostatin Receptor Fraction Predicts the
`Antiproliferative Effect of Octreotide in a Neuroendocrine
`Tumor Model: Implications for Dose Optimization
`
`Pedram Heidari1, Eric Wehrenberg-Klee1, Peiman Habibollahi1, Daniel Yokell1,
`Matthew Kulke2, and Umar Mahmood1
`
`Abstract
`
`Somatostatin receptors (SSTR) are highly expressed in well-differentiated neuroendocrine tumors (NET).
`Octreotide, an SSTR agonist, has been used to suppress the production of vasoactive hormones and relieve
`symptoms of hormone hypersecretion with functional NETs. In a clinical trial, an empiric dose of octreotide
`treatment prolonged time to tumor progression in patients with small bowel neuroendocrine (carcinoid) tumors,
`irrespective of symptom status. However, there has yet to be a dose optimization study across the patient
`population, and methods are currently lacking to optimize dosing of octreotide therapy on an individual basis.
`Multiple factors such as total tumor burden, receptor expression levels, and nontarget organ metabolism/
`excretion may contribute to a variation in SSTR octreotide occupancy with a given dose among different patients.
`In this study, we report the development of an imaging method to measure surface SSTR expression and
`occupancy level using the PET radiotracer 68Ga-DOTATOC. In an animal model, SSTR occupancy by octreotide
`was assessed quantitatively with 68Ga-DOTATOC PET, with the finding that increased occupancy resulted in
`decreased tumor proliferation rate. The results suggested that quantitative SSTR imaging during octreotide
`therapy has the potential to determine the fractional receptor occupancy in NETs, thereby allowing octreotide
`dosing to be optimized readily in individual patients. Clinical trials validating this approach are warranted. Cancer
`Res; 73(23); 6865–73. Ó2013 AACR.
`
`Introduction
`Neuroendocrine tumors (NET) are a heterogeneous group of
`malignancies that are thought to originate from endocrine
`progenitor cells located in various organ systems including the
`lung, pancreas, and gastrointestinal tract. Very commonly,
`these NETs secrete a variety of biologically active peptides
`and amines that can lead to symptoms of wheezing, nausea,
`abdominal pain, flushing, and diarrhea, among others (1).
`Somatostatin receptor (SSTR) agonists have been employed
`with great success for controlling these symptoms (1, 2). With
`80% to 100% of well-differentiated NETs expressing high levels
`of SSTR (3), somatostatin analogs, such as octreotide, have
`become the treatment of choice for symptomatic relief through
`the reduction of NET hormone production.
`
`Authors' Affiliations: 1Division of Nuclear Medicine and Molecular Imag-
`ing, Department of Radiology, Massachusetts General Hospital, Harvard
`Medical School and 2Department of Medical Oncology, Dana-Farber
`Cancer Institute, Harvard Medical School, Boston, Massachusetts
`
`P. Heidari and E. Wehrenberg-Klee contributed equally to this work.
`
`Corresponding Author: Umar Mahmood, Division of Nuclear Medicine
`and Molecular Imaging, Department of Radiology, Massachusetts General
`Hospital, Boston, MA 02114. Phone: 617-726-6477; Fax: 617-726-6165;
`E-mail: umahmood@mgh.harvard.edu
`
`doi: 10.1158/0008-5472.CAN-13-1199
`Ó2013 American Association for Cancer Research.
`
`Over the past several years, it has further been demon-
`strated that SSTR agonists may also have an antiproliferative
`effect on NETs (4, 5) and may have a role as antineoplastic
`therapy for NETs. It was shown in a double-blind randomized
`controlled trial that patients with metastatic well-differen-
`tiated mid-gut NETs who received monthly intramuscular
`injections of a standard dose (30 mg) of long-acting octreo-
`tide (octreotide LAR) had a significantly increased progres-
`sion-free survival (PFS) compared to those who received
`placebo (15.6 vs. 5.9 months, respectively; ref. 6). The patients
`in this study benefited from octreotide LAR therapy regard-
`less of tumor functional status. The results of this trial
`suggest that octreotide may possess antiproliferative prop-
`erties on NETs that are enacted through SSTR-mediated
`signaling, irrespective of activation of pathways involved in
`bioactive peptide and amine production. This paradigm shift
`in the use of SSTR agonists for their antitumor effects has
`been empiric in nature, without an understanding of what
`fraction of the somatostatin receptors were bound during
`agonist therapy, how this changed over the course of a
`monthly treatment cycle, and the association with prolifer-
`ation. The development of a noninvasive approach to quan-
`tify the concentration of SSTR in tumors, the change in free
`receptor fraction with therapy, and the resultant downstream
`effects on proliferation would provide a method to optimize
`this treatment, both at the individual level and across treat-
`ment populations.
