throbber
Clinical
`Cancer
`Research
`
`Cancer Therapy: Clinical
`
`Weekly nab-Rapamycin in Patients with Advanced
`Nonhematologic Malignancies: Final Results of a Phase I Trial
`
`Ana M. Gonzalez-Angulo1, Funda Meric-Bernstam2,3, Sant Chawla4, Gerald Falchook3, David Hong3,
`Argun Akcakanat2, Huiqin Chen1, Aung Naing3, Siqing Fu3, Jennifer Wheler3, Stacy Moulder1,
`Thorunn Helgason3, Shaoyi Li6, Ileana Elias6, Neil Desai6, and Razelle Kurzrock5
`
`Abstract
`
`Purpose: This dose-finding phase I study investigated the maximum-tolerated dose (MTD) and safety of
`weekly nanoparticle albumin-bound rapamycin (nab-rapamycin) in patients with untreatable advanced
`nonhematologic malignancies.
`Experimental Design: nab-Rapamycin was administered weekly for 3 weeks followed by 1 week of rest,
`with a starting dose of 45 mg/m2. Additional doses were 56.25, 100, 150, and 125 mg/m2.
`Results: Of 27 enrolled patients, 26 were treated. Two dose-limiting toxicities (DLT) occurred at 150
`mg/m2 [grade 3 aspartate aminotransferase (AST) elevation and grade 4 thrombocytopenia], and two DLTs
`occurred at 125 mg/m2 (grade 3 suicidal ideation and grade 3 hypophosphatemia). Thus, the MTD was
`declared at 100 mg/m2. Most treatment-related adverse events (TRAE) were grade 1/2, including
`thrombocytopenia (58%), hypokalemia (23%), mucositis (38%), fatigue (27%), rash (23%), diarrhea
`(23%), nausea (19%), anemia (19%), hypophosphatemia (19%), neutropenia (15%), and hypertrigly-
`ceridemia (15%). Only one grade 3 nonhematologic TRAE (dyspnea) and one grade 3 hematologic event
`(anemia) occurred at the MTD. One patient with kidney cancer had a partial response and 2 patients
`remained on study for 365 days (patient with mesothelioma) and 238 days (patient with neuroendocrine
`tumor). The peak concentration (Cmax) and area under the concentration–time curve (AUC) of rapamycin
`increased with dose between 45 and 150 mg/m2, except for a relatively low AUC at 125 mg/m2. nab-
`Rapamycin significantly inhibited mTOR targets S6K and 4EBP1.
`Conclusions: The clinical dose of single-agent nab-rapamycin was established at 100 mg/m2 weekly (3 of
`4 weeks) given intravenously, which was well tolerated with preliminary evidence of response and stable
`disease, and produced a fairly dose-proportional pharmacokinetic profile in patients with unresectable
`advanced nonhematologic malignancies. Clin Cancer Res; 19(19); 5474–84. Ó2013 AACR.
`
`Introduction
`The prognosis for patients with advanced solid tumors is
`poor, as most malignancies are not responsive to standard
`treatments at the advanced stage. mTOR, a serine/threonine-
`specific protein kinase, is downstream of the phosphoinosi-
`tide 3-kinase (PI3K)/Akt pathway, and a key regulator of cell
`
`Authors' Affiliations: Departments of 1Breast Medical Oncology and Sys-
`tems Biology, 2Surgical Oncology, and 3Investigational Cancer Therapeu-
`tics, The University of Texas MD Anderson Cancer Center, Houston, Texas;
`4Sarcoma Oncology Center, Santa Monica; 5Division of Hematology-Oncol-
`ogy, University of California, San Diego, California; and 6Celgene, Summit,
`New Jersey
`
`Note: Supplementary data for this article are available at Clinical Cancer
`Research Online (http://clincancerres.aacrjournals.org/).
`
`Corresponding Author: Ana M. Gonzalez-Angulo, Department of Breast
`Medical Oncology and Department of Systems Biology, The University of
`Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Unit 1354,
`Houston, TX 77030. Phone: 713-792-2817; Fax: 713-794-4385; E-mail:
`agonzalez@mdanderson.org
`
`doi: 10.1158/1078-0432.CCR-12-3110
`Ó2013 American Association for Cancer Research.
`
`survival, proliferation, stress, and metabolism (1). mTOR
`inhibition with rapamycin and rapalogs (everolimus and
`temsirolimus) has proven to be effective in various solid
`tumors including renal cell carcinoma, neuroendocrine
`tumors, and breast cancer (2–12).
`Although rapamycin is an efficacious allosteric inhibitor of
`mTOR complex 1 (mTORC1), it has low oral bioavailability,
`poor solubility, and dose-limiting intestinal toxicity (13, 14).
