throbber
Eur J Nucl Med Mol Imaging
`DOI 10.1007/s00259-012-2330-6
`
`GUIDELINES
`
`The joint IAEA, EANM, and SNMMI practical guidance
`on peptide receptor radionuclide therapy (PRRNT)
`in neuroendocrine tumours
`
`John J. Zaknun & L. Bodei & J. Mueller-Brand &
`M. E. Pavel & R. P. Baum & D. Hörsch & M. S. O’Dorisio &
`T. M. O’Dorisiol & J. R. Howe & M. Cremonesi &
`D. J. Kwekkeboom
`
`# The Author(s) 2013. This article is published with open access at Springerlink.com
`
`Abstract Peptide receptor radionuclide therapy (PRRNT) is a
`molecularly targeted radiation therapy involving the systemic
`administration of a radiolabelled peptide designed to target with
`high affinity and specificity receptors overexpressed on
`tumours. PRRNT employing the radiotagged somatostatin re-
`ceptor agonists 90Y-DOTATOC ([90Y-DOTA0,Tyr3]-octreotide)
`or 177Lu-DOTATATE ([177Lu-DOTA0,Tyr3,Thr8]-octreotide or
`[177Lu-DOTA0,Tyr3]-octreotate) have been successfully used
`for the past 15 years to target metastatic or inoperable neuro-
`endocrine tumours expressing the somatostatin receptor
`
`subtype 2. Accumulated evidence from clinical experience
`indicates that these tumours can be subjected to a high absorbed
`dose which leads to partial or complete objective responses in
`up to 30 % of treated patients. Survival analyses indicate that
`patients presenting with high tumour receptor expression at
`study entry and receiving 177Lu-DOTATATE or 90Y-DOTA-
`TOC treatment show significantly higher objective responses,
`leading to longer survival and improved quality of life. Side
`effects of PRRNT are typically seen in the kidneys and bone
`marrow. These, however, are usually mild provided adequate
`
`L. Bodei
`Division of Nuclear Medicine, European Institute of Oncology,
`Milan, Italy
`
`J. J. Zaknun (*)
`Nuclear Medicine Section, Division of Human Health,
`International Atomic Energy Agency, IAEA, Vienna, Austria
`e-mail: J.zaknun@hotmail.com
`
`J. J. Zaknun
`e-mail: John.Zaknun@Zentralklinik.de
`
`J. J. Zaknun
`Zentralklinik Bad Berka, Center for Molecular Radiotherapy and
`Molecular Imaging, ENETS Center of Excellence, Bad Berka,
`Germany
`
`J. Mueller-Brand
`Klinik und Institut für Nuklearmedizin, Universitätsspital Basel,
`Basel, Switzerland
`
`M. E. Pavel
`Campus Virchow Klinikum, Klinik für Gastroenterologie,
`Hepatologie, Endokrinologie, Diabetes und
`Stoffwechsel-erkrankungen, Charité Universitaetsmedizin Berlin,
`Berlin, Germany
`
`R. P. Baum : D. Hörsch
`Zentralklinik Bad Berka, Department of Internal Medicine,
`Gastroenterology and Endocrinology, ENETS Center of
`Excellence, Bad Berka, Germany
`
`M. S. O’Dorisio
`RJ and LA Carver College of Medicine, Department of Pediatrics,
`University of Iowa, Iowa City, IA, USA
`
`T. M. O’Dorisiol
`RJ and LA Carver College of Medicine, Department of Internal
`Medicine, University of Iowa, Iowa City, IA, USA
`
`J. R. Howe
`RJ and LA Carver College of Medicine, Department of Surgical
`Oncology, University of Iowa, Iowa City, IA, USA
`
`M. Cremonesi
`Service of Health Physics, European Institute of Oncology, Milan,
`Italy
`
`D. J. Kwekkeboom
`Department of Nuclear Medicine, Erasmus Medical Center
`Rotterdam, Rotterdam, The Netherlands
`
`Evergeen Ex. 1034
`1 of 17
`
`

`

`protective measures are undertaken. Despite the large body of
`evidence regarding efficacy and clinical safety, PRRNT is still
`considered an investigational treatment and its implementation
`must comply with national legislation, and ethical guidelines
`concerning human therapeutic investigations. This guidance
`was formulated based on recent literature and leading experts’
`opinions. It covers the rationale, indications and contraindica-
`tions for PRRNT, assessment of treatment response and patient
`follow-up. This document
`is aimed at guiding nuclear
`medicine specialists in selecting likely candidates to receive
`PRRNT and to deliver the treatment in a safe and effective
`manner. This document is largely based on the book pub-
`lished through a joint international effort under the auspices
`of the Nuclear Medicine Section of the International Atom-
`ic Energy Agency.
