throbber
Hindawi Publishing Corporation
`Journal of Oncology
`Volume 2012, Article ID 320198, 10 pages
`doi:10.1155/2012/320198
`
`Clinical Study
`Radiolabeled Somatostatin Analogues Therapy in Advanced
`Neuroendocrine Tumors: A Single Centre Experience
`
`A. Filice,1 A. Fraternali,1 A. Frasoldati,2 M. Asti,1 E. Grassi,3 L. Massi,1
`M. Sollini,1, 4 A. Froio,1 P. A. Erba,1, 4 and A. Versari1
`
`1 Department of Nuclear Medicine, Azienda Ospedaliera Santa Maria Nuova, IRCCS Reggio Emilia, Via Risorgimento 80,
`42100 Reggio Emilia, Italy
`2 Department of Endocrinology, Azienda Ospedaliera Santa Maria Nuova, IRCCS Reggio Emilia, Via Risorgimento 80,
`42100 Reggio Emilia, Italy
`3 Department of Medical Physics, Azienda Ospedaliera Santa Maria Nuova, IRCCS Reggio Emilia, Via Risorgimento 80,
`42100 Reggio Emilia, Italy
`4 Nuclear Medicine Unit, University of Pisa, Via Roma 55, 56125 Pisa, Italy
`
`Correspondence should be addressed to A. Versari, versari.annibale@asmn.re.it
`
`Received 30 April 2012; Revised 25 June 2012; Accepted 26 June 2012
`
`Academic Editor: Marialuisa Appetecchia
`
`Copyright © 2012 A. Filice et al. This is an open access article distributed under the Creative Commons Attribution License, which
`permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`The aim of this study was to assess the efficacy of PRRT in patients with advanced neuroendocrine tumors (NETs). Patients
`and Methods. From January 2007 to August 2011, we enrolled 65 patients (m/f 38/27; mean age 65 years, range 33–83) with
`advanced NETs having enhanced SSTR expression, treated with PRRT. The enhanced expression of SSTR was assessed using
`68Ga-DOTATOC/DOTATATE PET/CT. Among all the enrolled patients, 6 of them were excluded from the present analysis
`since they voluntarily interrupted treatment. Mean activity/cycle of 2.6 GBq (90Y-DOTATOC/DOTATATE) or 6.0 GBq (177Lu-
`DOTATOC/DOTATATE) was administrated intravenously (max 9 cycles). Results. Complete response (CR) was found in 1/59
`(2%) patients, partial remission (PR) in 24/59 (40.5%) patients, stable disease (SD) in 24/59 (40.5%), and progression (PD) in
`10/59 (17%) patients. The overall tumor response rate (CR + PR) was 42.5%. In 40.5% of patients, the disease could be stabilized.
`Overall, 49 out of 59 patients had no tumor progression (83%). Twelve patients out of 59 (20%) had grade 2-3 hematological side
`effects including anemia, thrombocytopenia, and leukopenia. Long-term nephrotoxicity was observed in 3 patients (2 moderate,
`1 severe). Conclusions. PRRT is a promising perspective for patients with advanced NETs.
`
`1. Introduction
`
`Neuroendocrine tumors (NETs) are considered a class of
`rare neoplasms accounting <5% of all tumors. However,
`diagnosis of NETs has increased substantially over the last
`decades and prevalence is now greater than that of any other
`upper gastrointestinal tumor [1]. These tumors originate
`from dispersed neuroendocrine cells, distributed almost
`ubiquitously in the body [2], and occur in 5/100,000 people
`per year [1].
`The most frequent sites of NETs are gastroenteropancre-
`atic tract (GEP NETs), followed by lungs; less frequently skin,
`adrenal glands, thyroid, and genital tracts may be affected.
`
`Different nomenclature systems and classifications have been
`used for NETs.
`Current pathological staging and grading differ between
`Europe and USA; however, both classification systems are
`centered on the primary site of the tumor and histological
`grade. In Europe, the Ki-67 proliferative index is used to
`differentiate tumors of low (<2%), intermediate (2–20%)
`and high (>20%) grade, whereas in the USA, tumors are
`graded as “well-” and “poorly-” differentiated where “well”
`equates to low-intermediate grade and “poorly” equates to
`high-grade tumors [3, 4].
`Up to 80% of GEP NETs express somatostatin receptors
`(SSTR2 and SSTR5 primarily). Therefore, somatostatin
`
`Evergeen Ex. 1028
`1 of 10
`
`