`Other rapalogs, including everolimus and ridaforolimus, are
`also oral preparations and are often associated with signif-
`icant stomatitis (15). Temsirolimus, a prodrug of rapamycin,
`requires conversion by the CYP3A enzyme and also carries a
`significant risk for developing skin rash and stomatitis (16).
`Because none of the rapalogs are highly water soluble, they
`require surfactants and solvents in an intravenous formula-
`tion, such as polysorbate 80 for temsirolimus (17). The use of
`surfactants can potentially cause irritation, local inflamma-
`tion, and potential reduction of drug efficacy due to micellar
`sequestration, and the need for premedication to avoid
`potential hypersensitivity reactions (17). The nanoparticle
`albumin-bound rapamycin (nab-rapamycin; Celgene Inc.),
`
`5474
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`.At(~ AmericanAssoci,atwn for Cancer Research
`
`

`

`Translational Relevance
`In this first clinical evaluation of nanoparticle albu-
`min-bound rapamycin (nab-rapamycin), an mTOR
`inhibitor, it was well tolerated given intravenously in
`patients with unresectable advanced solid tumors. Most
`rapalogs are oral preparations requiring toxic surfactants
`for intravenous formulation due to poor water solubil-
`ity. The nab-technology exploits the natural properties of
`human albumin to achieve a solvent-free drug delivery.
`Dose-limiting toxicities (DLT) including mucositis/sto-
`matitis that were observed with mTOR inhibitors were
`not dose-limiting with nab-rapamycin. Notably, 27% of
`patients were 65 years or older, a frail population that are
`more prone to toxicities and receive lesser benefits than
`younger patients from everolimus/temsirolimus. Pre-
`liminary proof-of-efficacy was observed in this phase I
`study. nab-Rapamycin produced a fairly dose-propor-
`tional peak concentration (Cmax) and area under the
`concentration–time curve (AUC) increase of rapamycin,
`and significantly inhibited mTOR targets.
`
`with a mean particle size of about 100 nm, is freely dis-
`persible in saline and is suitable for intravenous adminis-
`tration, and may be an advantageous alternative to oral
`rapamycin or oral rapalogs. Human albumin has broad
`binding affinity and accumulates in tumors, making it an
`ideal candidate for drug delivery (18, 19). In preclinical
`studies, nab-rapamycin was safe and highly effective in
`multiple tumor types; it reduced cell viability and decreased
`downstream signaling in various xenograft cancer models,
`including pancreatic, colorectal, multiple myeloma, and
`breast cancer (20–23). In addition, in human breast xeno-
`graft models, nab-rapamycin alone produced 75% tumor
`growth inhibition without weight loss and antitumor activ-
`ity was further enhanced with the combination of doxoru-
`bicin (a topoisomerase inhibitor), SAHA [an histone dea-
`cetylase (HDAC) inhibitor], erlotinib (an EGF tyrosine
`kinase inhibitor), and perifosine (an Akt inhibitor) with
`15% or less weight loss, indicating high tolerability in the
`combination regimens (24).
`On the basis of the promising preclinical results, this dose-
`finding phase I study investigated the maximum-tolerated
`dose (MTD) and safety of intravenous single-agent weekly
`nab-rapamycin in patients with untreatable advanced non-
`hematologic malignancies.
`
`Patients and Methods
`This study was conducted at MD Anderson Cancer Center
`(Houston, TX), and the Sarcoma Oncology Center (Santa
`Monica, CA). The study was approved by the Institutional
`Review Board of both participating medical institutions and
`was conducted in compliance with the World Medical
`Association Declaration of Helsinki and Good Clinical
`Practice, Guidelines of the International Conference on
`
`nab-Rapamycin in Advanced Nonhematologic Malignancies
`
`Harmonization (25). Written informed consent was obt-
`ained from all patients before study initiation.
`
`Patients
`Eligible patients were 18 years or older, had histologically
`or cytologically confirmed diagnosis of stage IV cancer that
`was not amenable to curative therapy. Advanced disease was
`defined as metastatic disease or locally advanced disease
`that was surgically unresectable and considered unmanage-
`able with standard therapies such as radiation or systemic
`therapies. Patients had a measurable disease by Response
`Evaluation Criteria in Solid Tumor (RECIST) v1.0, life
`expectancy 3 or more months, an Eastern Cooperative
`Oncology Group (ECOG) performance status of 0–1, ade-
`quate renal function (serum creatinine <1.5 mg/dL and/or
`creatinine clearance 60 mL/min), and were off all therapy
`for at least 4 weeks before study drug administration.
`Patients were excluded from the study if they had brain
`metastasis, history of interstitial lung disease and/or pneu-
`monitis, or a history of allergy or hypersensitivity to the
`study drug or any compounds of similar chemical or bio-
`logic composition.