`
`Keywords Peptide receptor radionuclide therapy . PRRNT .
`PRRNT, neuroendocrine tumours, guideline/s, renal protection
`
`Purpose
`
`This guidance document is aimed at assisting and guiding
`nuclear medicine specialists in:
`
`1. Assessing patients with neuroendocrine tumours (NETs)
`for their eligibility to undergo treatment with 90Y- or
`177Lu-radiolabelled somatostatin analogues.
`2. Providing guidance on performing peptide receptor ra-
`dionuclide therapy (PRRNT) and implementing this
`treatment in a safe and effective manner.
`3. Understanding and evaluating the outcome of PRRNT,
`namely treatment results and possible side effects in-
`cluding both renal and haematological toxicities.
`
`A committee of international experts was assembled
`under the auspices of the International Atomic Energy
`Agency (IAEA), in cooperation with the EANM Thera-
`py, Oncology and Dosimetry Committees and with the
`Society of Nuclear Medicine and Molecular Imaging.
`They worked together to create this guidance document
`on the use of somatostatin analogue-based PRRNT. This
`guidance document was compiled taking into account
`recent literature and experts’ opinion.
`
`Regulatory issues
`
`Applicable in all countries Clinicians involved in unsealed
`source therapy must be knowledgeable about and compliant
`with all applicable national and local legislation and
`regulations.
`
`Applicable in the USA The radiopharmaceuticals used for
`the diagnostic and therapeutic procedures addressed in this
`
`Eur J Nucl Med Mol Imaging
`
`guidance document are not approved by the Food and Drug
`Administration (FDA) in the USA. Therefore in the USA
`these procedures should be performed only by physicians
`enrolled in an investigational protocol pursuant to a valid
`Investigational New Drug application or Radioactive Drug
`Research Committee approval and under the purview of an
`appropriate institutional review board.
`
`Background information and definitions
`
`Definitions
`
`PRRNT
`
`Somatostatin
`
`Somatostatin
`receptors
`
`PRRNT (or PRRT) involves the systemic
`administration of a specific well-defined
`radiopharmaceutical composed of a β-
`emitting radionuclide chelated to a pep-
`tide for the purpose of delivering cyto-
`toxic radiation to a tumour. The
`oligopeptides are designed to target cel-
`lular proteins, commonly cell surface
`receptors, such as the somatostatin recep-
`tor (sstr) subtype 2 (sstr2) that is overex-
`pressed on the cell surface of NETs in a
`tumour-specific fashion, thereby ensuring
`a high level of specificity in the delivery
`of the radiation to the tumour. Hence,
`PRRNT is a molecularly targeted radia-
`tion therapy, and thus is distinct from ex-
`ternal beam radiation therapy.
`The naturally occurring somatostatin
`molecule is an oligopeptide comprising
`either 14 or 28 amino acids with a
`limited half-life in blood due to rapid
`enzymatic degradation. Somatostatin
`exerts an antisecretory endocrine and
`exocrine effect in addition to tumour
`cell-growth inhibition. Stabilized ana-
`logues of somatostatin (SSA) show
`prolonged duration of action [1].
`In humans five sstr subtypes have been
`identified. Each sstr is a transmembrane
`molecule weighing approximately
`80 kDa. Somatostatin exerts its action by
`inhibiting G-protein-dependent 3′,5′-
`cyclic monophosphate (cAMP) formation
`in a dose-dependent manner at
`subnanomolar concentrations. Sstr2 is
`overexpressed in NETs. Sstr2 is the key
`target molecule for both cold and
`radiolabelled SSA. Upon binding to its
`receptor, the complex (SSA-sstr) under-
`goes cellular internalization thereby
`
`Evergeen Ex. 1034
`2 of 17
`
`

`

`Eur J Nucl Med Mol Imaging
`
`Yttrium-90
`
`Lutetium-177
`
`DOTATOC
`
`DOTATATE
`
`enhancing the therapeutic effect of the
`radiolabelled drug [2].