`

`2
`
`Journal of Oncology
`
`analogues have been used for both diagnosis and treatment
`of NETs. 111In-labeled SST-analogues SPECT and 68Ga SST-
`analogues PET/CT represent an accurate methods for NETs
`diagnosis peptide radioreceptor therapy (PRRT) indication
`and patients management [5–8].
`When beta-emitters isotopes as 90Y (T1/2 of 2.67 days,
`maximum range of tissue irradiation of 12 mm) or 177Lu
`(T1/2 of 6.73 days, maximum range of irradiation of 1.5 mm)
`are used to label SST-analogues linked to a chelator, PRRT
`may be performed. After the i.v. injection, the radiophar-
`maceutical will distribute in the body, selectively bind to
`SSTRs, and actively be taken up by the cells through a
`process called receptor-ligand internalization [9, 10]. The
`internalization will ultimately lead to a selective accumu-
`lation of radioactivity in the tumor, thus determining cell
`death. The majority of clinical trials data available is from
`non-randomized retrospective case series. Due to variation
`in patients selection, dosing, scheduling, and total number
`of treatments it can be challenging to draw firm conclusions
`from the literature. However, it seems to be a benefit for
`selected patients with response rates in the range of 40% [11–
`14].
`Here we present the results of a phase II study designed to
`treat disseminated or nonoperable NETs patients with PRRT.
`Patients demonstrated enhanced SSTR expression at PET/CT
`with 68Ga-peptide (DOTATOC/DOTATATE).
`
`2. Materials and Methods
`
`2.1. Study Design. This was a prospective nonrandomized
`single-arm clinical trial performed at the Department of
`Nuclear Medicine, Santa Maria Nuova Hospital, Reggio
`Emilia (Italy). All patients with advanced, progressive NET
`fulfilling the study inclusion criteria were first evaluated
`with 68Ga-peptide PET/CT followed by 111In-peptide dosi-
`metric evaluation to determine both the presence of SSTR
`expression as a target for the following treatment and
`eligibility to PRRT, that is in presence of provisional adsorbed
`doses: (a) >10 Gy to tumor, (b) <10 Gy to the kidneys,
`(c) <6 Gy for the liver, (d) <1.5 Gy for red marrow, (e)
`<3 Gy for lung, and (f) <8 Gy for whole body. In case
`of 177Lu-PRRT (177Lu-DOTATOC/DOTATATE), dosimetric
`evaluation was performed acquiring images during the first
`cycle of therapy. A fractionated treatment protocol was
`followed with the intravenous administration of an average
`activity of 2.6 GBq/cycle for 90Y-PRRT and 6.0 GBq/cycle for
`177Lu-PRRT, respectively, with an interval of about 2 months.
`Toxicity and tolerability were recorded through all the
`study and for additionally 6 months after the study com-
`pletion. Serial follow-up 68Ga-peptide PET/CT imaging was
`repeated after each PRRT cycle during the first part of the
`study as required by our ethic committee. The clinical trial
`was subsequently amended and the number of PET/CT
`examinations reduced to baseline, intermediate (after 2-3
`PRRT cycles), and end-treatment (3–6 months after the
`last PRRT) scans. In order to homogenize data analysis,
`treatment response was assessed comparing PET/CT studies
`performed at baseline and at the end of treatment as well
`
`as patient’s clinical response. The intermediate PET/CT
`evaluation was used only to assess the early progressive
`disease (PD).
`The study was conducted in accordance with Interna-
`tional Conference on Harmonization Good Clinical Practice
`guidelines, the Declaration of Helsinki and it was approved
`by local and national authorities (EudraCT numbers 2006-
`000897-65 and 2008-000983-17).
`
`2.2. Patients. From January 2007 to August 2011, we enrolled
`65 patients (38 men and 27 females; mean age = 65 years,
`range 33–83). All patients presented progressive disease and
`fulfilled the following inclusion/exclusion criteria.
`
`2.2.1. Inclusion Criteria. The inclusion criteria were as fol-
`lows:
`
`inoperable or
`
`(i) Age > 18 years;
`(ii) histological confirmation of NET;
`metastatic disease;
`(iii) presence of at least one measurable lesion;
`(iv) positive 68Ga-peptide PET/CT defined as radiophar-
`maceutical uptake in tumor and/or metastasis higher
`than liver, evaluated within 3 months before PRRT
`(qualitative analysis);
`(v) adequate hematological parameter: hemoglobin level
`(Hb) ≥ 10 g/dL; leucocytes (WBC) ≥ 2.5 × 103/mL;
`platelets (PLT) ≥ 100 × 103/mL;
`(vi) adequate liver and renal function: bilirubin levels
`<2.5 mg/dL; creatinine levels <2 mg/dL;
`(vii) ECOG performance status <2;
`(viii) Signed informed consent;
`(ix) discontinuation of cold SST-analogues treatment at
`least 4 weeks before PRRT;
`(x) Life expectancy of at least 6 months.
`
`2.2.2. Exclusion Criteria. The exclusion criteria were as fol-
`lows:
`
`(i) other treatment (such as chemotherapy or radiother-
`apy) or participation in any investigational drug trial
`within 1 month of PRRT and for the following 2
`months;
`(ii) Pregnancy or lactation;
`(iii) Bone marrow involvement >25%;
`(iv) other concomitant tumors, except “in situ” basal cell
`carcinoma and tumors of the uterine cervix treated
`with radical surgery.
`
`Additionally, before each PRRT cycle the following
`parameters should be maintained: Hb ≥ 10 g/dL, WBC ≥
`2.5 × 103/mL; PLT ≥ 100 × 103/mL, creatinine levels
`<2 mg/dL; bilirubin levels <2.5 mg/dL.
`The final analysis was based on a total of 59 patients
`(m/f 33/26) since 6 patients (2 with GI tumor, 1 with
`
`Evergeen Ex. 1028
`2 of 10
`
`