`
`Study design
`This dose-finding study evaluated MTD and dose-limit-
`ing toxicities (DLT) of nab-rapamycin in patients with
`advanced nonhematologic malignancies. Following base-
`line evaluations, patients entered into the treatment period.
`nab-Rapamycin was administered by intravenous infusion
`for 30 minutes weekly for 3 weeks followed by 1 week of rest
`(28-day cycle), with a starting dose of 45 mg/m2. The
`starting dose of nab-rapamycin was chosen on the basis of
`nonclinical toxicology data of nab-rapamycin. Additional
`dose levels were 56.25, 100, 150, and 125 mg/m2. The
`original protocol was amended to add the 125 mg/m2 dose
`cohort for refinement of MTD.
`The first cycle was considered the treatment interval for
`determination of DLTs and the MTD. The MTD for nab-
`rapamycin was determined using a standard 3þ3 design,
`where 3 patients were enrolled at each dose level. The
`protocol was amended to ensure that all patients at a given
`dose level complete one cycle of therapy before patients
`were enrolled at the next dose level. If no DLT was observed,
`3 additional patients were enrolled at the next dose level. If
`one DLT was observed, the dose level was expanded to 6
`patients. If two DLTs were observed at a given dose level, the
`MTD was considered to be exceeded. Of the 6-patient
`expanded cohort, if 1 of 6 patients experienced a DLT,
`this was defined as the MTD. All patients at a given dose level
`completed one cycle of
`therapy before patients were
`enrolled at the next dose level.
`A DLT was defined [using the National Cancer Institute
`Common Terminology Criteria of Adverse Events (NCI
`CTCAE) v3.0] as any grade 3/4 nonhematologic toxicity,
`grade 3/4 nausea, or vomiting that occurred despite tre-
`atment, grade 4 thrombocytopenia of any duration and
`grade 4 uncomplicated neutropenia (i.e., without fever
`or infection) lasting more than 7 days, grade 4 febrile
`
`www.aacrjournals.org
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`5475
`
`

`

`Gonzalez-Angulo et al.
`
`neutropenia that required hospitalization, and any grade 3
`hematologic toxicity that required treatment delay beyond
`3 weeks.
`Throughout the study, patients were routinely assessed
`for toxicities, response, and possible need for a dose mod-
`ification. Patients continued on treatment until they expe-
`rienced progressive disease or unacceptable toxicity, with-
`drew consent, or their physician felt it was no longer in their
`best
`interest
`to continue on treatment. Discontinued
`patients completed the end of study evaluation and entered
`into a 30-day follow-up period.
`
`Assessments and statistical methods
`All patients who received at least one dose of study drug
`(treated population) were evaluated for safety. Safety and
`tolerability endpoints included the incidence of treatment-
`related adverse events (TRAE) by NCI CTCAE v3.0 and the
`percentage of patients experiencing TRAEs that required
`dose delays/modifications, and/or premature discontinua-
`tion of the study drug.
`The exploratory efficacy analysis included the summary
`of percentage of patients who achieved an objective with
`confirmed complete or partial tumor response (CR or PR,
`respectively) and the percentage of patients with confirmed
`stable disease for at least 12 weeks, using RECIST v1.0. The
`objective tumor responses of target or nontarget lesions
`were classified individually based on RECIST v1.0. The
`overall tumor response was determined by taking into
`account the responses of target lesions and nontarget
`lesions as well as the presence of new lesions.
`Tumor response assessments were carried out every 12
`weeks. A waterfall plot was used to illustrate the percentage
`change of target lesion from baseline for all patients with
`target tumor evaluation. The corresponding objective target
`lesion responses, dose level cohorts, and tumor types were
`also provided in the graph.
`
`Molecular analyses
`Evaluation of PTEN loss was carried out with immu-
`nohistochemistry (IHC) using monoclonal mouse anti-
`human PTEN antibody clone 6H2.1 from Dako at 1:100
`dilution, as described by Gonzalez-Angulo and colleagues
`(26). Briefly, both cytoplasmic and nuclear PTEN staining
`in the tumor and non-neoplastic ductal epithelium and
`stroma were quantified. PTEN expression level was scored
`semiquantitatively on the basis of staining intensity (SI)
`and distribution using the immunoreactive score (IRS) as
`follows: IRS ¼ SI  percentage of positive cells. Staining
`intensity was determined as 0, negative; 1, weak; 2,
`moderate; and 3, strong. Percentage of positive cells was
`defined as 0, <1%; 1, 1%–10%; 2, 11%–50%; 3, 51%–
`80%; and 4, >80% positive cells. Tumors with IRS of 0
`were considered to have PTEN loss. A mass spectroscopy–
`based approach evaluating single-nucleotide polymorph-
`isms (SNP) was used to detect known mutations in
`members of the PI3K pathway. Molecular analysis was
`conducted in patients who showed clinical benefit using
`archival tissue.