`The radiometal 90Y is a pure β-emitting
`isotope with a physical half-life of 64 h.
`The maximum and mean β-particle ener-
`gies are 2.28 MeV and 0.934 MeV, re-
`spectively. The maximum and mean β-
`particle penetration depths in soft tissue
`are 11 mm and 3.9 mm, respectively.
`177Lu is a β- and γ-emitting radionuclide
`with a physical half-life of 162 h (6.73
`days). Compared to 90Y, 177Lu has lower
`maximum and mean β-particle energies
`(0.498 MeVand 0.133 MeV, respectively).
`These translate to maximum and mean
`soft-tissue penetration depths of 1.7 mm
`and 0.23 mm, respectively. 177Lu has two
`main gamma emission lines: 113 keV (6 %
`relative abundance) and 208 keV (11 %
`relative abundance). The latter properties
`of 177Lu allow posttreatment imaging and
`dosimetry assessments.
`DOTATOC is a derivatized somatostatin
`analogue peptide. DOTATOC is the
`abbreviated form of [DOTA0,Tyr3]-
`octreotide, where DOTA stands for the
`bifunctional chelating molecule 1,4,7,10-
`tetraazacyclo-dodecane-1,4,7,10-
`tetraacetic acid, and Tyr3-octreotide is the
`modified octreotide. This peptide shows a
`high affinity for sstr2 (IC50 14±2.6 nM),
`but lower affinities for sstr5 (IC50 393±84
`nM) and sstr3 (IC50 880±324 nM) [3].
`DOTATATE is also a derivatized
`somatostatin analogue peptide.
`DOTATATE is the abbreviated form of
`[DOTA0,Tyr3,Thr8]-octreotide or
`[DOTA0,Tyr3]-octreotate, and DOTA
`stands for the bifunctional metal-
`chelating molecule. This peptide shows
`a six- to ninefold higher affinity for
`sstr2 (IC50 1.5±0.4 nM) than DOTA-
`TOC, but has no affinity for either
`sstr5 (IC50 547±160 nM) or sstr3
`(IC50 >1,000 nM) [4].
`
`Background
`
`NETs have proven to be ideal neoplasms for PRRNT, as the
`majority of these slow-growing malignancies overexpress
`sstrs. Appropriate candidates for PRRNT are patients present-
`ing with well-differentiated or moderately differentiated neu-
`roendocrine carcinomas, defined as NETs of grade 1 or 2
`
`according to the WHO classification of 2010 [5]. The inci-
`dence of NETs has been rising over the past 30 years, partic-
`ularly those arising from the midgut and pancreas [6]. The
`incidence of NETs in the USA rose from 10.9 to 52.4 per
`million between 1973 and 2004 (SEER database). NETs can
`occur in children and young adults, being diagnosed as early as
`at the age of 5 years, while their incidence increases with age.
`The clinical presentation may vary depending on the site of
`tumour origin. About 72 % of NETs arise in gastrointestinal
`structures, 25 % are bronchopulmonary in origin, and less than
`5 % arise at other sites (e.g. thymus, breast and genitourinary
`system). Frequently, these tumours are discovered when meta-
`static or locally advanced and therefore inoperable. NETs can
`be either functioning or nonfunctioning in nature. Functioning
`tumours are associated with clinical syndromes, such as the
`carcinoid syndrome (due to the release of serotonin). Other
`secreting tumours include insulinomas (inducing hypoglycae-
`mia), gastrinomas (inducing Zollinger-Ellison syndrome),
`VIPomas (associated with the watery diarrhoea, hypokalaemia
`and achlorhydria syndrome; WDHA syndrome).
`Anatomical
`imaging of NETs should be as detailed
`and extensive as possible to provide accurate informa-
`tion about site and extent of the primary tumour, and
`the location and extent of regional and distant metasta-
`ses. An exact assessment of liver metastases and degree
`of liver involvement using ultrasonography, CT or MRI
`is central for accurate staging and for assessing the
`response to treatment [7].
`Functional imaging procedures applying sstr imaging
`using 111In-pentetreotide (OctreoScan) with SPECT or
`PET with 68Ga-labelled SSA, combined with morpho-
`logical imaging procedures, are used to collect essential
`information for staging, assessing sstr status and making
`decisions on the most appropriate therapy regimens [8,
`9]. Serial morphological examinations are mandatory to
`monitor therapy and detect recurrent disease. Emerging
`data indicate that 18F-FDG PET may have additional
`prognostic value [10]. This information, however, needs
`validation in larger studies.
`Multiple treatment approaches are now available for
`patients presenting with metastatic disease, considering re-
`cently introduced molecular targeted therapies and multi-
`modality treatment options. For the choice of the most
`appropriate treatment, information regarding anatomical lo-
`cation and local invasion of adjacent structures, tumour
`functionality, sstr status, histological grading and staging
`are required to facilitate the decision-making process within
`the multidisciplinary tumour board. If the disease is restrict-
`ed to the liver, surgical and locoregional approaches should
`be considered primarily. Chemotherapy is appropriate for
`highly proliferating NETs and pancreatic NETs, keeping in
`mind the fact that the vast majority of NETs are rather
`insensitive to this treatment.
`
`Evergeen Ex. 1034
`3 of 17
`
`

`

`Treatment options in NETs
`
`Patients with NETs may present with local tumours, with or
`without regional or distant metastases. The common site of
`metastasis is the liver. These tumours may remain clinically
`silent until a significant liver tumour burden is present.
`Therapeutic options include surgery, SSA, interferon, che-
`motherapy, molecularly targeted agents, locoregional thera-
`pies and PRRNT. Supportive palliative care and pain control
`also play an important role in patient management. These
`options are not mutually exclusive and, for the most part, are
`interchangeable. Options of care, including PRRNT, should
`be chosen and implemented in a correct sequence by an
`experienced multidisciplinary team. This approach should
`provide the highest benefit while minimizing the risks and
`side effects and ensuring the best quality of life achievable
`for the patient. Surgery with curative intent should always
`be performed whenever feasible; in selected cases, and
`within a multidisciplinary approach, PRRNT may be bene-
`ficial as a neoadjuvant therapy to render a patient accessible
`to surgery. For functionally active tumours, cytoreductive
`strategies, e.g. transarterial chemoembolization (TACE),
`transarterial embolization (TAE), radiofrequency ablation
`(RFA) and other techniques such as selective internal radi-
`ation therapy (SIRT), should always be considered with the
`intention of ameliorating clinical symptoms. The optimal
`management of NETs is early surgical removal prior to the
`development of regional or distant metastases. Unfortunate-
`ly, many patients are diagnosed with metastatic disease,
`when complete eradication of their tumours will not be
`possible. Removal of the primary tumour is indicated to
`prevent complications such as bleeding or small-bowel ob-
`struction. Even in the presence of liver metastases, removal
`of the primary tumour has several advantages and seems to
`have a positive prognostic impact on survival [11–14]. Sol-
`itary or isolated liver metastases can be surgically removed,
`while a more diffuse liver infiltration is usually treated using
`a locoregional approach.
`Locoregional approaches or local ablative therapies target
`predominantly liver metastases aiming at achieving local
`tumour control and alleviating functional secretory syn-
`dromes. Different techniques are applied depending on in-
`dividual findings (number size and distribution of liver
`lesions, their morphology, focal or diffuse, and their vascu-
`larization), functional activity of the NET and locally avail-
`able expertise. In an individual with few liver lesions with a
`preferably resected primary lesion, liver lesions can be trea-
`ted by surgical resection with or without RFA or laser-
`induced interstitial thermoablation. In those with multifocal
`or diffuse liver disease causing a high tumour load, TACE
`and TAE are the preferred choices [15, 16]. Local emboli-
`zation techniques are particularly useful when treating
`patients with functionally active liver metastases. Following
`
`Eur J Nucl Med Mol Imaging
`
`TACE, symptomatic response rates of 60–95 % and bio-
`chemical response rates of 50–90 % are achieved and radio-
`logical response of 33–80 % have been reported [17–19].
`Response duration is between 18 and 24 months. Similar
`response rates are achieved with TAE alone [16]. In general,
`the procedure may require more than one treatment session
`to ensure effectiveness and consolidation of the treatment
`and to minimize the risk of complications.
`The recently introduced SIRT has shown variable re-
`sponse rates among individual centres [20]. Prospective
`studies are however lacking. In a single prospective study
`in 34 patients the objective response rate was 50 % [19].
`Given the lack of comparative studies of the different tech-
`niques used for local ablative and locoregional therapies, the
`choice of technique will be highly dependent on the physi-
`cians’ experience in the different centres and on individual
`criteria such as number, size, vascularization and distribu-
`tion of the lesions.
`Among the medical treatments, octreotide and lanreotide
`are the two most used sstr agonists. They play an essential
`role in the control of both symptomatic and asymptomatic
`NETs and should be regarded as first-line therapy. SSA can
`be used with virtually all of the other therapeutic options
`available. As the vast majority (87–92 %) of NETs express
`sstr2, patients should always be offered this therapy along-
`side other concurrent therapeutic options, and for supportive
`care. Long-acting SSA possess secretory inhibiting action,
`and are approved for alleviating the symptoms of the carci-
`noid syndrome, such as flushing and diarrhoea or bronchial
`obstruction, and to prevent carcinoid crisis. It is reported
`that treatment with SSA may control the clinical syndrome
`in 40–90 % of patients subject to tumour load and dosage
`[21, 22]. Nevertheless, patients may become refractory to
`syndrome control, and need incremental dosage increases of
`SSA. However, most patients with tumour progression re-
`quire an additional treatment, including the use of PRRNT.
`The recent PROMID study conducted in Germany showed
`the effectiveness of long-acting SSA as an antiproliferative
`therapeutic agent in midgut NET. In this study, time to
`tumour progression in patients given octreotide LAR
`30 mg intramuscularly monthly was more than double that
`in patients receiving only placebo (6.0 vs. 14.3 months). The
`NCCN guidelines and very recently the ENETS guidelines
`have added octreotide as an option for antiproliferative
`treatment [23, 24].
`Interferon-alpha (IFN-α) has been used for treating
`patients with NETs, especially those with the carcinoid
`syndrome, for more than 25 years. It is considered the main
`antisecretory drug used for the treatment of functional
`tumours [25]. IFN-α effectively reduces hypersecretion-
`related symptoms in patients with carcinoid syndrome in a
`similar manner to SSA. Partial tumour growth responses are
`also observed in 10–15 % of patients with malignant
`
`Evergeen Ex. 1034
`4 of 17
`
`

`

`Eur J Nucl Med Mol Imaging
`
`carcinoids, and stabilization in 39 %. IFN-α has also been
`demonstrated to be effective in endocrine pancreatic
`tumours [26]. The very common side effect of IFN-α,
`namely a ’flu-like syndrome, limits both the use of high
`dosages and the duration of treatment due to intolerance
`forcing its interruption. Systemic chemotherapy is effective
`in some patients, especially those with poorly differentiated
`NET/neuroendocrine carcinoma (grade 3, WHO 2010) or
`progressive NET of the pancreas. However, in well-
`differentiated midgut NETs/NETs (grade 1/2, WHO 2010)
`the response rates to chemotherapy are low (7–20 %) and a
`survival advantage has not been demonstrated [27, 28]. The
`standard treatment for neuroendocrine carcinoma (grade 3)
`is cisplatin and etoposide. The response rate with this com-
`bination is 42–67 %, and its duration is often short, ranging
`from 8 to 9 months [32]. The combination of irinotecan and
`cisplatin [29] or FOLFOX chemotherapy (5-fluorouracil or
`capecitabine and oxaliplatin) may be an alternative [30].
`PRRNT is very rarely a suitable treatment option for neuro-
`endocrine carcinoma (grade 3), because of the low expres-
`sion of sstr but it may be considered following the failure of
`chemotherapy and if 111In-pentetreotide (OctreoScan) or
`68Ga-DOTATOC/DOTATATE PET/CT demonstrates mod-
`erate to high sstr expression.
`Systemic chemotherapy based on streptozotocin (Zano-
`sar, STZ) is considered a standard therapy for progressive
`pancreatic NETs with low or moderate proliferative capac-
`ity. Combinations of STZ and 5-fluorouracil and/or doxoru-
`bicin have been shown to lead to partial remission rates of
`35–40 % [31–33]. Recent phase II studies have shown
`efficacy of temozolomide based chemotherapy either with
`antiangiogenic drugs or capecitabine [34, 35]. Standards of
`care for the use of chemotherapy have been defined by
`ENETS [36]. In recent years, the efficacy of molecular
`targeted therapies for treating NETs has been assessed in
`clinical trials. These therapies include angiogenesis inhib-
`itors, single or multiple tyrosine kinase inhibitors and the
`novel SSA analogue pasireotide, for which clinical trials are
`ongoing. The drugs with the highest evidence of efficacy are
`sunitinib and everolimus (RAD-001). Both lead to extension
`of progression-free survival (PFS) of patients with advanced
`pancreatic NET. For everolimus, an mTOR inhibitor, there
`is evidence of efficacy in controlling NET arising from other
`sites associated with the carcinoid syndrome [37]. The most
`developed antiangiogenic drugs are sunitinib and the anti-
`VEGF antibody bevacizumab. In a phase II study bevacizu-
`mab in combination with octreotide LAR led to partial
`tumour remission in 18 % of patients and stable disease in
`77 % [38]. A recent international phase III study of sunitinib
`versus placebo in patients with progressive well-
`differentiated endocrine pancreatic tumour was interrupted
`prematurely due to the striking superiority of sunitinib evi-
`dent by a PFS of 11.1 vs. 5.5 months [39]. The objective
`
`remission rate was less than 10 %. The drug was recently
`approved by the US FDA and the European Medicines
`Agency for the treatment of advanced and progressive
`well-differentiated pancreatic NETs.
`Everolimus has been studied in more than 1,000 patients
`with NET and has been included in several clinical trials
`(RADIANT-1, RADIANT-2, RADIANT-3 trials, RAM-
`SETE trial). Antitumour activity of everolimus has been
`confirmed in RADIANT-1 in patients with progressive
`metastatic pancreatic NETs after failure of at least one line
`of cytotoxic chemotherapy. The trial studied 160 patients
`divided into two groups with or without monthly intramus-
`cular octreotide acetate therapy. The combination therapy
`group showed significantly longer PFS (16.7 vs. 9.7 months)
`[40]. The efficacy of everolimus has been confirmed in a
`large international placebo-controlled trial including 410
`patients with progressive pancreatic NET (RADIANT-3)
`[41]. Everolimus significantly reduced the risk of disease
`progression and led to a prolongation of PFS by 6.4 months
`(11 vs. 4.6 months) compared to placebo. Objective tumour
`response was low (4.8 % partial remissions). Disease control
`rate (partial response + stable disease) was, however, higher
`with everolimus than with placebo with best supportive care
`(77.7 % vs. 52.7 %). Side effects were rarely grade 3 or 4;
`the most frequently reported side effects included stomatitis,
`anaemia and hyperglycaemia. In May 2011 the US FDA
`approved everolimus for the treatment of progressive NETs
`of pancreatic origin in patients with nonresectable, locally
`advanced, or metastatic disease.
`In the global supportive approach to the patient, and
`when delivering PRRNT, nutrition and pain control are an
`essential component of care. Treatment of pain in patients
`with NET follows the general principles followed in adult
`and paediatric oncology [42]. Effective treatment of NETs,
`such as PRRNT, may alleviate pain, including bone pain.
`Treatment of painful bone metastasis is also mandatory with
`the administration of bisphosphonates as supportive therapy.
`
`PRRNT a historical overview
`
`PRRNT using radiolabelled octreotide was first attempted in
`the 1990s. The initial phase I trial investigated the safety and
`efficacy of using high activities of the diagnostic compound
`111In-octreotide as a therapeutic radiopharmaceutical. The
`results in terms of clinical efficacy were attributed to the effect
`of intracellular emission of the Auger and conversion elec-
`trons by 111In following the internalization of the peptide–
`receptor complex. Partial remissions were exceptional, and
`furthermore three patients developed leukaemia or myelodys-
`plastic syndrome from the group receiving the highest cumu-
`lative dose (90–100 GBq) [43]. In Europe, 111In-pentetreotide
`was abandoned as a therapy option in favour of the more
`efficient β emitters 90Y and 177Lu. 111In-pentetreotide is,
`
`Evergeen Ex. 1034
`5 of 17
`
`

`

`however, still used by some in the USA mainly due to the lack
`of availability of β-emitting radiotracers. High-energy β emit-
`ters, such as 90Y with a longer β range in soft tissue, were
`considered more promising for the treatment of bulky tumour.
`A novel analogue, Tyr3-octreotide, with a similar affinity
`profile for sstrs, was developed. Linked to a macrocyclic
`chelator (DOTA), it allows simple and stable radiolabelling
`of 111In and 90Y to [DOTA0,Tyr3]-octreotide (90Y-DOTA-
`TOC) [44]. PRRNT using 90Y-DOTATOC was first used in
`1996 in a patient in Basel, Switzerland. The excellent subjec-
`tive and objective response following several cycles of 90Y-
`DOTATOC led to high expectations as to the therapeutic
`potential of PRRNT in patients with NET. Since then other
`centres worldwide have conducted clinical trials with 90Y-
`DOTATOC [45]. Since the year 2000, octreotate (Tyr3,Thr8-
`octreotide), a newer analogue with improved affinity for sstr2,
`has been synthesized. The chelated analogue [DOTA0,Tyr3]-
`octreotate (DOTATATE) can be labelled with the β- and γ-
`emitting isotope 177Lu and has been used in clinical studies.
`
`Indications and contraindications
`
`Indications
`
`PRRNT is indicated for the treatment of patients with
`positive expression of sstr2, or metastatic or inoperable
`NET [46–50]. Candidate patients for PRRNT using
`radiolabelled somatostatin analogues are mainly those
`with sstr2-expressing NET of the gastroenteropancreatic
`and bronchial tracts, but may also include patients with
`phaeochromocytoma, paraganglioma, neuroblastoma [51]
`or medullary thyroid carcinoma [52–56]. The ideal can-
`didates for PRRNT are those with well-differentiated
`and moderately differentiated neuroendocrine carcinomas
`defined as NET grade 1 or 2 according to the recent
`WHO 2010 classification [4].
`
`Contraindications
`
`Absolute
`
`&
`&
`&
`
`Pregnancy.
`Severe acute concomitant illnesses.
`Severe unmanageable psychiatric disorder.
`
`Relative
`
`& Breast feeding (if not discontinued).
`&
`Severely compromised renal function: for PRRNT with
`a 90Y-labelled peptide age-adjusted normal renal func-
`tion is essential. Patients with compromised renal func-
`tion may still be considered for 177Lu-labelled peptide
`
`Eur J Nucl Med Mol Imaging
`
`treatment. For 177Lu-labelled peptide a mild to moderate
`grade of renal impairment can be tolerated (e.g. creati-
`nine ≤1.7 mg/dl). Glomerular filtration rate (GFR) and
`tubular extraction rate (TER) should be at least 60 % of
`mean age-adjusted normal values.
`Severely compromised bone marrow: noncompromised
`haematological reserve should be present before
`PRRNT. Suggested reference values are:
`
`&
`
`WBC <3,000/μl, with absolute neutrophil count <1,000/μl
`PLT <75,000/μl for 177Lu-DOTATATE, <90,000/μl for
`90Y-DOTATOC,
`RBC <3,000,000/μl.
`
`Special warnings
`
`Renal function
`
`the activities
`The kidney is the dose-limiting organ at
`normally used for PRRNT. Side effects involving the
`kidney and the bone marrow are mild if adequate renal
`protection and fractionation are used. Renal function
`should be assessed by means of laboratory tests (creat-
`inine and BUN), or calculation of creatinine clearance
`(e.g. Cockroft-Gault formula). Additional studies, e.g.
`measurement of GFR with 24-h urine collection or
`nuclear medicine methods (e.g. 99mTc-MAG3 with
`TER determination, 99mTc-DTPA GFR or effective renal
`plasma flow using hippuran), should be performed in
`patients with risk factors for
`renal
`toxicity or with
`compromised renal function, and in all children.
`
`Aggravating conditions (caveats)
`
`&
`
`& Renal outflow obstruction, potentially leading to
`hydronephrosis, and, ultimately,
`to loss of
`renal
`function, should always be ruled out or otherwise
`corrected before PRRNT.