`

`Journal of Oncology
`
`3
`
`carcinoid tumor of the lung, and 3 with pancreatic tumor)
`voluntarily interrupted the treatment. Tumor was localized
`in the gastrointestinal tract in 19/59 cases (32%), followed by
`pancreas in 16/59 cases (27%) and lung in 13/59 cases (22%).
`In 11/59 patients (19%), the origin was unknown.
`All patients at enrollment had metastatic (stage IV) PD
`(Table 1). Histopathological findings including grading were
`not reported for patients since histological diagnosis was
`performed in different centers thus features were reported
`in different not comparable modalities. Previous treatments
`are reported in Table 2. Diabetes was present in 11/59
`cases and 16/59 patients suffered from blood hypertension.
`Additionally, 9/59 had previous tumors (3/9 prostate cancers,
`3/9 breast cancers, 2/9 large-bowel cancers and 1/59 stomach
`cancer) with a minimal time of free disease of 5 years. Main
`baseline clinical signs and symptoms were diarrhea (18/59),
`pain (12/59), weight loss (7/59), flush (5/59), cough (4/59),
`constipation (3/59), nausea (2/59), and carcinoid syndrome
`(1/59). Additionally, 32/59 patients presented at enrolment
`a variable grade of asthenia. Twenty-seven patients were
`asymptomatic at baseline. Serum baseline CgA levels were
`normal in 19/59 patients.
`
`90Y-, and
`2.3. Radiopharmaceuticals Preparation. 111In-,
`177Lu-peptide (DOTATOC or DOTATATE) were synthesized
`by following internal protocol [15]. Every preparation
`was obtained by carrying out the following steps: (a) a
`3 mL syringe was filled with a 1 mL solution containing
`30 μg of sodium ascorbate and an amount of a 4 mg/mL
`peptide solution proportional to the 90Y-, and 177Lu- or
`111In- activity in order to achieve a radiolabeling specific
`activity of 106 MBq/nmol, 48 MBq/nmol, and 6 MBq/nmol,
`respectively (b) this solution was added to a 3 mL Schott vial
`containing an activity ranging between 7.4 to 30 GBq of 90Y
`chloride solution, between 15 to 60 GBq of 177Lu chloride
`solution or between 222 to 444 MBq of 111In- chloride
`solution (Perkin Elmer, Boston, MA, United States) in 0.05 M
`hydrochloric acid obtaining a 4.6 pH solution; (c) the Schott
`◦
`C in a heating block;
`vial was heated for 30 minutes at 90
`(d) a 5 μL aliquot of the solution was withdrawn for carrying
`out the quality controls by using solid phase extraction or
`chromatographic methods [16]; (e) only for 90Y and 177Lu-
`peptide: the preparation was transferred to a bigger vial
`containing 0.5 mL of 1 mM DTPA solution and diluted with
`20 mL of 0.9% sodium chloride solution [17]; (f) single doses
`for the patients were obtained by fractioning the mother
`solution in vials containing 2 mL of an ascorbic acid/sodium
`ascorbate buffer solution in order to decrease the effects of
`radiolysis. The radiochemical purity of the 111In-, 177Lu-, and
`90Y-peptide preparations was always >99.8%.