`
`Pharmacokinetics
`Whole-blood samples (4 mL each) were collected in
`vacutainer tubes containing EDTA as the anticoagulant for
`determination of rapamycin. Samples were obtained only
`during cycle 1 and were taken immediately predose (before
`infusion), during the infusion (15 and 30 minutes before
`end of the infusion), and postinfusion at 1.0, 1.5, 2, 4, 6, 8,
`24, 48, 72, 96, and 168 hours. The samples were stored
`frozen at a temperature between 20
`C and 80
`
`
`C until
`shipment for analysis to St. George’s Hospital at the Uni-
`versity of London (London, United Kingdom).
`The whole-blood samples were analyzed for total (free þ
`bound) rapamycin using high-performance liquid chroma-
`tography–tandem mass
`spectrometry (HPLC/MS-MS).
`Rapamycin concentrations in whole blood were validated
`from 10 to 2,000 ng/mL with 32-desmethoxyrapamycin
`used as an internal standard. Analytes were extracted using
`a solvent mixture and detected and quantified by reverse
`phase HPLC with detection via turbo ion-spray mass
`spectrometry.
`The concentration-versus-time data for rapamycin in
`whole blood were analyzed using a noncompartmental
`analysis technique and WinNonlin software. Pharmacoki-
`netic analysis was based on whole-blood concentrations
`due to the known instability of rapamycin in plasma.
`Calculated parameters included peak concentration (Cmax),
`half-life (t1/2), area under the concentration–time curve
`(AUC), clearance (CL), and steady-state volume of distri-
`bution (Vss). A simple regression model was applied to
`assess the relationship of the pharmacokinetic parameters
`with dose.
`
`Peripheral blood mononuclear cells and reverse phase
`protein arrays
`Whole blood for pharmacodynamics evaluation was
`collected only during cycle 1 at four time points: C1 D1
`(pretreatment), C1 D2, C1 D4, and C1 D8 (immediately
`before next dose) in an 8-mL cell preparation tube with
`sodium citrate (Becton, Dickinson and Company). Sepa-
`ration of peripheral blood mononuclear cells (PBMC) from
`whole blood was accomplished through density gradient
`centrifugation using Ficoll following the manufacturer’s
`recommendations. After centrifugation, plasma compo-
`nent from the upper half of the tube was transferred to
`cryotubes and snap-frozen. The layer containing the cells
`was transferred to a fresh tube, washed, and centrifuged.
`After removal of the supernatant, PBMC pellet was also
`snap-frozen.
`Reverse phase protein array (RPPA) was conducted in the
`MD Anderson Cancer Center Functional Proteomics RPPA
`Core Facility as described previously (27). PBMC samples
`were resuspended in RPPA lysis buffer containing 0.25%
`sodium deoxycholate. Protein concentrations were deter-
`mined using BCA method (Pierce) and 4 SDS sample
`buffer was added. Final protein concentration was adjusted
`to 3 mg/mL. Samples were probed with antibodies that were
`validated for RPPA. A total of 135 proteins and 21 replicates
`were analyzed,
`including S6 S240/244, S6 S235/236,
`
`5476
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`Clinical Cancer Research
`
`

`

`nab-Rapamycin in Advanced Nonhematologic Malignancies
`
`Table 1. Baseline patient demographics and
`characteristics
`
`MTD
`n ¼ 7
`57 (36, 76)
`4 (57)
`3 (43)
`
`5 (71)
`2 (29)
`
`0
`0
`6 (86)
`1 (14)
`
`2 (29)
`5 (71)
`0
`
`7 (100)
`
`Age, median years (range)
`<65 years, n (%)
`65 years, n (%)
`Gender, n (%)
`Male
`Female
`Race
`Asian, n (%)
`African heritage, n (%)
`Caucasian, n (%)
`Hispanic, Latino, n (%)
`ECOG, n (%)
`0
`1
`2
`Stage at current diagnosis, n (%)
`IV
`Site of primary diagnosis, n (%)
`1 (14)
`Bladder
`0
`Breast
`2 (29)
`Colorectal
`1 (14)
`Esophagus
`1 (14)
`Head and neck
`0
`Kidney
`1 (14)
`Lung/thoracic
`0
`Prostate
`0
`Stomach
`1 (14)
`Uterus
`0
`Other
`Histology of primary diagnosis, n (%)
`Carcinoma/adenocarcinoma
`5 (71)
`Sarcoma/sarcomatoid
`2 (29)
`Site of metastasis, n (%)
`Visceral
`Nonvisceral
`
`S6KT389, 4EBP1 T37/46, and 4EBP1 T70. Proteomics
`assessment of S6 S240/244 and 4EBP1 T37/46 was carried
`out using Meso Scale discovery (MSD) phosphoprotein
`assays (Meso Scale Discovery).