`
`www.aacrjournals.org
`
`6865
`
`Downloaded from
`
`cancerres.aacrjournals.org
`
`on June 14, 2017. © 2013 American Association for Cancer Research.
`
`Ex. 1089-0001
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Heidari et al.
`
`Radionuclide imaging of SSTR through 111In-octreotide or
`68Ga–DOTATOC has largely been qualitative in nature, using the
`high contrast gained from the images to locate foci of disease not
`apparent on other imaging modalities or to monitor disease
`progression by the assessment of tumor volume changes over
`time (7–9). In fact, tumor imaging with these agents is typically
`performed either for primary staging before initiation of octreo-
`tide LAR therapy or monitoring of disease progression during
`the expected nadir blood levels of octreotide LAR to aid in tumor
`visualization (10, 11). The objective in this study was to use the
`quantitative nature of 68Ga–DOTATOC PET imaging to develop
`a technique that allows one to compute the receptor density in a
`tumor volume and monitor the fraction that is occupied with
`agonist treatment at a given time, based on changes in the
`available (unbound) receptor density. In combination with
`0
`0
`another positron-emitting radiotracer, 18F-fluoro-3
`-deoxy-3
`-
`L-fluorothymidine (18F-FLT), a proliferation marker (12), we
`were able to correlate the changes in unoccupied SSTR with
`proliferation status in an animal model. The imaging techniques
`and quantitation methods thus developed have the potential to
`be readily translated to patients with NETs to more effectively
`monitor treatment and improve dosing regimens.
`
`Materials and Methods
`Cell culture
`AR42J [American Type Culture Collection (ATCC)], an STTR
`expressing rat pancreatic carcinoma, was cultured in F-12K
`medium (ATCC), supplemented by 20% (v/v) FBS, 100 U/mL
`penicillin, and 100 mg/mL streptomycin. A549 (ATCC), an
`SSTR-negative human alveolar basal epithelial carcinoma cell
`line, was cultured in F-12K medium (ATCC), supplemented by
`10% (v/v) FBS, 100 U/mL penicillin, and 100 mg/mL strepto-
`mycin. Cultures were maintained in a humidified incubator at
`
`37
`C, 5% CO2. Subculturing was performed employing a 0.25%
`trypsin/0.1% EDTA solution. The cell lines were obtained from
`ATCC and were used in this study for less than 6 months after
`resuscitation. Cell lines undergo comprehensive quality con-
`trol and authentication procedures by ATCC before shipment.
`These include testing for mycoplasma by culture isolation,
`Hoechst DNA staining, and PCR, together with culture testing
`for contaminant bacteria, yeast, and fungi. Authentication
`procedures used include species verification by DNA barcoding
`and identity verification by DNA profiling.
`
`68Ga labeling of DOTATOC
`A 68Ge/68Ga generator (iThemba Labs) was eluted with 6 mL
`of 0.6 N HCl. The eluant was added to a buffer system of 2 mol/L
`HEPES at pH 3.5 to 4.0 with 5 mg of DOTATOC. The reaction
`
`solution was heated at 100
`C for 20 minutes. The reaction
`product was loaded on a reverse-phase C18 Sep-Pak mini
`cartridge and eluted with 200 mL of 200-proof ethanol. The final
`formulation was adjusted to 10% ethanol in saline. The chemical
`and radiochemical purity of 68Ga-DOTATOC was measured
`through radio thin-layer chromatography (TLC; refs. 13, 14).
