throbber
Vol. 10, 6111– 6118, September 15, 2004
`
`Clinical Cancer Research
`
`6111
`
`Phase II Study of the Proteasome Inhibitor Bortezomib (PS-341) in
`Patients with Metastatic Neuroendocrine Tumors
`
`Manisha H. Shah,1 Donn Young,2
`Hedy L. Kindler,3 Iain Webb,4 Barbara Kleiber,1
`John Wright,5 and Michael Grever1
`1Division of Hematology-Oncology, Department of Internal Medicine,
`and 2Center for Biostatistics, The Ohio State University, Columbus,
`Ohio; 3Department of Medicine, University of Chicago, Chicago,
`Illinois; 4Millennium Pharmaceuticals, Inc., Cambridge,
`Massachusetts; and 5Cancer Therapy Evaluation Program, Bethesda,
`Maryland
`
`ABSTRACT
`Purpose: This phase II study was undertaken to assess
`objective response, toxicity, tumor marker response, and
`pharmacodynamics of bortezomib in patients with meta-
`static neuroendocrine (carcinoid and islet cell) tumors.
`Experimental Design: A total of 16 patients with meas-
`urable metastatic carcinoid (n ⴝ 12) or islet cell (n ⴝ 4)
`tumors received i.v. bolus of single agent bortezomib at a
`dose of 1.5 mg/m2 on days 1, 4, 8, and 11 every 21 days.
`Tumor response was assessed at 12-week intervals using
`Response Evaluation Criteria in Solid Tumors (RECIST)
`criteria. All patients were chemotherapy naı¨ve and had
`Eastern Cooperative Oncology Group performance status of
`0 to 1.
`Results: No patient achieved a partial or a complete
`remission. The patients received total of 264 doses of therapy
`with a median of 15 doses per patient. Grade 4 toxicities
`were not observed. The most common grade 3 adverse
`events included peripheral sensory neuropathy (37%), diar-
`rhea (25%), vomiting (18%), and ileus (18%). Six of 10
`patients who experienced grade 2 to 3 peripheral sensory
`neuropathy also had grade 2 to 3 dizziness (n ⴝ 2), ortho-
`static hypotension (n ⴝ 2), syncope (n ⴝ 1), ileus (n ⴝ 2), or
`abdominal cramps (n ⴝ 1). Changes in tumor marker levels
`did not correlate with tumor response. The mean percentage
`of 20S proteasome inhibition achieved in whole blood at 1
`and 24 hours after bortezomib administration was 68 and
`30%, respectively.
`Conclusions: Despite achieving the surrogate biologic
`end point, single-agent bortezomib did not induce any ob-
`
`Received 3/2/04; revised 4/27/04; accepted 5/5/04.
`Grant support: NIH Grant CA63185.
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advertisement in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`Requests for reprints: Manisha H. Shah, The Ohio State University,
`A438 Starling-Loving Hall, 320 West Tenth Avenue, Columbus,
`OH 43210. Phone:
`(614) 293-8629; Fax:
`(614) 293-3112; E-mail:
`shah-2@medctr.osu.edu.
`©2004 American Association for Cancer Research.
`
`jective responses in patients with metastatic carcinoid or
`islet cell tumors. Additional investigation is warranted to
`clarify the possible association of autonomic neuropathy
`with bortezomib.
`
`INTRODUCTION
`Because of the pivotal role of the proteasome in controlling
`key cellular processes, the proteasome has emerged as an at-
`tractive novel target for the development of anticancer therapy
`(1). The 26S proteasome is an ATP-dependent multicatalytic
`protease that is central to the ubiquitin-proteasome-degradative
`pathway. The 26S proteasome acts as a housekeeper to eliminate
`damaged or misfolded proteins. In addition, many regulatory
`proteins governing the cell cycle, transcription factor activation,
`apoptosis, and cell trafficking, are the substrates for proteasome-
`mediated degradation (1). The proteasome plays a significant
`role in degradation of cyclin-dependent kinase inhibitors (p21,
`p27; refs. 2, 3) and tumor suppressor (p53; ref. 4) proteins that
`are required for cell cycle arrest. The proteasome is also re-
`quired for activation of transcription factor nuclear factor-␬B
`by degradation of its inhibitory protein inhibitor of nuclear
`factor-␬B (5). Nuclear factor-␬B is necessary, in part, to main-
`tain cell viability through transcription of inhibitors of apoptosis
`in response to stress or cytotoxic agents. Nuclear factor-␬B has
`also been implicated in controlling gene expression of endothe-
`lial cell surface adhesion molecules such as intercellular adhe-
`sion molecule 1, vascular cell adhesion molecule, and E-selectin
`(6), which are involved in tumor metastasis and angiogenesis.