`Previous myelotoxic chemotherapy and extended ex-
`ternal beam irradiation fields to the bone marrow
`(pelvis, spine), particularly if performed in the
`weeks preceding the PRRNT, do increase the risk
`of bone marrow failure after PRRNT. In doubtful
`cases of haematological compromise, a bone marrow
`biopsy might be indicated to assess bone marrow
`status in pretreated patients and to assess the risk
`when planning for multiple PRRNT cycles (e.g.
`intervals between cycles). Depending on the amount
`of 90Y-DOTATOC or 177Lu-DOTATATE activity
`injected, persisting depressed platelets values follow-
`ing prior PRRNT cycle(s) can impede the timing and
`dosing of subsequent cycles.
`
`Evergeen Ex. 1034
`6 of 17
`
`

`

`Eur J Nucl Med Mol Imaging
`
`& A patient with pending liver failure should be consid-
`ered with caution before being submitted to PRRNT.
`
`Patient preparation
`
`Renal protection
`
`Procedure
`
`Pretherapy assessment
`
`The availability of the following information is mandatory
`when considering a patient for PRRNT:
`& NET proven by histopathology (immunohistochemistry).
`& High sstr expression determined by functional whole-
`body imaging with 111In-pentetreotide (OctreoScan) or 68
`Ga-DOTA-peptide PET/CT or immunohistochemistry.
`
`The following criteria should be taken into consideration
`when deciding whether or not to perform PRRNT.
`
`& Karnofsky/Lansky performance status above 60 % or
`ECOG performance status less than 2.
`& Tumour differentiation, preferably grade 1/2.
`& Tumour proliferation rate, preferably with a Ki-67/mitotic
`index ≤20 %. In addition, the rate of tumour growth, as
`determined by CT or MRI, could be considered. Note that,
`in general, less-differentiated tumours showing high pro-
`liferation rates are better candidates for chemotherapy.
`
`Facility and personnel
`
`PRRNT is still considered an investigational treatment and
`its implementation must comply with national legislation
`and local requirements, as well as with ethical principles
`regarding human studies. The decision to provide PRRNT
`should be taken within a multidisciplinary tumour board,
`including all the specialists involved in the care of patients
`with NET. The facility requirements will depend on national
`legislation on the therapeutic use of radioactive agents. If
`inpatient therapy is required by national legislation, the
`treatment should take place in an approved facility. The
`facility must have appropriate personnel, radiation safety
`equipment, and procedures for waste management and han-
`dling accidental contamination of the site or personnel.
`90Y-DOTATOC or 177Lu-DOTATATE should be admin-
`istered by appropriately trained medical staff with support-
`ing nursing staff with a medical physics expert available.
`Physicians responsible for treating patients should have a
`general knowledge of the pathophysiology and natural history
`of the respective diseases, should be familiar with alternative
`forms of therapy, and should be able to closely liaise with
`other physicians involved in managing the patients. Clinicians
`involved in the utilization of unsealed radionuclide sources for
`therapy must also be knowledgeable about and compliant with
`applicable national legislation and local regulations.
`
`Together with the bone marrow, the kidneys are the critical
`organs in PRRNT particularly when using 90Y-DOTATOC.
`Proximal tubular reabsorption of the radiopeptide and subse-
`quent retention in the interstitium result in excessive renal
`irradiation. Nephrotoxicity may be aggravated by risk factors,
`such as preexisting hypertension or diabetes mellitus [57]. To
`counteract and reduce the high kidney retention of radiopep-
`tides, positively charged amino acids, such as L-lysine and/or
`L-arginine, are coinfused to competitively inhibit the proximal
`tubular reabsorption of the radiopeptide. The coadministration
`of these amino acids leads to a significant reduction in the
`renal absorbed dose, which ranges from 9 % to 53 % [58].
`Renal absorbed dose is further reduced by up to 39 % by
`extending the infusion time of the amino acid solution over
`10 h, and up to 65 % by extending the protection over 2 days
`following radiopeptide administration, thereby covering the
`renal elimination phase more efficiently [59, 60].
`
`Amino acid protection protocols
`
`Lysine and/or arginine should be diluted appropriately in
`large volumes of normal saline in order to hydrate the
`patient, unless the patient suffers from cardiac in

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