`
`Competitive binding study
`To evaluate the specific binding of 68Ga-DOTATOC, a com-
`petitive binding assay using a fixed concentration of radio-
`
`tracer and increasing concentration of octreotide acetate was
`performed. AR42J (SSTR2-expressing) and A549 (SSTR-nega-
`tive) cells were seeded in 24-well plates (2.5  105/well) and
`allowed to grow to 80% confluence. Wells were incubated for 1
`hour with 0.01 to 1,000 mmol/L concentration of octreotide
`acetate (Abbiotec). Then, 25 mCi 68Ga-DOTATOC (9.5 nmol/L
`DOTATOC peptide) was added to each well and plates were
`
`incubated at 37
`C for 1 hour. The medium was removed and
`
`C PBS. Cells were collected
`wells were washed 3 times using 4
`after trypsin treatment, and the number of cells in each well
`was counted using an automated cell counter (Countess,
`Invitrogen). 68Ga activity in the cells in each well was assayed
`using an automated gamma counter (Wizard 2480, Perkin
`Elmer) and decay corrected for the beginning of incubation
`with 68Ga-DOTATOC.
`
`In vitro cell-cycle assay
`To assess the effect of octreotide on cell-cycle progression,
`AR42J and A549 were seeded in 6-well plates and incubated at
`
`C for 24 hours (A549) or 2 days (AR42J) in cell culture
`37
`medium. The medium was then removed and fresh medium
`was added to each well. Wells were randomized to receive
`octreotide acetate at a concentration of 1 mmol/L or no
`
`octreotide acetate and all wells were incubated at 37
`C for
`0
`-deoxyuridine;
`24 hours before the addition of EdU (5-ethynyl-2
`Click-it EDU kit, Invitrogen), a fluorescent DNA analog that is
`incorporated during DNA synthesis. Treated cells were then
`sorted for cell-cycle phase using fluorescence-activated cell
`sorting (FACS) and the percentage of the cells in S-phase
`determined in octreotide treated versus nontreated groups
`for each cell line. Studies were performed in triplicate.
`
`Western blotting for SSTR2 expression
`Nude (nu/nu) mice were injected subcutaneously with 106
`AR42J cells suspended in Matrigel (BD Biosciences) in the left
`upper flank. After PET imaging, AR42J tumors were removed
`and extracted and whole protein extract purification per-
`formed. Protein samples (30 mg) were loaded onto SDS-PAGE
`and run at 120 V and 14 mA for 1.5 hours. Gels were blotted on
`polyvinylidene difluoride (PVDF) membrane and the blots
`
`incubated overnight at 4
`C with SSTR2 monoclonal antibody
`(Abcam) at 1:500 dilution. b-Actin monoclonal antibody (Santa
`Cruz) at 1:1,000 dilution was used as an internal control.
`Detection was performed using the BM Chemiluminescence
`Western Blotting Kit (Mouse/Rabbit; Roche) and imaged on
`the Carestream In-Vivo Multispectral FX Imaging System.
`Quantitation of SSTR2 and b-actin expression was performed
`by drawing a region of interest around the protein bands on
`chemiluminescence images acquired with the Carestream
`Molecular Imaging Software. The acquired data were normal-
`ized using b-actin expression and corrected for tumor weight.
`
`In vivo imaging studies
`AR42J-bearing mice were divided randomly in 4 groups
`(n ¼ 3 in each group) that received vehicle, 1.25, 2.5, or 10
`mg/kg octreotide acetate, delivered via intraperitoneal injec-
`tion every 6 hours for a total of 5 injections to reach steady-
`state blood levels. Five hours following injection of the fifth
`
`6866
`
`Cancer Res; 73(23) December 1, 2013
`
`Cancer Research
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0002
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Free Somatostatin Receptor Predicts Proliferation
`
`In vitro studies. A, 68Ga-DOTATOC demonstrated significantly higher binding to AR42J compared to A549 cells (P < 0.0001). B, in vitro competitive
`Figure 1.
`binding studies demonstrate binding affinity of 68Ga-DOTATOC to SSTR. Octreotide concentration  10 mmol/L completely binds all sites, whereas doses of
`1 mmol/L allow for 68Ga-DOTATOC accumulation in graded manner in AR42J cells. A549 serve as a negative control with minimal expression of
`SSTR. C, gated FACS EDU binding data demonstrates that treatment with octreotide significantly decreased (P < 0.001) the number of AR42J cells in S-phase,
`whereas it had no effect on the number of A549 cells in S-phase. D, gated FACS EDU graph shows decreased proliferation in octreotide-treated versus
`control cultures for AR42J (top row), compared to A549 cells (second row), which showed no difference. Arrows, the cells in the S-phase.