`Furthermore, the proteasome is implicated in in vivo angiogen-
`esis, a phenomenon essential for tumor growth and metastasis
`(7).
`
`Bortezomib [Velcade (Millennium Pharmaceuticals, Inc.,
`Cambridge, MA), previously known as PS-341] is a dipeptidyl
`boronic acid that is a specific, potent and reversible inhibitor of
`the 26S proteasome. Preclinical work revealed significant anti-
`tumor activity of bortezomib in vitro and in vivo. In the cell line
`screen of the National Cancer Institute, bortezomib demon-
`strated a unique pattern of growth inhibitory and cytotoxic
`activity against a broad range of solid tumors (8). Bortezomib
`also significantly reduced tumor volumes in several murine and
`human tumor xenograft models including the Lewis lung,
`HT-29 human colon, PC-3 human prostate, squamous cell car-
`cinoma, and pancreatic cancer (9 –13). The initial dose and
`schedule of bortezomib for phase I clinical trials was chosen
`based on the acute and multiple dose toxicity studies performed
`on rodents and primates (14). Preclinical studies in animals
`showed that bortezomib was rapidly removed from the vascular
`compartment and distributed widely, quickly approaching the
`limits of detection. Therefore, a sensitive, accurate and repro-
`ducible ex vivo 20S proteasome inhibition assay to monitor
`bortezomib activity in whole blood was developed (15). Several
`phase I and/or II clinical trials done to date showed that the
`
`Downloaded from
`
`
`on July 27, 2017. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`Research.
`
`Ex. 1112-0001
`
`

`

`6112 Bortezomib in Metastatic Neuroendocrine Tumors
`
`target proteasome inhibition of ⬃70% can be achieved in cancer
`patients with a reasonable safety profile. Moreover, complete
`responses, partial responses, or minor responses were observed
`in the variety of solid tumors and hematologic malignancies
`(16 –20). Bortezomib was recently approved by the Food and
`Drug Administration for its use in the patients with refractory or
`relapsed multiple myeloma.
`Carcinoid and islet cell tumors are generally classified at
`the less-aggressive end of the spectrum of neuroendocrine tu-
`mors. They are thought to derive from enterochromaffin cells,
`which are distributed throughout the gastrointestinal and respi-
`ratory system. Although plasma chromogranin A and 24-hour
`urine for 5-hydroxy indole acetic acid are known prognostic
`markers for survival in patients with carcinoid tumors, the role
`of other tumor markers is not well described (21, 22). Although
`local or locoregional carcinoid or islet cell tumors are surgically
`manageable, metastatic disease is present in ⬃50% of patients at
`the time of diagnosis, and the overall 5-year survival for patients
`with distant metastasis is only 22%. Somatostatin analogues,
`IFN-␣ or hepatic artery chemoembolization provide good pal-
`liation of the symptoms of carcinoid syndrome associated with
`such tumors. However, no systemic therapy has been shown to
`consistently elicit tumor responses and prolong survival. The
`ineffectiveness of systemic chemotherapy in metastatic neu-
`roendocrine tumors may relate to their unique biological fea-
`tures such as slow growth pattern or hypervascularity.
`Preclinical in vitro and in vivo work suggests that bort-
`ezomib has antitumor activity in a broad spectrum of solid
`tumors, including the PC-12 neuroendocrine (pheochromocy-
`toma) tumor cell line (23). Although primarily targeting the
`proteasome pathway, bortezomib has been shown to affect key
`proteins involved in cell cycle regulation, angiogenesis, and
`apoptosis. On the basis of this and the unique antitumor mech-
`anisms of bortezomib, we hypothesize that it may have clinical
`activity in metastatic carcinoid and islet cell tumors. We thus
`conducted a phase II clinical trial in patients with metastatic
`carcinoid and islet cell tumors. On the basis of toxicity and
`pharmacodynamic data available from initial phase I clinical
`trials in solid tumor and hematologic malignancies, the dose of
`1.5 mg/m2 administered as an i.v. bolus on days 1, 4, 8, and 11
`every 21 days was considered optimal for our trial. We report
`the results of the first prospective phase II study of bortezomib
`in patients with metastatic neuroendocrine tumors.