`The radiolabeling of 68Ga-peptide was performed by
`means of a modular lab synthesizer (Eckert & Ziegler, Berlin,
`Germany) as already described [18]. Briefly, the fraction of
`about 2 mL of the 68Ge/68Ga-generator eluate containing
`about 80% of the 68Ga activity in 0.1 M hydrochloric acid
`was selected and directed to a reactor vial containing a
`20 μL of peptide solution (1 mg/mL) and 200 μL of a 1.5 M
`sodium formate solution or 140 μL of a 1.5 M sodium acetate
`solution in order to obtain a pH ranging between 3.2 and
`
`Table 1: Site and number of metastasis in the 59 evaluated patients
`at the enrollment in the clinical trial.
`
`Site of metastasis
`
`Bone (21/59)
`Liver (42/59)
`Lung (4/59)
`Lymph nodes (34/59)
`Other (6/59)
`
`≤5
`0/21
`2/42
`0/4
`3/34
`1/6
`
`Number of metastasis
`>5
`21/21
`40/42
`4/4
`31/34
`5/6
`
`Table 2: List of previous treatments in the 59 evaluated patients
`order on the basis of their frequency.
`
`Number of patients
`Previous treatment
`39/59
`Surgery
`25/59
`“Cold” SST analogues
`13/59
`Chemotherapy
`7/59
`TACE or RFTA
`5/59
`External beam radiotherapy
`TACE: intra-arterial hepatic chemoembolization; RFTA: radiofrequency
`thermoablation.
`
`◦
`C for 5 minutes and,
`3.5. The mixture was heated at 100
`then, passed through a light C-18 cartridge. 68Ga-peptide was
`eluted with 0.5–1 mL of a 50% ethanol solution and diluted
`with 8 mL of 0.9% sodium chloride solution. The synthesis
`was carried out in 14 minutes with a mean yield of 63 ± 3%
`(not corrected for decay). Quality controls were performed
`by chromatographic methods as already described, obtaining
`a radiochemical purity always >95% [19].
`
`2.3.1. Pretherapeutic Somatostatin Receptor Imaging. Pre-
`imaging was performed by 68Ga-peptide
`therapeutic
`PET/CT. For this study, PET/CT scans were acquired on a
`GE Discovery at 60 min after injection of about 120 MBq
`of 68Ga-peptide. Seven or eight bed positions with 5 slices
`overlap were acquired for 4 min emission time in 3D. The
`CT-exposure factors for all examinations were 120 kVp and
`80 mA in 0.8 seconds. PET images were reconstructed using
`CT-attenuation correction (OSEM). All studies were visually
`and semiquantitatively assessed. SUV calculations were
`performed on a Xeleris workstation. Mean and maximum
`SUV (activity concentration corrected for patient weight
`and total injected dose) was determined in all lesions and
`recorded.
`
`2.3.2. Selection of Patients Eligible for PRRT. 68Ga-peptide
`PET/CT was considered positive in patients who showed
`uptake in the tumor lesions at least two-times higher than
`the liver; thus they were considered eligible for PRRT and,
`therefore, admitted to dosimetric evaluation.
`
`2.4. Dosimetry. Planar imaging was initially performed after
`the i.v. injection of 185 MBq of 111In-peptide with a dual-
`head gamma camera (Genesys, Philips, The Netherlands)
`
`Evergeen Ex. 1028
`3 of 10
`
`

`

`4
`
`Journal of Oncology
`
`using parallel-hole, medium-energy, general-purpose colli-
`mators. The windows were centered over both 111In- photon
`peaks (247 and 172 keV with a window width of 20%),
`whereas scatter fraction was evaluated at 140 keV (width
`20%).
`In all the patients, whole-body scan and, in selected cases,
`spot images of the abdomen were obtained after 1, 4, 20, 48,
`and 72 hours for control of biodistribution. To determine
`blood clearance, we drew blood samples at 30 and 60 minutes
`and at 4, 20, and 48 hours after injection. Radioactivity in
`blood was measured with a HPGe spectrometer (DSPEC jr
`2.0—Ortec). For dosimetric calculations, regions of inter-
`est were drawn manually on the whole-body scans from
`anterior and posterior projections and ULMDOS software
`(University of Ulm, Germany) was used. Background regions
`were placed on the abdomen or on the thigh for back-
`ground correction. Scans were corrected for background,
`self-absorption, patient thickness attenuation, and organ
`overlapping. Whole-body activity acquired immediately after
`injection was defined as 100% of the injected activity. Data
`were expressed as percentage injected activity as a function
`of time. The resulting time-activity points were fitted to a
`monoexponential or multiexponential curve for whole-body,
`kidneys, liver, spleen, and red marrow to calculate residence
`time. Patient-specific organ masses were also considered.
`The estimated doses delivered to critical organs and to the
`tumor were obtained by the software OLINDA/EXM [20].
`The activity in blood was fitted to a biexponential curve
`to determine the residence time in blood. The dose to
`the red marrow was calculated from the residence time in
`blood, assuming no specific uptake, a uniform distribution
`of activity, and clearance from red marrow equal to that from
`blood. A correction factor of 1 was used as described by
`Cremonesi et al. [21].
`In case of 177Lu-PRRT the dosimetric evaluation was
`performed acquiring images during the first cycle of therapy,
`thanks to the low gamma emission of this isotope.
`
`2.5. Therapy (Administration Protocol). A fractionated treat-
`ment protocol was followed with the intravenous adminis-
`tration of an average activity of 2.6 GBq and 6.0 GBq per
`cycle for 90Y-PRRT and for 177Lu-PRRT, respectively, with an
`interval of about 2 months. For each cycle, patients were hos-
`pitalized for 3 days in accordance with local requirements.
`Thirty minutes before administration of the radiopeptide
`2 L of amino acid solution of Hartmann-Hepa 8 (Ringer’s
`Lactate Hartmann, Proteinsteril Hepa 8%, Mg 5-sulfat) were
`infused, which were continued up to 3 hours after injection
`to inhibit tubular reabsorption of the radioactive tracer.
`Repeated treatments were performed in case of response and
`significant improvement in symptoms and quality of life,
`except in cases of renal toxicity and rejection by the patient
`for further treatment within 3 months. Additional cycles
`were suspended in case of PD.
`
`SPECT gamma camera (Genesys, Philips, The Netherlands)
`or with a dual-head SPECT/CT gamma camera (Symbia-
`T, Siemens, Germany) using parallel-hole, medium-energy,
`general-purpose collimators. The windows were centered
`over 177Lu-PRRT photon peaks (208 keV and 110 keV width
`20% in both cases; scatter window at 160 keV) in case
`of treatment with 177Lu-PRRT; while at 170 keV (20%)
`and 80 keV (55%) in case of treatment with 90Y-PRRT, as
`Bremsstrahlung planar scan. Whole-body scans (acquisition
`time: 25 minutes) and spot images (acquisition time: 10
`minutes) were obtained.
`
`2.7. Assessment of Clinical Benefit and Evaluation of PRRT
`Response. Clinical benefit was assessed comparing baseline
`clinical conditions with end-treatment parameters. In the
`clinical benefit evaluation the worsening of clinical con-
`ditions (i.e., appearance of new sign(s)/symptom(s)) were
`considered as PD. Indeed any significant variations in base-
`line clinical conditions was defined as stable disease (SD).
`Clinical benefit was defined as non-PD/SD. For the follow-
`up blood tests were evaluated, as described in the clinical
`protocol, repeated before and after each treatment cycle
`and every two weeks. Blood tests included hematological
`parameters,
`liver and renal function. Baseline and end-
`treatment serum CgA values were compared and the trend
`was defined as increased, stable (variation over time ≤10%)
`or decreased. All patients were followed for an additional
`6 months after the last radiopharmaceutical administration.
`Acute and long-term adverse events were graded according
`to the Common Terminology Criteria for Adverse Events,
`version 3.0 of the National Cancer Institute [22]. To assess
`response to treatment PET/CT studies performed at baseline
`and at the end of treatment were considered.
`Treatment responses assessed by PET/CT scan were
`defined as follows:
`
`(i) complete response (CR): disappearance of radiophar-
`maceutical uptake in all detectable lesions;
`(ii) partial response (PR): reduction of radiopharma-
`ceutical uptake (>50%) in all detectable lesions in
`absence of appearance of new lesion(s);
`(iii) stable disease (SD): no variation or reduction
`of radiopharmaceutical uptake (<50%) in some
`detectable lesions in absence of appearance of new
`lesion(s);
`(iv) progressive disease (PD): increase >25% of radio-
`pharmaceutical uptake in one or more lesions or
`appearance of new lesions and/or >10% increasing of
`tumor marker.
`
`In this series of patients, we did not assess treatment
`response based on the size of lesions using the CT component
`of PET/CT images or CT scan, but as described above we
`evaluate only the functional response.
`
`2.6. Biodistribution of the Radiotracer. In order to evalu-
`ate the biodistribution of therapeutic activity, after each
`treatment, planar imaging was performed with a dual head
`
`2.8. Statistical Analysis. All values are expressed as median
`and range, as customary for nonparametric data.Correlation
`analysis was performed using the Mann-Whitney test.
`
`Evergeen Ex. 1028
`4 of 10
`
`