`
`dose (trough blood level of octreotide acetate), the mice
`underwent dynamic PET imaging with 68Ga-DOTATOC.
`Immediately following, the mice were imaged in static mode
`and then a sixth dose of octreotide was administered. Five
`hours after receiving the last dose of treatment solution the
`mice were imaged using 18F-FLT PET in static mode.
`
`Static PET imaging protocol
`Approximately 400 mCi of 68Ga-DOTATOC prepared as
`described above and diluted into a final volume of 150 to
`200 mL that was injected intravenously via tail vein, and 1 hour
`later, static PET images were acquired for 15 minutes in 2 bed
`positions using the Sedecal Argus microPET. Static PET imag-
`ing with 18F-FLT was performed 2 hours after intravenous
`injection of 400 mCi 18F-FLT. Images were reconstructed using
`2D-OSEM (4 iterations, 16 subsets) and were corrected for
`scatter and randoms. The mean standard uptake value (SUV-
`mean) for each tumor was calculated in a 3-dimensional (3D)
`region of interest autodrawn around the tumor using a 30%
`isocontour threshold.
`
`Dynamic PET imaging protocol and compartmental
`modeling
`Mice were placed under anesthesia with 2% isoflurane in O2
`and positioned on the scanner such that heart and tumor were
`both in the field of view. Dynamic PET data were acquired in
`list mode for 60 minutes beginning immediately before injec-
`tion of 400 mCi 68Ga-DOTATOC in 150 to 200 mL of volume via
`tail vein. The list mode data were then reframed in 40 fifteen-
`second, 20 thirty-second, 16 sixty-second, and 16 ninety-sec-
`ond frames. Scans were reconstructed and a 3D region of
`interest was set around the tumor, as described above. The
`input function was measured from a spherical region of
`interest with a 3-mm diameter over the center of the mouse
`heart. Time activity curves were plotted for tumor and blood
`pool. To determine the best compartmental model fit for 68Ga-
`DOTATOC binding, an octreotide challenge study was per-
`formed during a dynamic 68Ga-DOTATOC PET study in AR42J-
`bearing mice. The octreotide dose (150 mg) was injected via tail
`vein 10 minutes after the scan start and the administration of
`68Ga-DOTATOC.
`
`www.aacrjournals.org
`
`Cancer Res; 73(23) December 1, 2013
`
`6867
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0003
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Heidari et al.
`
`Ki-67 staining
`To determine tumor cell proliferation changes in response
`to octreotide therapy in vivo, Ki-67 staining was performed.
`Tumor samples from AR42J tumor–bearing mice from the
`different treatment groups described above were excised and
`kept in frozen tissue-embedding fixative (Fisher Scientific) at
`80
`
`C for further immunofluorescent staining. Briefly, slide-
`mounted 5-mm-thick sections were prepared and fixed using
`ice-cold acetone for 10 minutes followed by 3 wash with PBS
`(5 minutes each). Tissue sections were blocked for 30 minutes
`with 1% bovine serum albumin in PBS with Tween, washed
`with PBS, and incubated overnight at þ4
`
`C with Ki-67 anti-
`body (Abcam). The slides were washed again with PBS,
`mounted with mounting medium for fluorescence-containing
`DAPI (Vector Laboratories), and visualized by confocal fluo-
`rescent microscopy (Nikon). Ki-67–stained and -unstained
`cells in the resulting images were segmented using ImageJ,
`software and the percentage of cells stained for Ki-67 was
`determined.
`
`Statistical analysis
`The statistical analysis was performed using GraphPad
`Prism 5. Unpaired t test was used to compare the number of
`cells in S-phase in control and treatment groups. One-way
`ANOVA was employed to discern the differences in SUVmean
`and Ki among different treatment groups in mice. Tukey
`multiple comparison test was used to compare the significance
`between groups. The 68Ga-DOTATOC influx rate and SUV-
`mean were plotted against 18F-FLT SUVmean, and correlation
`between the measurements was determined using linear
`regression. P < 0.05 was considered statistically significant.
`Mean values are reported  SEM.