`
`PATIENTS AND METHODS
`Patient Selection. Eligibility criteria included histologi-
`cally confirmed well-differentiated neuroendocrine carcinoma;
`metastatic and measurable disease; no more than one prior
`systemic chemotherapy regimen; no IFN-␣, systemic chemo-
`therapy, or radiation therapy within the past 4 weeks; no hepatic
`artery chemoembolization within the past 12 weeks; age ⱖ 18
`years; Eastern Cooperative Oncology Group performance status
`0 to 2; life expectancy ⱖ 6 months; and adequate organ func-
`tions: total bilirubin ⱕ 1.5 mg/dL, aspartate aminotransferase/
`alanine aminotransferase ⱕ 2.5⫻ upper limit of normal, abso-
`lute neutrophil count ⱖ 1500/␮L, platelet count ⱖ 100,000/␮L,
`and serum creatinine of ⱕ1.5 mg/dL. The standard dose (10 to
`40 mg every 3 to 4 weeks) of intramuscular long-acting oct-
`
`reotide [Sandostatin LAR (Novartis Pharmaceuticals Co., East
`Hanover, NJ)] was permitted for the control of carcinoid syn-
`drome symptoms provided that the dose was stable for 3 months
`before study entry. Exclusion criteria were pregnancy, breast-
`feeding, and uncontrolled intercurrent illness.
`Study Design. Bortezomib was administered as an i.v.
`bolus at a dose of 1.5 mg/m2 on days 1, 4, 8, and 11 every 21
`days. Antiemetic therapy or growth factor support was not used
`on a prophylactic basis. Therapy was given for a total of 24
`weeks unless patients met one of the following criteria: progres-
`sive disease; unacceptable adverse event; off of study drug for
`⬎2 weeks; or patient withdrawal from the study. Patients were
`maintained at the full dose of 1.5 mg/m2 unless patients had
`grade 3 or 4 adverse event. In case of grade 3 or 4 adverse event,
`treatment was withheld for 1 week. Upon reevaluation, patients
`who showed a partial (improvement to grade 1 or 2 adverse
`event) or a full resolution were started on bortezomib at the
`reduced dose of 1.0 mg/m2 that was escalated to 1.3 mg/m2 after
`1 week and to 1.5 mg/m2 after the second week as tolerated. The
`patients with persistent grade 3 or 4 adverse event were taken
`off of the protocol.
`History, physical examination, complete blood count, and
`serum chemistry were obtained within 14 days before initiation
`of therapy and within 2 to 4 weeks after the last dose of
`bortezomib (posttreatment). Baseline tumor measurements were
`performed with computed tomography scan within 4 weeks
`before initiation of therapy and repeated every 12 weeks during
`study and at the posttreatment visit. In addition, patients had a
`history and physical examination performed every 6 weeks and
`had complete blood count, serum chemistries, and adverse event
`evaluation performed once a week during their treatment weeks
`(week 1, 2, 4, 5, and so on). Objective response was assessed
`according to Response Evaluation Criteria in Solid Tumors
`(RECIST) criteria, and toxicity was assessed according to Na-
`tional Cancer Institute Common Toxicity Criteria, version 2.0
`(24).6
`Tumor Marker Studies. Serum tumor markers (pancre-
`astatin, gastrin, calcitonin, pancreatic polypeptide, vasoactive
`intestinal peptide, neurotensin, substance-P, gastrin-releasing
`peptide, glucagon, somatostatin, and serotonin) and 24-hour
`urine for 5-hydroxy indole acetic acid were obtained within 14
`days before initiation of therapy every 12 weeks during therapy
`and within 2 to 4 weeks posttreatment.
`Pharmacodynamic Studies. We used an ex vivo 20S
`proteasome inhibition assay to monitor bortezomib activity in
`whole blood using the previously described method (15). This
`fluorogenic kinetic assay consisted of measuring proteasome
`activity at chymotryptic and tryptic sites within the 20S core of
`the proteasome and determining the degree of inhibition con-
`ferred by bortezomib. Using the ratio of these chymotryptic:
`tryptic activities and the catalytic mechanism of the proteasome,
`percentage of inhibition was calculated compared with predose
`whole blood obtained from the same patient.
`Blood samples were collected at 0 hour (just before admin-
`istration of bortezomib), 1 hour, and 24 hours after bortezomib
`
`6 Internet address: http://ctep.info.nih.gov/reporting/ctc.html.
`
`Downloaded from
`
`
`on July 27, 2017. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`Research.
`
`Ex. 1112-0002
`
`

`

`Clinical Cancer Research
`
`6113
`
`on days 1 and 4 of the first course of therapy. For each time
`point, 8-mL of blood were collected in one heparinized green
`top tube, immediately placed on ice, and then frozen at ⫺70°C.
`Batched samples were shipped on dry ice to Millennium Phar-
`maceuticals, Inc., for performing an assay. On the basis of the
`preclinical efficacy and toxicity studies, a target level of ⬃70%
`proteasome inhibition was considered optimal.