`

`Journal of Oncology
`
`5
`
`from previous chemotherapies. Asthenia (grade 2-3, 28/59)
`nausea (grade 1-2, 14/59), vomiting (grade 2-3, 5/59), were
`frequently observed. Stomatitis (grade 2) and gastritis (grade
`1) were also reported in 1 case each. Long-term nephro-
`toxicity was observed in 3 patients (2 moderate; 1 severe
`requiring dialysis). Patients who developed nephrotoxicity
`were treated with 90Y-PRRT receiving 5 (2/3) and 6 (1/3)
`cycles. One of them suffered from both diabetes and blood
`hypertension. Patient who required dialysis had only one
`kidney. Clinical benefit was recorded in 21/59 patients while
`a worsening of clinical conditions was observed in 9/59
`patients. All patients which were asymptomatic at baseline
`and not present modifications of their clinical conditions.
`Best objective response was CR in 1/59 patient (2%), PR
`in 24/59 (40.5%), SD in 24/59 patients (40.5%) while PD
`was demonstrated in 10/59 (17%) of patients. The overall
`tumor response rate considering both CR and PR was 42.5%.
`SUVmax values in the main lesion for both baseline and
`end-treatment 68Ga-peptide PET/CT scans were reported in
`Table 4 based on functional response. A significant difference
`in cumulated administered activity between PD and non-PD
`patients was found as shown by Figure 4.
`Table 5 shows treatment responses based on primary
`tumor site. Table 6 shows results of treatment responses
`based on the type of treatment (90Y-PRRT, 177Lu-PRRT,
`or combined 90Y-PRRT and 177Lu-PRRT) while treatment
`responses based on the numbers of PRRT cycles are reported
`in Table 7. In Table 8 functional response was tabulated
`on the basis of clinical benefit assessment. In the eleven
`patients with both normal baseline and end-treatment serum
`CgA levels functional response assessed by 68Ga-peptide
`PET/CT resulted in 1/11 CR, 5/11 PR, 4/11 SD, and 1/11
`PD. Discordant results between serum CgA levels trend
`over time and 68Ga-peptide results were found in 23/59
`patients. Despite the increase of CgA values 68Ga-peptide
`PET/CT documented a PR in 7 patients and a SD in 6 cases,
`respectively (Table 9). In one patient classified as SD by 68Ga-
`peptide PET/CT, serum CgA levels completely normalized
`after PRRT.
`
`4. Discussion
`
`The development of imaging agents specifically designed to
`target tumor metabolic pathways and associated antigens
`including membrane receptors opens new horizon both for
`the selection of patients candidate to target treatment by the
`in vivo detection of enhanced target expressions as well as for
`the development of new multimodality treatment strategies.
`The expression of SSTRs by NETs made molecular imag-
`ing with specific SST-analogues for specific SSTR subtypes
`the method of choice for their diagnostic workup. In fact
`111In-labeled SST analogues scintigraphy and more recently
`68Ga-DOTA-peptides significantly change the diagnostic
`approach to neuroendocrine tumors. In our study, 68Ga-
`peptide PET/CT as first-selection procedure to determine the
`presence of high SSTR expression and a tumor uptake at least
`two times higher than the liver were considered the criteria
`to be eligible for dosimetric evaluation with 111In-peptide.
`
`Evergeen Ex. 1028
`5 of 10
`
`Figure 1: 68Ga-DOTATOC PET/CT: liver, lung, lymph node, and
`bone metastases from NET of unknown origin.
`
`3. Results
`
`68Ga-DOTATOC/DOTATATE PET/CT was performed in
`all the patients to evaluate the eligibility. Baseline 68Ga-
`DOTATOC/DOTATATE PET/CT demonstrated at least one
`site of radiopharmaceutical uptake.
`Figure 1 shows an example of 68Ga-DOTATOC accu-
`mulation in tumor lesions. 68Ga-DOTATOC positive lesions
`were preferentially localized at liver, lymph nodes, lung, and
`skeleton.
`An end-treatment 68Ga-peptide PET/CT was performed
`in all treated patients about 3–6 months after the last
`PRRT administration except for 6 patients in which PD was
`determined on the basis of worsening of clinical conditions.
`Figures 2 and 3 represent examples of pre- and postther-
`apeutic 68Ga-DOTATOC PET/CT.
`Dosimetric estimates for kidney and bone marrow are
`summarized in Table 3. No toxicities were recorded after
`radiopharmaceutical injection administered for dosimetric
`purpose.
`PRRT cycles were administered at 70 ± 24.6 days apart
`(range 35–140) with a median cumulative activity of 5.5 GBq
`(range 3.6–7.4 GBq). Thirty-five patients received 4 or 5
`PRRT cycles, 10/59 more than 5 cycles while 14/59 patients
`had <4 PRRT cycles. 90Y-PRRT was administered in 33/59
`patients (56%), 177Lu-PRRT in 9/59 patients (15%) while
`17/59 patients (29%) received both 90Y-PRRT and 177Lu-
`PRRT in different cycles. Posttherapeutic scintigraphy con-
`firmed a correct distribution of the radiopharmaceutical in
`all patients.
`In 18/59 (30%) patients no adverse effects after adminis-
`tration of the radiopharmaceuticals were observed. Hema-
`tological toxicity including grade 2-3 anemia, thrombocy-
`topenia and leukopenia occur in 12/59 patients (20%). Two
`of the 12 patients who had hematological toxicity presented
`baseline grade 1 anemia and thrombocytopenia resulting
`
`

`

`6
`
`Journal of Oncology
`
`Figure 2: Male, 56 years old, with pancreatic NET and multiple liver metastases. 68Ga-DOTATOC PET/CT before therapy (a) and after
`PRRT (b). The result was a partial response.
`
`(a)
`
`(b)
`
`Figure 3: Male, 73 year old, pancreas NET with liver metastases. 68Ga-DOTATOC PET/CT before (left) and after therapy (right). PRRT with
`90Y-DOTATOC (2 cycles) and 177Lu-DOTATOC (4 cycles) was administered at interval of 2 months. The response was complete in the liver
`but partial in the pancreatics region (arrow).
`
`(a)
`
`(b)
`
`Table 3: Dosimetric estimates for kidney and bone marrow.
`
`90Y-kidney dose (Gy/GBq)
`90Y-bone marrow dose (Gy/GBq)
`177Lu-kidney dose (Gy/GBq)
`177Lu-bone marrow dose (Gy/GBq)
`
`Mean
`2.4E + 00
`9.7E − 02
`3.9 E − 01
`2.81E − 02
`
`Median
`1.5E + 00
`5.8E − 02
`2.5E − 01
`1.29E − 02
`
`SD
`1.9
`0.1
`0.3
`0.04
`
`Range
`0.32–8.90
`0.0047–0.51
`0.05–1.47
`0.0163–0.256
`
`Evergeen Ex. 1028
`6 of 10
`
`

`

`Journal of Oncology
`
`7
`
`9
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Cumulated administered activity (GBq)
`
`9
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Cumulated administered activity (GBq)
`
`PD
`
`Non-PD
`
`PD
`
`Objective response
`
`(a)
`
`(b)
`
`9
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Cumulated administered activity (GBq)
`
`PD
`
`Objective response
`
`SD
`
`(c)
`
`Figure 4: (a) Mann-Whitney test plot shows a significant difference in cumulated administered activity between PD and non-PD patients
`(P < 0.001). Similar results (b) have been obtained by using Mann-Whitney test excluding patients who had SD and thus considering only
`patients who had objective response (P = 0.002). When all subgroups of patients are considered (c), the cumulated administered activity
`remains significantly different in PD group versus both objective response and SD groups while no significant differences have been found
`in cumulated administered activity comparing patients who presented objective response and stable disease (P = 0.435). PD = patients who
`presented progressive disease; non-PD = patients who presented complete or partial response or disease stabilization; objective response =
`patients who presented complete or partial response; SD = patients who presented stable disease.
`
`Originally, the study was designed for radiolabelled DOTA-
`TOC but because of problem of commercial availability we
`were obliged to amend the study protocol and substitute
`DOTATOC with DOTATATE. Using 68Ga-peptide PET/CT
`high SSTR expression to be eligible for subsequent PRRT
`was found in our study in all patients. Dosimetric estimates
`confirmed the eligibility of all patients and demonstrated
`that a fractionated treatment protocol with the intravenous
`administration of an average activity of 2.6 GBq/cycle for 90Y-
`PRRT and 6.0 GBq/cycle for 177Lu-PRRT, respectively, with
`an interval about 2 months was within the safety threshold
`of toxicity, particularly for the kidney (the critical organ for
`PRRT) and the bone marrow. Initially for PRRT, we used
`90Y since in our department 177Lu was allowed from 2009.
`Subsequently, the criteria of choice of radionuclide used for
`PRRT was mainly based on tumor size [23] (reserving 90Y
`
`for lesion(s) >2 cm, 177Lu for lesion(s) <2 cm, and 90Y/177Lu
`in presence of both conditions) and on dosimetric estimates.
`Administration of 90Y-PRRT (average activity of about
`2.6 GBq/cycle) and 177Lu-PRRT (average activity of about
`6.0 GBq/cycle) induced disease control in 83% of patients (1
`CR, 24 PR, and 24 SD) with a duration of response of at least
`6 months. In the majority of cases, objective response was
`associated to symptomatic response with an improvement of
`quality of life.
`These responses rates are comparable with data from
`literature [11, 24, 25] demonstrating radiological response
`of 34.1% and clinical response in 29.7% for 90Y-PRRT
`with longer median survival in responders compared to
`nonresponders (44.7 versus 18.3 months) and response
`rates of up to 30% with median time to progression of
`40 months for 177Lu-PRRT [26]. Interestingly,
`in these
`
`Evergeen Ex. 1028
`7 of 10
`
`