`Results
`Competitive binding and in vitro cell-cycle assays
`68Ga-DOTATOC demonstrated 8.6-fold greater binding to
`AR42J compared to A549 cells (P < 0.0001; Fig. 1A). This is
`consistent with the high expression of SSTR type 2 in AR42J
`cells and undetectable expression of SSTR2 in A549 cells. In
`the competition receptor-binding assay, nonlabeled octreotide
`competed specifically with the 68Ga-DOTATOC for binding to
`the AR42J cells. As shown in Fig. 1B, treatment of AR42J cells
`with increasing doses of octreotide acetate led to decreased
`68Ga-DOTATOC uptake. 68Ga-DOTATOC influx was complete-
`ly inhibited at an octreotide concentration of 10 mmol/L or
`higher. A549 did not show considerable 68Ga-DOTATOC
`uptake or displacement with octreotide treatment. This find-
`ing shows that 68Ga-DOTATOC can be used to monitor the
`SSTR octreotide occupancy in SSTR expressing cells. Cell-cycle
`assays were performed to demonstrate the effect of octreotide
`on cell proliferation. As seen in Fig. 1C and D, for SSTR-
`expressing AR42J cells, treatment with octreotide decreased
`the percentage of cells in S-phase by 53% compared to control
`(P < 0.001). In SSTR nonexpressing A549 cells, treatment with
`octreotide did not lead to a significant difference in S-phase
`compared to control (P > 0.5). These data suggest that octreo-
`tide exerts a downstream inhibitory effect on cell proliferation
`through somatostatin receptors.
`
`Correlation of imaging findings with SSTR2 protein
`levels
`There was a consistent ratio between SSTR2 expression
`level and 68Ga-DOTATOC quantitative imaging measures
`irrespective of tumor size (tumors with diameter 3.9–8.2
`mm) in AR42J tumors. The total 68Ga-DOTATOC uptake of
`the tumors as measured by the molecular tumor burden
`(MTB; ref. 15) on PET studies, which were acquired in the
`static mode, strongly correlates with the total SSTR2 content
`in tumors (R2 ¼ 0.99, P < 0.0001), as seen in Fig. 2A. MTB is
`the product of SUVmean and molecular tumor volume (total
`volume of the voxels in the region of interest with 68Ga-
`DOTATOC uptake above the defined threshold; ref. 16).
`Moreover, the mean 68Ga-DOTATOC uptake in tumors
`(SUVmean) also strongly correlates (Fig. 2B) with the expres-
`sion of SSTR2 normalized for b-actin expression (R2 ¼ 0.85,
`P < 0.0004). These findings demonstrate that the noninva-
`sively measured 68Ga-DOTATOC uptake is a true reflection
`of the SSTR2 levels in the tumors.
`
`Dynamic 68Ga-DOTATOC PET imaging of AR42J tumors
`Octreotide challenge studies showed that 68Ga-DOTATOC
`is partially displaced by octreotide but a large fraction was not
`
`Figure 2. Western blotting results demonstrate a consistent ratio between
`SSTR2 expression level and 68Ga-DOTATOC quantitative measures
`irrespective of tumor size (tumors with diameter 3.9–8.2 mm) in AR42J
`tumors. A, total 68Ga-DOTATOC uptake of the tumors (MTB) strongly
`correlates with the total SSTR2 content in tumors. B, the mean 68Ga-
`DOTATOC uptake in tumors (SUVmean) also strongly correlates with the
`SSTR2/b-actin expression ratio.