`Statistical Considerations. For this phase II study, the
`minimax two-stage design of Simon was chosen, resulting in a
`trial with a decision to proceed to the second stage based on
`efficacy seen in the first 16 patients. Bortezomib was to be
`considered ineffective or uninteresting if the true response prob-
`ability was ⬍10% (p0). The regimen was to be worthy of further
`study if the true response probability or target response rate was
`ⱖ30% (p1). These figures resulted in a two-stage design of 16
`and 25 patients, with an ␣ of 0.10 and ␤ of 0.10. If one or no
`responses were seen in the first 16 patients, the study was to be
`terminated early, and this regimen was deemed ineffective for
`this patient population. If two or more patients respond in the
`first 16, an additional 9 patients were to be treated for a total
`of 25.
`The primary end point was to assess the objective response
`(partial remission or complete remission) of bortezomib in met-
`astatic carcinoid and islet cell tumors. Stable disease was not
`considered an objective response to therapy given the relatively
`slow-growing nature of these cancers and due to a single-arm
`study design. Patients who had stable disease were taken off
`from the protocol at 24 weeks of therapy. Secondary end points
`were to assess the toxicity of bortezomib in patients with met-
`astatic neuroendocrine tumors, to assess tumor marker response,
`and to evaluate pharmacodynamics of bortezomib by ex vivo
`20S proteasome inhibition assay.
`
`RESULTS
`Patients. After obtaining informed consent, 16 patients
`with metastatic neuroendocrine tumor were enrolled on to the
`Institutional Review Board-approved phase II study at The Ohio
`State University (n ⫽ 14) and at University of Chicago (n ⫽ 2)
`between May 2001 and December 2001. Patient characteristics
`are outlined in Table 1. A majority of patients (n ⫽ 12; 75%)
`had carcinoid tumors, whereas the rest (n ⫽ 4, 25%) had islet
`cell tumors. Of the four patients with islet cell tumors, one
`patient had symptoms of diarrhea related to carcinoid syndrome,
`whereas the rest of the patients had no symptoms associated
`with abnormally high hormone production (pancreatic polypep-
`tide, gastrin, glucagon, or calcitonin) related to their islet cell
`tumors.
`Treatment Administered.
`If the best response achieved
`was stable disease, treatment with bortezomib was continued for
`up to 24 weeks (32 doses). All 16 patients received median of 15
`doses (range, 1 to 32 doses), with a median cumulative total
`dose of 21.8 (range, 1.5 to 48) mg/m2. Of the total 264 doses
`given in the study, 42 doses in seven patients were given at the
`reduced doses per protocol guidelines because of grade 3 ad-
`verse events [neutropenia (n ⫽ 2), peripheral neuropathy (n ⫽
`2), ileus (n ⫽ 2), and thrombocytopenia (n ⫽ 1)] or recurrent
`grade 2 vomiting. The median duration of therapy for all pa-
`tients was 12 weeks (range, 0.5 to 24 weeks) and only 4 of 16
`
`Table 1 Patient characteristics
`
`Characteristic
`
`Total patients
`Age (y)
`Median
`Range
`Sex
`Female
`Male
`Eastern Cooperative Oncology Group
`performance status
`0
`1
`Type of neuroendocrine tumor
`Carcinoid tumor
`Islet cell tumor
`Primary sites
`Pancreas
`Ileocecal
`Colon
`Stomach
`Lung
`Unknown
`Metastatic sites
`Liver alone
`Liver and lymph nodes
`Liver, lymph nodes, and bone
`Carcinoid syndrome/concurrent octreotide therapy
`Present
`Absent
`Prior treatment
`Debulking or palliative surgery
`Hepatic artery chemoembolization
`Systemic chemotherapy
`Baseline elevation of serum tumor markers
`Pancreastatin
`Glucagon
`Neurotensin
`Gastrin
`Pancreatic polypeptide
`Calcitonin
`
`No.
`16
`
`60
`26–72
`
`10
`6
`
`13
`3
`
`12
`4
`
`4
`7
`1
`1
`1
`2
`
`6
`6
`4
`
`6
`10
`
`11
`6
`0
`
`13
`5
`5
`4
`2
`2
`
`%
`
`62
`38
`
`81
`19
`
`75
`25
`
`25
`43
`6
`6
`6
`12
`
`37
`37
`25
`
`38
`62
`
`68
`38
`0
`
`81
`31
`31
`25
`12
`12
`
`patients (25%) completed 24 weeks of therapy. The reasons for
`leaving the study before 24 weeks of planned therapy in the 12
`patients included progressive disease (n ⫽ 3), persistent grade 3
`adverse events [peripheral neuropathy (n ⫽ 2), diarrhea (n ⫽ 1),
`fatigue (n ⫽ 1)], and patient withdrawal [grade 3 hypertension/
`atrial fibrillation (n ⫽ 1); grade 2 nausea (n ⫽ 1), vomiting (n ⫽
`1) or seizure (n ⫽ 1); and social reason (n ⫽ 1)] (Fig. 1).