`

`8
`
`Journal of Oncology
`
`Table 4: Baseline and end-treatment SUV max values recorder for all patients in the main lesion assessed by 68Ga-peptide PET/CT tabulated
`on the basis of patients’ functional response.
`
`Baseline SUV max value
`Range
`Median
`Mean
`Functional response
`—
`45.3
`45.3
`CR
`6.3–119.9
`29.2
`79.7
`PR
`6.6–82.0
`20.9
`31.1
`SD
`PDc¸
`11.7–55.8
`22
`27.9
`Data from the 4 patients in which PD was assessed using 68Ga-peptide PET/CT.
`
`c¸
`
`Main lesion
`
`End-treatment SUV max value
`Median
`Range
`0.9
`—
`17.8
`3.2–49.1
`28.2
`11.2–61.3
`26
`15.7–74.7
`
`Mean
`0.9
`39.2
`31.2
`35.7
`
`Table 5: Results of treatment responses tabulated on the basis of primary tumor site.
`
`Site of primitive tumor
`GI (19/59)
`Pancreas (16/19)
`Lung (13/59)
`Unknown origin (11/59)
`
`CR
`—
`1/16 (6%)
`—
`—
`
`PR
`8/19 (42%)
`5/16 (31%)
`8/13 (62%)
`3/11 (27%)
`
`SD
`9/19 (47%)
`6/16 (38%)
`3/13 (23%)
`6/11 (55%)
`
`PD
`2/19 (11%)
`4/16 (25%)
`2/13 (15%)
`2/11 (18%)
`
`studies the degree of uptake on the pretreatment 111In-
`peptide was found to be predictive of response to treatment
`and overall survival. Despite the small number of patients,
`combined treatments with labeled peptide using both 90Y
`and 177Lu seem to perform better (no evidence of PD)
`when compared to “single radionuclide” PRRT (8 and 2
`cases of PD administering only 90Y-peptide and 177Lu-
`peptide, resp.). In our patients population, the SUVmax
`value in tha main lesion at baseline 68Ga-peptide PET/CT
`examination compared to end-treatment scan was in line
`with the functional response evaluation. Additionally, in
`our series of patients, cumulated administered activity was
`significantly different in responders and no-responders. In
`agreement with previous reports in literature [25] our data
`supported the hypothesis that progression at baseline could
`be a prognostic factor of objective response to PRRT. Indeed
`previous reports showed also that both SD and objective
`response (CR+PR) in previously progressive patients showed
`the same favorable trend [25]. Finally, PRRT showed bene-
`ficial effect on symptoms in the majority of patients (36%)
`and all asymptomatic patients (46%) remained stable over
`the time. In 39% of our patients discordant results between
`serum CgA trend and 68Ga-peptide findings were observed.
`Particularly, CgA values increased (with variation up to
`+263%) in 68% of patients in which PRRT determined PR
`or SD. Our results on CgA trend and antitumor activity are
`in contrast with previous reported ones in the literature [25].
`However, the majority of our patients assumed proton-pump
`inhibitor as prophylactic therapy and as well known these
`drugs may cause substantial increase of blood CgA levels
`[27] underling the need of a more reliable biomarker to
`monitoring NETs [28].
`The prevalence of partial responses and stable disease
`obtained is mainly related to the advanced stage of the disease
`wh

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