`
`6868
`
`Cancer Res; 73(23) December 1, 2013
`
`Cancer Research
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0004
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Free Somatostatin Receptor Predicts Proliferation
`
`Figure 3. Dynamic 68Ga-DOTATOC scans of 2 AR42J-bearing mice. A, in this dataset, octreotide IV challenge at 10 minutes resulted in partial displacement of
`tracer from the receptor, but a component of the uptake could not be displaced even though the Patlak plots (B) show essentially no tracer influx after
`octreotide challenge in steady-state condition (orange dots). Please note that the slope of the best-fit line (Ki) in the steady state is approximately zero
`(R2 ¼ 0.98). C, without an octreotide drug challenge, the tracer accumulates in the tumor with a constant rate, as is evident in the Patlak plot (D); in the steady-
`state condition, the orange dots the slope of the best-fit line (Ki) is 0.69 (R2 ¼ 0.99). E, this pattern of tracer uptake is compatible with an irreversible two-tissue
`Rt
`CPðtÞdt
`in the x-axis is plotted against CTissueðtÞ
`compartment model. F, the x- and y-axes of the Patlak plots (such as B and D) are calculated using formula 1;
`CPðtÞ
`CPðtÞ
`the y-axis to draw the Patlak plot and K is measured using linear regression. The net tracer influx rate (Ki) is measured using formula 2. t, time after
`tracer injection; CTissue, the amount of tracer in the region of interest; CP(t), the concentration of tracer in plasma; K, the rate of entry into the
`irreversible compartment; V0, the distribution volume of the tracer in the central compartment.
`
`in
`
`0
`
`displaceable following competitive challenge. The displace-
`able fraction was the 68Ga-DOTATOC bound to SSTR but not
`yet internalized, whereas the remaining component was
`already internalized and non-displaceable (Fig. 3A and C).
`These findings were compatible with an irreversible 2-com-
`partment tissue model (Fig. 3E; ref. 17). Thus, on the basis of
`the equations in Fig. 3F, we employed a Patlak graphical plot
`and calculated the tumor influx constant (Ki; refs. 18, 19). We
`noted that for all studies, steady state was achieved in less
`than 25 minutes, and thus a 25-minute cutoff was used for
`fitting the Ki. The net 68Ga-DOTATOC influx rate, measured
`using a Patlak plot, following IV challenge of octreotide
`1/min
`1 (Fig.
`decreased to 0.0 (mL plasma)/(mL tissue)
`
`3B), whereas the influx rate was approximately 0.7 (mL
`1/min
`1 without an octreotide chal-
`plasma)/(mL tissue)
`lenge (Fig. 3D).
`The parameter Ki, which is the net rate of 68Ga-DTOATOC
`influx, is independent of tumor perfusion and reflects the
`number of available receptors as well as the rate of receptor
`trafficking (17–19). Measurement of the Ki in AR42J tumors
`demonstrated
`that
`octreotide
`treatment
`significantly
`decreased the rate of tracer influx in all treatment groups
`compared to control and that the mean Ki monotonically
`decreased with higher doses. The mean Ki was 0.67  0.02,
`0.61  0.03, 0.23  0.02, and 0.17  0.03 (mL plasma)/(mL
`
`1/min1 in vehicle, 1.25, 2.5, and 10 mg/kg treatment
`tissue)
`
`www.aacrjournals.org
`
`Cancer Res; 73(23) December 1, 2013
`
`6869
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0005
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Heidari et al.
`
`Figure 4. Dynamic PET imaging of AR42J tumors in vehicle (A), 1.25 mg/kg (B), 2.5 mg/kg (C), and 10 mg/kg (D) octreotide groups with accompanying Patlak
`plot on top right corner of each panel (orange). There is a graded decrease in the slope of the Patlak plot (Ki) after reaching steady state with increasing doses
`of octreotide treatment (blue, blood pool; red, tumor uptake; purple, tumor/blood pool ratio; green, data points in Patlak plot before reaching steady-state
`blood levels; orange, data points in Patlak plot after reaching steady-state blood levels).
`
`groups, respectively, as shown in Fig. 4. There was a significant
`decrease in the Ki of all treatment groups compared to the
`vehicle group. The decreasing Ki reflects the number of recep-
`tors occupied by octreotide and that are unavailable to bind
`68Ga-DOTATOC.
`
`Static 68Ga-DOTATOC PET imaging of AR42J tumors
`Static scan results confirmed and paralleled the results
`of dynamic 68Ga-DOTATOC PET imaging. Treatment with
`increasing doses of octreotide acetate led to progressively
`significant decreases in tumor SUVmean compared to con-
`trol. Representative images are shown in Fig. 5. The mean
`SUVmean was 0.96  0.05, 0.88  0.08, 0.42  0.03, and 0.21 
`0.04 in vehicle, 1.25, 2.5, and 10 mg/kg treatment groups,
`respectively (Fig. 6B). As shown in the Fig. 6D, there was a
`very strong correlation between tumor SUVmean and Ki
`measured by static and dynamic 68Ga-DOTATOC PET imaging
`(R2 ¼ 0.95, P < 0.0001). The high agreement of quantitative
`parameters between static and dynamic 68Ga-DOTATOC PET
`in our study suggests that SUVmean of the tumors in static
`PET, although not as accurate as Ki measured by dynamic
`PET for free receptor density, can be effectively employed to
`monitor SSTR occupancy with octreotide treatment.