`Objective Response. All 16 patients were considered
`evaluable for response in an intent-to-treat analysis. No pa-
`tients achieved a partial response or a complete response.
`Stable disease was noted in 11 of 16 patients (69%) at the
`median evaluation time of 12 weeks (range, 3 to 24 weeks)
`after the start of therapy. Five patients (31%) had progressive
`disease noted at weeks 0.5 (n ⫽ 1), 12 (n ⫽ 2), 16.5 (n ⫽ 1),
`and 24 (n ⫽ 1).
`Tumor Marker Response. Serum pancreastatin was
`consistently elevated at the prestudy evaluation in 13 patients,
`whereas other tumor markers (pancreatic polypeptide, gastrin,
`glucagon, calcitonin, or neurotensin) were elevated in only a
`few patients at baseline (Table 1). Tumor markers were evalu-
`able in 10 patients, and marker levels did not correlate with
`
`clincancerres.aacrjournals.org Downloaded from
`
`on July 27, 2017. © 2004 American Association for Cancer
`
`Research.
`
`Ex. 1112-0003
`
`

`

`6114 Bortezomib in Metastatic Neuroendocrine Tumors
`
`Fig. 1 Treatment administered and reasons for off study. f indicates maximum doses planned,
`indicates individual patient and data label indicates
`the reason for off study (type of response). PD, progressive disease; SD, stable disease; HTN, hypertension, AF, atrial fibrillation.
`
`objective tumor response in most patients. Table 2 summarizes
`serum pancreastatin results in 10 evaluable patients. Of eight
`patients who showed stable disease, three patients had an 11 to
`24% reduction in serum pancreastatin, whereas five patients had
`an increase (median increase, 44%; range, 7 to 55%) in serum
`pancreastatin compared with the prestudy levels. Serum pancre-
`astatin was increased by 13 to 18% as compared with baseline
`in patients who showed progressive disease.
`
`Pharmacodynamic Studies. A pharmacodynamic assay
`was performed to correlate the degree of proteasome inhibition
`with tumor response or adverse events. The percentage of 20S
`proteasome inhibition was compared with the predose (0 hour)
`value obtained on the same day in each individual patient.
`Peripheral blood samples were evaluable in 15 of total 16
`patients on the study. The mean percentage of 20S proteasome
`inhibition achieved in whole blood at 1 and 24 hours after
`
`Table 2 Correlation of serum tumor marker response with objective response
`Serum pancreastatin (pg/mL) (normal value ⬍ 135 pg/mL)
`
`Subject
`Posttherapy
`Pretherapy
`no.
`755
`851
`105
`260
`346
`110
`1,200
`1,580
`113
`1,620
`1,520
`112
`1,420
`1,070
`108
`8,210
`12,500
`107
`853
`555
`104
`8,790
`5,650
`101
`389
`343
`106
`570
`484
`103
`Abbreviations: SD, stable disease; PD, progressive disease.
`
`% change with therapy
`⫺11
`⫺21
`⫺24
`7
`33
`44
`54
`55
`13
`18
`
`Best clinical response
`SD
`SD
`SD
`SD
`SD
`SD
`SD
`SD
`PD
`PD
`
`Downloaded from on July 27, 2017. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`
`
`
`Research.
`
`Ex. 1112-0004
`
`

`

`Clinical Cancer Research
`
`6115
`
`bortezomib. Moreover, one patient had neuropathy symptoms
`starting at 3 weeks after the last of bortezomib. Grade 3 neu-
`ropathy symptoms in this patient improved to grade 2 and to
`grade 1 at week 12 and at week 18 after the last of bortezomib,
`respectively, despite subsequent treatment with etoposide and
`cisplatin. Although peripheral neuropathy is a common side
`effect of cisplatin, it is usually cumulative and progressive
`during the course of cisplatin chemotherapy. Thus, spontaneous
`improvement of neuropathy from grade 3 to grade 1 in our
`patient during the six cycles of cisplatin-based chemotherapy
`indicates its likely attribution to bortezomib. Seven of 10 pa-
`tients had improvement up to grade 1 (n ⫽ 5) or grade 0 (n ⫽
`2) peripheral neuropathy at a median of 18 weeks (range, 2 to 76
`weeks) after the last dose of bortezomib.