`The RPPA spot signal intensity data obtained from Micro-
`Vigene automated RPPA module (VigeneTech, Inc.) were
`analyzed using the R package SuperCurve (version 1.4.3;
`ref. 28), available at "http://bioinformatics.mdanderson.
`org/OOMPA". RPPA raw data were treated with median
`centering across samples, and then a centering by the
`sample median was undertaken on the treated data and
`the final normalized data were obtained by applying medi-
`an absolute deviation (MAD) scaling to the data. Linear
`mixed models and ANOVA tests were developed and
`applied to test the pre- versus posttreatment and inhibition
`effects at each dose level and each pair of time points. Tukey
`tests were also used for pairwise comparisons. To test the
`association of proteins expression on patients’ response,
`patients with stable disease and progressive disease were
`also compared using logistic models adjusted by time
`points, and their interactions were also taken into account.
`
`Results
`Patients
`Twenty-seven patients were enrolled in the study and 26
`patients were treated of which 19 have evaluable tumor
`assessment data. Specifically, 7 patients were treated in the
`45 mg/m2 arm, 1 additional patient was added after a pat-
`ient did not complete a full cycle, 3 in the 56.25 mg/m2, 7
`in the 100 mg/m2, 2 in the 150 mg/m2, and 7 in 125 mg/m2
`arm. Seven patients had no tumor assessments beyond
`the baseline evaluation as a result of loss to follow-up
`(3 patients), patient request (1 patient), drug shortage
`(1 patient), and incomplete tumor evaluation (1 patient).
`All patients had discontinued therapy at the time of this
`analysis. Eighteen (69%) patients discontinued treatment
`because of disease progression, 4 (15%) due to adverse
`events/toxicities, 2 (8%) for patient request, and 2 (8%) for
`drug shortage. Patient baseline demographics and charac-
`teristics were described in Table 1. Briefly, the median age
`was 60.5 years, and with the majority of patients were male
`(62%), Caucasian (81%), and had a baseline ECOG score
`of 1 (73%). The most common sites of primary tumor
`diagnosis were head and neck, colorectal, and kidney
`(12% each). Most patients had a carcinoma/adenocarcino-
`ma (54%) and the rest had sarcoma. All patients had visceral
`metastases. The most common sites of metastases were
`lung/thoracic (69%), liver (46%), lymph node (42%), and
`abdomen/peritoneal (42%).
`
`Treatment exposure
`For all patients, the median number of cycles adminis-
`tered was three (1–11, 15, 29), with 27% of patients having
`more than three cycles of therapy. The median cumulative
`rapamycin dose was 405 mg/m2 (100–2,200), with the
`median dose intensity of 68.9 mg/m2/wk (11.4–150.0).
`At the MTD, the median number of cycles was also three
`
`All treated
`patients
`n ¼ 26
`60.5 (18, 78)
`19 (73)
`7 (27)
`
`16 (62)
`10 (38)
`
`1 (4)
`2 (8)
`21 (81)
`2 (8)
`
`5 (19)
`19 (73)
`2 (8)
`
`26 (100)
`
`1 (4)
`1 (4)
`3 (12)
`2 (8)
`3 (12)
`3 (12)
`2 (8)
`1 (4)
`1 (4)
`1 (4)
`8 (31)
`
`14 (54)
`12 (46)
`
`26 (100)
`0
`
`7 (100)
`0
`
`(1–3, 15, 29), with the median cumulative dose of 800
`mg/m2 (100–900) and median dose intensity of 78.9
`mg/m2/wk (51.1–100.0).
`
`Safety results
`MTD. Following dose escalation to 100 mg/m2, nab-
`rapamycin dose was initially escalated to 150 mg/m2. Two
`DLTs occurred in the 150 mg/m2 cohort: a grade 3 elevation
`of aspartate aminotransferase (AST) and a grade 4 throm-
`bocytopenia. After observing DLTs at the 150 mg/m2 cohort,
`a new dose level of 125 mg/m2 was added for refinement of
`MTD. At the 125 mg/m2 dose level, two DLTs occurred
`(grade 3 suicidal ideation and grade 3 hypophosphatemia);
`therefore, the MTD was reached and declared at 100 mg/m2.
`
`www.aacrjournals.org
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`5477
`
`

`

`Gonzalez-Angulo et al.
`
`TRAEs. For all cohorts and all grades, 25 of 26 (96%)
`patients experienced at least one TRAE. The most common
`nonhematologic TRAEs reported were mucosal inflamma-
`tion (10 patients; 38%), fatigue (7 patients; 27%), rash (6
`patients; 23%), diarrhea (6 patients; 23%), and nausea (5
`patients; 19%; see Table 2). Most of these adverse events
`were grade 1/2 events, with only three grade 3 nonhema-
`tologic adverse events (two elevated AST and one dyspnea).
`Specifically, at the MTD (100 mg/m2), all 7 patients expe-
`rienced at least one TRAE of any grades, and the most
`common adverse events were mucositis and fatigue (5
`patients; 71% each). Four (15%) patients experienced at
`least one treatment-related serious adverse event, including
`arrhythmia (grade 2) and mood alteration (grade 3) both in
`the 125 mg/m2 cohort, vomiting (grade 3) in the 45 mg/m2
`cohort, and dyspnea (grade 3) in the 100 mg/m2 cohort.
`The most common hematologic TRAE, for all cohorts and
`grades, were thrombocytopenia (58%), followed by hypo-
`kalemia (23%), anemia and hypophosphatemia (19%
`each), and neutropenia and hypertriglyceridemia (15%
`each; see Table 2). Most of these events were grade 1/2,
`and only one grade 4 hematologic event occurred (throm-
`bocytopenia in the 150 mg/m2 arm). At the MTD, the only
`hematologic adverse event was a grade 3 anemia.
`Treatment-related study drug reductions, delays, and dis-
`Five (19%) patients experienced TRAEs
`continuations.
`that required study drug dose reductions and 50% of dose
`reductions occurred at cycle 2. Only 1 patient at the MTD
`had an adverse event that required a dose reduction, which
`
`occurred at cycle 4. The specific events requiring dose
`reductions were one grade 2 thrombocytopenia and one
`grade 2 dyslipidemia in the 100 mg/m2 cohort, and two
`grade 3 thrombocytopenia and one grade 3 suicidal idea-
`tion in the 125 mg/m2 cohort. The patient who experienced
`suicidal ideation had been on antidepressants before the
`trial. After the onset of grade 3 suicidal ideation (end of cycle
`1), this patient received two cycles of nab-rapamycin at a
`reduced dose (100 mg/m2), during which no suicidal
`ideation was reported. In addition, there was a dose reduc-
`tion for a grade 2 elevated AST in the 45 mg/m2 cohort. The
`dose was reduced to 30 mg/m2, which was not specified in
`the protocol. This patient responded to treatment and the
`physician felt that continuing the treatment at a lower dose
`was in the best interest for this patient.
`Sixteen (62%) patients had TRAEs requiring a dose delay:
`4 (57%) patients in the 45 mg/m2, 1 (33%) in the 56.25
`mg/m2, 4 (57%) in the 100 mg/m2, 2 (100%) in the 150
`mg/m2, and 5 (71%) in the 125 mg/m2 cohort. Specifically
`in the 100 mg/m2 cohort, the treatment-related dose delays
`were due to three grade 2 thrombocytopenia, a grade 2
`elevated triglycerides, a grade 2 mucosal inflammation, and
`a grade 3 dyspnea. Only 1 patient had a TRAE that resulted in
`study drug discontinuation (150 mg/m2 cohort; 1 patient
`with a grade 4 thrombocytopenia and a grade 2 diarrhea).
`
`Pharmacokinetics
`Whole-blood samples obtained during cycle 1 of treat-
`ment at
`the specified time points were analyzed for
`
`Table 2. Treatment-related grade 1–4 hematologic and nonhematologic adverse events reported in 10% or
`more of all treated patients
`
`NCI CTCAE v 3.0
`Hematologic AEs, n (%)
`Anemia
`Hypokalemia
`Hypophosphatemia
`Hypertriglyceridemia
`Neutropenia
`Thrombocytopenia
`Nonhematologic AEs, n (%)
`AST
`Constipation
`Diarrhea
`Dyspnea
`Fatigue
`Infection, oral cavity
`Mucositis/stomatitis
`Nausea
`Rash
`Weight loss
`
`MTD (100 mg/m2)
`n ¼ 7
`G3
`
`G1
`
`G2
`
`G4
`
`G1
`
`G2
`
`All treated patients
`n ¼ 26
`G3
`
`0
`1 (14)
`0
`1 (14)
`1 (14)
`1 (14)
`
`0
`0
`1 (14)
`0
`1 (14)
`1 (14)
`3 (43)
`1 (14)
`1 (14)
`0
`
`0
`0
`1 (14)
`1 (14)
`0
`4 (57)
`
`0
`1 (14)
`0
`1 (14)
`4 (57)
`1 (14)
`2 (29)
`1 (14)
`0
`0
`
`1 (14)
`0
`0
`0
`0
`0
`
`0
`0
`0
`1 (14)
`0
`0
`0
`0
`0
`0
`
`0
`0
`0
`0
`0
`0
`
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`0
`5 (19)
`1 (4)
`2 (8)
`2 (8)
`5 (19)
`
`1 (4)
`1 (4)
`3 (12)
`1 (4)
`1 (4)
`3 (12)
`7 (27)
`3 (12)
`4 (15)
`1 (4)
`
`3 (12)
`0
`2 (8)
`1 (4)
`1 (4)
`6 (23)
`
`0
`2 (8)
`3 (12)
`2 (8)
`6 (23)
`2 (8)
`3 (12)
`2 (8)
`2 (8)
`2 (8)
`
`2 (8)
`1 (4)
`2 (8)
`1 (4)
`1 (4)
`3 (12)
`
`2 (8)
`0
`0
`1 (4)
`0
`0
`0
`0
`0
`0
`
`G4
`
`0
`0
`0
`0
`0
`1 (4)
`
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`Abbreviations: AE, adverse event; G, grade.
`
`5478
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`Clinical Cancer Research
`
`

`

`nab-Rapamycin in Advanced Nonhematologic Malignancies
`
`211.11(41.62)
`296.74(20.79)
`215.09(29.03)
`135.70(25.71)
`107.36(11.35)
`(%CV)
`Vss,L/m2
`
`36.03(2.25)
`78.80(20.05)
`44.34(44.72)
`39.35(34.88)
`31.29(15.34)
`(%CV)
`CL,mL/min/m2
`
`462.67(2.25)
`219.39(21.34)
`440.73(40.31)
`455.12(30.01)
`545.87(18.74)
`(%CV)
`ng h m2/mL/mg
`AUCinf/dose,
`
`69,400.48(2.25)
`27,423.48(21.34)
`44,072.64(40.31)
`25,600.46(30.01)
`24,564.19(18.74)
`(%CV)
`AUCinf,ng h/mL
`
`25.52(96.39)
`34.09(41.01)
`32.28(38.00)
`51.23(36.38)
`39.47(40.30)
`(%CV)
`ng m2/mL/mg
`Cmax/dose,
`
`3,828.45(96.39)
`4,261.24(41.01)
`3,227.61(38.00)
`2,881.77(36.38)
`1,776.27(40.30)
`(%CV)
`Cmax,ng/mL
`
`1.10(72.86)
`0.36(37.42)
`0.46(20.35)
`0.44(21.66)
`0.46(20.27)
`tmax,h(%CV)
`
`90.75(5.42)
`54.93(22.74)
`63.10(35.77)
`46.31(33.36)
`39.57(26.79)
`HL,h(%CV)
`
`150(2)
`125(7)
`100(7)
`56.25(3)
`45(7)
`
`mg(n)
`Dose,
`
`Abbreviations:HL,half-life;tmax,timeatpeakplasmaconcentration.
`NOTE:meanvaluesarepresentedforallvariables.
`
`Table3.Summarystatisticsofpharmacokineticvariableestimates
`
`rapamycin concentration and noncompartmental phar-
`macokinetic analyses were conducted. Of 27 enrolled
`patients, 26 were evaluable for pharmacokinetic analyses
`(patient demographics in Table 1). There was a rapid
`decline in whole-blood levels of rapamycin in the first
`2 hours following the 30-minute infusion of nab-rapa-
`mycin, which was followed by a slower elimination phase
`(Table 3 and Fig. 1). The Cmax increased proportionally
`over the dose range of 45 to 150 mg/m2 as did the AUC,
`except for a relatively low AUC in the 125 mg/m2 dose
`cohort (Table 3).
`
`Efficacy results
`Of 19 patients evaluable for efficacy with best overall
`tumor response assessments, which included assessment
`of target, nontarget, and new lesions across all cycles, 1
`patient (5%) in the 45 mg/m2 cohort diagnosed with
`adenocarcinoma of the kidney and with bone and intra-
`thoracic metastases had a confirmed PR. The target lesion
`of this patient was reduced by 35.1% and the duration of
`response lasted 183 days. Two (11%) patients had an
`overall tumor evaluation of stable disease (confirmed): 1
`patient with mesothelioma had stable disease for 365
`days and 1 patient with a neuroendocrine tumor in the
`left axillary node had stable disease for 238 days. Eight
`patients had stable disease that could not be confirmed
`either due to absence of follow-up tumor evaluation after
`the first stable disease, or due to progression after the first
`stable disease.