`
`Static 18F-FLT PET imaging and its correlation with
`68Ga-DOTATOC PET imaging
`Treatment with octreotide acetate decreased the SUVmean
`of 18F-FLT tumor uptake in all treatment groups compared to
`the control group. As with dynamic and static 68Ga-DOTATOC
`
`PET imaging, higher doses of octreotide led to monotonically
`decreasing 18F-FLT PET SUVmean. The mean SUVmean was
`1.40  0.10, 1.30  0.08, 0.34  0.06, and 0.18  0.03 in vehicle,
`1.25, 2.5, and 10 mg/kg treatment groups, respectively, as seen
`in Fig. 6. There was a strong correlation between the SUVmean
`of the tumors in 18F-FLT PET scans and both SUVmean and
`Ki of tumors in 68Ga-DOTATOC PET scans (R2 ¼ 0.95, P <
`0.0001 and R2 ¼ 0.97, P < 0.0001, respectively; Fig. 6E and F).
`This demonstrates that increased occupancy of SSTR with
`octreotide results in a reduced rate of tumor proliferation in
`vivo, assessed by static18F-FLT PET; the magnitude of reduc-
`tion in tumor proliferation rate is directly correlated with
`and dependent on the level of SSTR octreotide occupancy
`and thus can be potentially monitored using 68Ga-DOTA-
`TOC PET imaging.
`
`Ki-67 staining results
`Treatment of AR42J tumors with increasing doses of octreo-
`tide led to a reduction in the percentage of cells staining for Ki-
`67. The mean percentage of cells stained for Ki-67 was 25  1.2,
`23  0.8, 11  0.9, and 5  1.1 in the vehicle, 1.25, 2.5, and 10
`mg/kg treatment groups, respectively. Increase in the SSTR
`octreotide occupancy results in a corresponding decrease in
`the rate of tumor proliferation shown by the decrease in
`relative number of cells stained for Ki-67 (Fig. 5). These results
`are consistent with the results obtained by noninvasive
`18F-FLT PET imaging and further confirm the enhanced anti-
`proliferative effects of octreotide on tumor cells with increas-
`ing level of SSTR octreotide occupancy.
`
`6870
`
`Cancer Res; 73(23) December 1, 2013
`
`Cancer Research
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0006
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Free Somatostatin Receptor Predicts Proliferation
`
`Figure 5.
`Imaging of AR42J tumors
`using static 68Ga-DOTATOC (A-D),
`18F-FLT (E-H), and staining for Ki-
`67 in tumors samples (I-L) in 4
`groups of mice treated with vehicle,
`1.25, 2.5, or 10 mg/kg groups.
`There is high 68Ga-DOTATOC and
`18F-FLT uptake as well as 25%
`cells stained for Ki-67 in tumors in
`the control group; there is a graded
`decrease across all the biomarkers
`with increased dose of octreotide
`treatment. The 10 mg/kg
`octreotide dose shows near
`background levels of 68Ga-
`DOTATOC and 18F-FLT uptake
`and 5% staining for Ki-67.
`
`Discussion
`Octreotide continues to play a key role in the treatment of
`patients with metastatic NET, both for the control of symp-
`toms of hypersecretion and, more recently, for control of tumor
`growth (1, 20–22). In an efficacy clinical trial called the PRO-
`MID trial (6), it was shown that a fixed monthly injection of
`octreotide LAR significantly increased the time to tumor
`progression in patients with metastatic mid-gut carcinoids by
`8.3 months compared to placebo, demonstrating the antipro-
`liferative effect of octreotide LAR on well-differentiated mid-
`gut NETs. However, the potential for optimized individual
`dosing of octreotide to lead to further significant improve-
`ments in PFS has never been explored. Quantitative noninva-
`sive assessment of somatostatin receptor occupancy and
`downstream pharmacodynamic assessment of proliferation,
`as demonstrated in this study, could directly guide such
`personalized dosing optimization.