`
`DISCUSSION
`Given the broad spectrum of in vitro and in vivo antineo-
`plastic activity and safety of single-agent bortezomib, we stud-
`ied its efficacy in patients with metastatic neuroendocrine tu-
`mors. Despite achieving the surrogate biological end point of
`20S proteasome inhibition in peripheral blood mononuclear
`cells obtained from 15 patients, our study failed to show any
`objective tumor response in this patient population. Although
`stable disease was noted in 11 of 16 patients (69%) at the
`median evaluation time of 12 weeks (range, 3 to 24 weeks) after
`
`Table 3 Bortezomib-related adverse events reported in
`all 16 patients
`
`Adverse event
`Gastrointestinal
`Anorexia
`Nausea
`Vomiting
`Diarrhea
`Constipation
`Ileus
`Abdominal cramps
`Dehydration
`Dysgeusia
`Neurological
`Neuropathic pain
`Neuropathy-sensory
`Dizziness
`Syncope
`Seizure
`Constitutional
`Fatigue
`Weight loss
`Hematologic
`Neutropenia
`Thrombocytopenia
`Others
`Skin rash
`Mouth sores (cold sores)
`Infection without neutropenia
`Hyperglycemia
`Orthostatic hypotension
`Hypertension
`Atrial fibrillation
`Conjunctivitis
`
`Number (%)
`
`Grade 1
`
`Grade 2
`
`Grade 3
`
`4 (25)
`1 (6)
`1 (6)
`3 (18)
`1 (6)
`0
`0
`0
`3 (18)
`
`0
`0
`1 (6)
`0
`0
`
`7 (43)
`2 (12)
`
`0
`1 (6)
`
`2 (12)
`0
`1 (6)
`1 (6)
`0
`0
`0
`0
`
`5 (31)
`9 (56)
`3 (18)
`1 (6)
`4 (25)
`0
`0
`3 (18)
`1 (6)
`
`4 (25)
`4 (25)
`1 (6)
`0
`1 (6)
`
`5 (31)
`1 (6)
`
`1 (6)
`1 (6)
`
`2 (12)
`3 (18)
`2 (12)
`1 (6)
`2 (12)
`0
`0
`0
`
`0
`2 (12)
`3 (18)
`4 (25)
`0
`3 (18)
`2 (12)
`0
`0
`
`6 (37)
`6 (37)
`0
`1 (6)
`0
`
`1 (6)
`0
`
`2 (12)
`2 (12)
`
`0
`0
`0
`1 (6)
`0
`1 (6)
`1 (6)
`1 (6)
`
`Fig. 2 Pharmacodynamic assay for bortezomib (n ⫽ 15). Percentage of
`20S proteasome inhibition in whole blood at indicated time points
`during the first course of bortezomib (mean ⫾ SD). Percentage of 20S
`proteasome inhibition is compared with the predose value obtained on
`the same day in each individual patient.
`
`bortezomib administration on day 1 was 68 (SD ⫾6) and 30 (SD
`⫾6), respectively. Similar results were achieved on day 4 of
`bortezomib administration (Fig. 2). The target level of 20S
`proteasome inhibition was achieved in our study patients, and
`there was no correlation with type or degree of adverse events or
`clinical outcome.
`Adverse Events. Bortezomib was generally well toler-
`ated. There were no grade 4 adverse events or deaths related to
`the study. All of the drug-related adverse events reported in the
`16 study patients are outlined in Table 3. The worst grade
`experienced by individual patient was used to report the adverse
`events. Although the most common adverse event seen was
`nausea (74%), the most common grade 3 toxicities were periph-
`eral sensory neuropathy (37%), followed by diarrhea (25%). It is
`noteworthy that four patients (25%) developed viral-associated
`infections during the study period. Three of these patients had
`recurrent grade 2 cold sores, whereas one patient had grade 3
`conjunctivitis in absence of neutropenia. Three patients (18%)
`had ileus occur at the cumulative dose (mg/m2) of 6, 7.5, or
`10.5. All of these patients had prior laparotomy, two patients
`had active mesenteric carcinoid tumor, and one patient was on
`a stable dose of tincture of opium. Ileus resolved within 2 to 3
`days of hospitalization with conservative treatment, and patients
`subsequently received one, three, or eight doses of bortezomib
`without recurrent ileus. Of the two patients who experienced
`grade 3 abdominal cramps, one had ileus as noted above.
`A drug-related grade 2 to 3 peripheral sensory neuropathy
`manifested by pain, tingling, and numbness occurred in 10
`patients (62%). In general, tingling and numbness were more
`intense in the feet and legs than in the hands, whereas neuro-
`pathic pain was mainly localized to legs and feet. All of these
`patients required narcotics and gabapentin for pain control. The
`median cumulative dose at the onset of neuropathy was 13.3
`(range, 1.5 to 28.5) mg/m2 (Table 4). Five of these patients had
`worsening of neuropathy during the off-study period at a median
`of 3.5 weeks (range, 3 to 9 weeks) after the last dose of
`
`clincancerres.aacrjournals.org Downloaded from
`
`on July 27, 2017. © 2004 American Association for Cancer
`
`Research.
`
`Ex. 1112-0005
`
`

`

`6116 Bortezomib in Metastatic Neuroendocrine Tumors
`
`Table 4 Analysis of bortezomib-associated neurotoxicity
`
`Subject
`no.
`101
`
`Total cumulative
`dose (mg/m2)
`17.4
`
`Worst grade
`of PSN
`3
`
`Grade of PSN
`at recovery
`1
`
`3
`
`1
`
`3
`
`1
`
`1
`
`102
`
`27.3
`
`103
`
`104
`
`105
`
`21.1
`
`10.5
`
`15
`
`106
`
`22.5
`
`107
`
`24
`
`108
`
`109
`110
`
`48
`
`36
`39.3
`
`3
`
`3
`
`3
`
`3
`
`3
`
`2
`
`2
`
`2
`2
`
`Cumulative total dose at
`the onset of PSN
`16.1 mg/m2: grade 2
`17.4 mg/m2: grade 3
`10.5 mg/m2: grade 1
`
`13.5 mg/m2: grade 2
`21.3 mg/m2: grade 3
`10.5 mg/m2: grade 1
`
`Status of PSN during
`off study period
`Wk after the last
`dose of bortezomib:
`grade of PSN
`Wk 7: grade 2
`Wk 12: grade 1
`Wk 8: grade 3
`
`Wk 3–5: grade 3
`
`18.1 mg/m2: grade 2
`6 mg/m2: grade 2
`
`Wk 24: grade 1
`Wk 3–102: grade 3
`
`1.5 mg/m2: grade 1
`
`Wk 6–34: grade 3
`
`12 mg/m2: grade 2
`
`No symptoms during
`study
`
`Wk 56: grade 2
`Wk 76: grade 1
`Wk 3 (d 7, 1st cycle
`VP/cis): grade 3
`Wk 12 (4th cycle
`VP/cis): grade 2
`Wk 18 (6th cycle
`VP/cis): grade 1
`Wk 23–27: grade 1
`Wk 2: grade 1
`
`Wk 4–12: grade 2
`Wk 16: grade 1 T/N
`Wk 16: grade 0 pain
`Wk 2–30: grade 0
`
`Wk 9–20: grade 2
`Wk 12: grade 1
`
`1 T/N
`
`10.5 mg/m2: grade 1
`
`0 pain
`
`0
`
`2
`0
`
`21 mg/m2: grade 1
`27 mg/m2: grade 2
`28.5 mg/m2: grade 1
`27 mg/m2: grade 1
`
`Symptoms possibly related to
`autonomic neuropathy
`(Cumulative dose of
`bortezomib at onset)
`
`Grade 2 orthostatic
`hypotension (18 mg/m2)
`
`Grade 1 dizziness, grade 3
`ileus (10.5 mg/m2)
`
`Grade 3 ileus (6 mg/m2)
`
`Grade 2 dizziness
`(12 mg/m2)
`
`Grade 2 orthostatic
`hypotension, grade 3
`syncope (21 mg/m2)
`
`Grade 3 abdominal cramps
`(3 mg/m2)
`
`Grade 3 ileus (7.5 mg/m2)
`
`Wk 24: grade 0
`Lost to follow up
`0
`8.5
`111
`Abbreviations: VP, VP-16; cis, cisplatin; PSN, peripheral sensory neuropathy; T/N, tingling/numbness.
`
`28.5 mg/m2: grade 2
`
`the start of therapy, our study was not designed to evaluate
`progression-free survival. Given the slow growing nature of
`these tumors, we cannot attribute stable disease to the antitumor
`effect of bortezomib in our single-arm study. Given that protea-
`some inhibition in tumor tissue was not evaluated in our study,
`it is conceivable that the ineffectiveness of bortezomib may
`relate to an insufficient level or duration of 26S proteasome
`inhibition in the tumors. Although bortezomib affects the nu-
`clear factor-␬B pathway, cyclin-dependent kinase inhibitors
`(e.g., p21, p27), or tumor suppressor (e.g., p53) proteins, it is
`possible that redundant pathways exist for tumor progression
`and survival of metastatic neuroendocrine tumors.
`Bortezomib was generally well tolerated at the dose and
`schedule used in our study. Similar to previously reported
`studies, the most common adverse events in this study in-
`cluded gastrointestinal, neurologic, constitutional, and hema-
`tologic. However, we observed a higher (37 versus 12%)
`
`incidence of grade 3 peripheral sensory neuropathy in our
`chemo-naı¨ve patients compared with the phase II trial of
`bortezomib in multiple myeloma (16). It is unclear if grade 3
`pain in limb reported in 7% of their study patients was
`neuropathic pain. Given that 86% of patients in the reported
`myeloma study had prior thalidomide (and likely Vinca al-
`kaloids), it is possible that an attribution of neuropathy to
`bortezomib might have been difficult. Although the median
`duration of therapy administered in both studies was roughly
`similar (12 versus 15 weeks), the dose of bortezomib was
`higher (1.5 versus 1.3 mg/m2/dose) in our study. Thus, it is
`possible that the higher dose may account for the higher
`incidence of neuropathy seen in our study. A recently re-
`ported phase II study used a dose and schedule similar to our
`study and reported only 9% grade 3 peripheral neuropathy
`but 47% incidence for all grades of neuropathy (25). Al-
`though we did not evaluate predisposing factors for neurop-
`
`Downloaded from
`
`
`on July 27, 2017. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`Research.
`
`Ex. 1112-0006
`
`

`

`Clinical Cancer Research
`
`6117
`
`athy in a prospective manner, no obvious risk factors (such as
`diabetes mellitus, concomitant or prior neurotoxic medica-
`tions, or B12 deficiency) were found in our patients. Periph-
`eral sensory neuropathy was generally dose related, cumula-
`tive, and reversible up to less than or equal to grade 1 (Table
`4). The median time in improvement or resolution of neu-
`ropathy was 18 weeks (range, 2 to 76 weeks) after the last
`dose of bortezomib. However, patients were not required to
`follow-up at the specific intervals during the off-study pe-
`riod. The clinical pattern seen in our patients with peripheral
`neuropathy was most consistent with small-fiber painful sen-
`sory neuropathy where A-␦ (small myelinated) and nocicep-
`tive C (unmyelinated) nerve fibers are affected. With the
`striking association of peripheral neuropathy with bort-
`ezomib, it is possible that the ileus, abdominal cramps, syn-
`cope, dizziness, and orthostatic hypotension seen in seven of
`our patients (43%) could be a result of bortezomib-related
`autonomic neuropathy. Six of seven patients who developed
`such symptoms also developed grade 2 to 3 peripheral neu-
`ropathy during the course of their therapy. Although existing
`predisposing factors (e.g., prior laparotomy, narcotic use, or
`active bowel carcinoid tumor) could explain the occurrence
`of ileus in three of our patients (18%), possible association of
`ileus with bortezomib cannot be excluded. Interestingly,
`there were eight patients on the study who had prior laparot-
`omy for tumor debulking who did not develop ileus. The high
`incidence of nausea and vomiting seen our study suggests
`that patients might benefit from prophylactic antiemetic ther-
`apy. Although the given patient population could have diar-
`rhea because of carcinoid syndrome, the baseline symptoms
`were taken into consideration in grading diarrhea, and no
`simultaneous worsening of other carcinoid syndrome symp-
`toms was seen. Hematologic toxicity was of short duration
`alleviating the need for transfusion or growth factor support.
`We also question whether bortezomib administration might
`be associated with viral reactivation given that 25% of our
`patients had either cold sores or conjunctivitis in the absence
`of neutropenia. Of note, we did not observe any of the
`electrolyte abnormalities seen in phase I studies.
`In conclusion, single-agent bortezomib does not have ac-
`tivity in patients with metastatic neuroendocrine tumors. Bort-
`ezomib is safe at the schedule and dose used in our study, with
`the most significant clinical adverse event being a peripheral
`sensory neuropathy. The detailed evaluation of bortezomib-
`associated neurotoxicity, including autonomic neuropathy and
`value of prophylactic strategies, merits future investigation.
`These studies will be particularly helpful because this agent may
`be used with other neurotoxic agents in the future. Furthermore,
`with growing preclinical data demonstrating synergism between
`bortezomib and cytotoxic chemotherapy (26 –30), future studies
`should evaluate bortezomib in combination with chemotherapy
`in solid tumors but caution should be taken in combining chem-
`otherapy that may have overlapping gastrointestinal or neuro-
`logic toxicity.
`
`REFERENCES
`1. Adams J. Proteasome inhibitors as new anticancer drugs. Curr Opin
`Oncol 2002;14:628 –34.
`
`2. Blagosklonny MV, Wu GS, Omura S, el-Deiry WS. Proteasome-
`dependent regulation of p21WAF1/CIP1 expression. Biochem Biophys
`Res Commun 1996;227:564

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