`The waterfall plot in Fig. 2 illustrates the percentage
`change in the target tumors in 18 evaluable patients with
`various tumor types and histologies. Two (11%) patients
`with adenocarcinoma of the kidney had more than 30%
`decrease in the target lesion, which included the patient
`dosed at 45 mg/m2 (the patient mentioned above who had
`a confirmed PR) and another patient in the 56.25 mg/m2
`cohort, whose target lesion was reduced by 34.7% and a
`duration of response lasting 104 days. As seen in Fig. 2, 13
`(72%) patients had a target lesion objective response
`evaluation of stable disease. These patients had cancer of
`the bladder, colorectal, esophagus, head and neck, pros-
`tate, retroperitoneal, or uterus. It is notable that many of
`these patients with a target tumor evaluation of stable
`disease did not have a confirmed overall tumor evaluation
`of stable disease, which in addition to target lesions also
`accounted for nontarget and new lesions. In addition, 3
`(17%) patients had target lesion objective response of pro-
`gressive disease. As seen in Fig. 2, patients with any decrease
`in the target tumor lesion had carcinoma/adenocarcinoma
`of the kidney, bladder, esophagus, or neuroendocrine can-
`cer. Of note, the molecular analyses of the tumor biopsy
`obtained from the patient with mesothelioma achieving the
`longest clinical benefit revealed no loss in PTEN, or an
`activating mutation in PIK3CA or AKT. However, a SNP was
`observed on PHLPP2 (PH domain leucine-rich repeat pro-
`tein phosphatase 2), a gene that codes for a protein phos-
`phatase that mediates dephosphorylation of serine 473 in
`Akt1 (30).
`
`www.aacrjournals.org
`
`Clin Cancer Res; 19(19) October 1, 2013
`
`5479
`
`

`

`nab-Rapamycin treatment was associated with a signifi-
`cant decrease of S6K T389 on D2 with persistent inhibition
`at D8 (Fig. 3A and B) at all doses. nab-Rapamycin treatment
`was associated with significant decrease of 4EBP1 T70 levels
`on D2 and D4, but with recovery by D8 (Fig. 3A and C). nab-
`Rapamycin at 56.25 dose level was not associated with a
`decrease in 4EBP1 T70 levels, whereas a significant decrease
`in 4EBP1 T70 levels was seen with higher doses (Fig. 3B).
`These results show that nab-rapamycin has a dose-depen-
`dent effect on mTOR signaling, with pathway inhibition
`being seen at 56.25 mg/m2 and higher doses. The duration
`of inhibition differs between downstream targets, and is
`longer for S6K T389 than for 4EBP1 T70.
`Next, we determined whether the pharmacokinetic data
`correlated with pathway inhibition on RPPA. There was a
`moderate negative correlation between PBMC 4EBP1 T70
`levels and serum rapamycin concentrations (r2 ¼ 0.446)
`as well as between S6K T389 and serum rapamycin con-
`centration (r2 ¼ 0.517). Unfortunately, PBMCs were not
`available for the patient who had a PR. There was no
`significant difference in the inhibition 4EBP1 and S6K
`phosphorylation between patients who had stable disease
`and patients who had progressive disease.
`
`Discussion
`The results of this phase I dose-finding study showed that
`the MTD for nab-rapamycin in patients with advanced
`nonhematologic malignancies was 100 mg/m2, which pro-
`duced favorable safety profile without the DLTs typically
`
`45
`56.25
`100
`125
`150
`
`Gonzalez-Angulo et al.
`
`4,000
`
`3,000
`
`2,000
`
`1,000
`
`(ng/mL)
`
`Concentration
`
`0
`0.25 0.5 1
`
`2
`
`16 32 64 128 256
`8
`4
`Time (h)
`
`Figure 1. nab-Rapamycin plasma concentration by time.
`
`Pharmacodynamics
`PBMCs were collected from 18 patients. The effect of nab-
`rapamycin on mTOR signaling was assessed by evaluating
`the phosphorylation of mTOR targets, including 4EBP and
`S6K, and S6K target ribosomal S6 on phosphorylation sites,
`with two different assays (MSD and RPPA) previously used
`to show rapamycin-mediated inhibition of downstream
`signaling (31). The MSD phosphoprotein assays revealed
`very low baseline expression of S6 S240/244 and 4EBP T37/
`46 in pretreatment PBMC samples, thus we were unable to
`assess regulation by nab-rapamycin treatment. Therefore,
`the effect of nab-rapamycin on the functional proteomic
`profile was assessed by RPPA.
`
`Progressive disease, i.e., ≥20% increase
`
`-
`
`Stable disease
`
`-
`
`PR, i.e., ≥30 % decrease
`
`-
`
`30
`
`20
`
`10
`
`0
`
`–10
`
`–20
`
`–30
`
`Figure 2. Waterfall plot showing
`percentage change of target lesion
`from baseline by patient and tumor
`types.
`
`Percentage change of target tumor from baseline
`
`mg/m2
`
`100
`
`125
`
`45
`
`56.25
`
`45
`
`100
`
`45
`
`45
`
`100
`
`125
`
`150
`
`125
`
`100
`
`45
`
`100
`
`56.25
`
`45
`
`Kidney
`
`Kidney
`
`Bladder
`
`Esophagus
`
`Esophagus
`
`Neuroendocrine
`
`Retroperitoneal
`
`x
`
`Colorectal
`
`Colorectal
`
`Retroperitoneal
`
`Pleural mesothelioma
`
`Uterus
`
`Prostate
`
`Head and neck
`
`Head a

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