`This paradigm shift from the use of imaging for disease
`detection to disease characterization, including the direct and
`downstream molecular effects of therapy on specific tumors,
`provides an opportunity to use such assessment to prospec-
`tively guide tailored therapy rather than retrospective report-
`ing on treatment effectiveness measured by tumor volume
`changes. Such paired imaging of upstream intracellular sig-
`naling from cell surface receptors and downstream effects such
`as proliferation changes or alterations in apoptosis rates could
`be applied to a broad range of targeted therapies for individual
`patient drug dosing optimization. This includes therapies
`
`targeted at receptor tyrosine kinases, estrogen receptors, and
`androgen receptors, among other targets.
`The quantitative nature of the 68Ga-DOTATOC PET measure-
`ments allows an indirect assessment of fractional SSTR occu-
`pancy. Our kinetic model is similar to that developed by Henze
`and colleagues (17) for characterization of the kinetics of 68Ga-
`DOTATOC uptake in brain meningiomas. Using dynamic PET
`imaging, we established that 68Ga-DOTATOC and octreotide
`directly compete for binding to SSTR and that binding of
`somatostatin analogs results in irreversible internalization
`(23) of the ligand receptor complex. Using this tracer kinetic
`model, we could then reliably calculate SSTR free fraction with
`increasing doses of octreotide. We demonstrated a highly sig-
`nificant correlation between the net tracer influx rate (Ki) and
`the SUV measurement. The net tracer influx rate (Ki) is mea-
`sured by Patlak graphical analysis of dynamic PET data based on
`an irreversible 2-compartment kinetic model, which removes
`the effects of perfusion from the calculated receptor-mediated
`uptake values, whereas SUV, which is calculated from static PET
`data, is more routinely employed in the clinic and does not
`separate perfusion effects from the receptor mediated uptake.
`For clinical translation, both the net tracer influx rate (Ki) and
`SUV measurements could be determined in patients. Given the
`typical enhancement pattern seen on CT scanning of carcinoid
`tumors (24, 25) suggestive of high tumoral perfusion, these may
`correlate clinically as well as in preclinical assessment per-
`formed in this study, providing a means to more simply translate
`this approach for patient assessment.
`
`www.aacrjournals.org
`
`Cancer Res; 73(23) December 1, 2013
`
`6871
`
`Downloaded from
`
`
`
`on June 14, 2017. © 2013 American Association for Cancer Research. cancerres.aacrjournals.org
`
`Ex. 1089-0007
`
`

`

`Published OnlineFirst September 30, 2013; DOI: 10.1158/0008-5472.CAN-13-1199
`
`Heidari et al.
`
`Figure 6. A–C, results of PET imaging in 4 treatment groups of mice bearing AR42J tumors (n¼ 3 in each treatment group). A, net tracer influx rate (Ki) in dynamic
`68Ga-DOTATOC shows a graded and significant decrease in uptake with increased dose of octreotide. B, the same pattern of decrease is observed in
`SUVmean of tumors in static 68Ga-DOTATOC PET (B) and 18F-FLT PET (C). D, there is strong correlation between SUVmean of tumor in static 68Ga-DOTATOC
`PET scans and Ki in dynamic 68Ga-DOTATOC PET analysis. E and F, correlation between SUVmean of tumor in 18F-FLT PET scans and Ki in dynamic
`68Ga-DOTATOC PET (D) as well as SUVmean in static 68Ga-DOTATOC PET (E). There is very strong correlation between 18F-FLT uptake in the tumor and both
`Ki and SUVmean of tumor in 68Ga-DOTATOC PET scans. , P < 0.05.
`
`One possible limitation to measuring receptor occupancy
`level using PET imaging is that it is most useful when the
`receptor-targeted therapy such as octreotide is in the subsatur-
`ating range. If the administered dose of octreotide is high enough
`to completely saturate receptors, then there will be no tracer
`uptake in the intracellular compartment;
`in these circum-
`stances, receptor quantitation using PET imaging shows com-
`plete occupancy and the degree of excess octreotide dose cannot
`be assessed. In practice, the majority of carcinoid patients is
`given subsat

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket