throbber
CANCER TREATMENT REVIEWS 2003; 29 (Suppl. 1): 33–39
`doi:10.1016/S0305-7372(03)00080-X
`
`Clinical update: proteasome inhibitors in
`hematologic malignancies
`
`Paul Richardson
`
`Dana-Farber Cancer Institute, Boston, MA 02115, USA
`
`The proteasome inhibitor bortezomib (VELCADE; formerly PS-341, LDP-341, MLN341) is a novel dipeptide boronic acid. In
`cell culture and xenograft models, bortezomib showed potent activity, enhanced the sensitivity of cancer cells to traditional
`chemotherapeutics, and appeared to overcome drug resistance. In vitro, bortzomib downregulated the NF-jB pathway. NF-
`jB is a transcription factor that enhances the production of growth factors (e.g., IL-6), cell-adhesion molecules, and anti-ap-
`optotic factors, all of which contribute to the growth of the tumor cell and/or protection from apoptosis. Phase II trials have
`been conducted in patients with relapsed and refractory multiple myeloma (SUMMIT trial, 202 patients) or relapsed my-
`eloma (CREST trial, n ¼ 54) using a 1.3 mg=m2 dose given twice weekly for 2 weeks (days 1, 4, 8, 11; rest days 12–21).
`Both trials showed responses (including complete responses) with manageable toxicities, forming the basis for an ongoing
`phase III trial comparing response to bortezomib versus high-dose dexamethasone.
`C 2003 Elsevier Science Ltd. All rights reserved.
`
`Key words: Proteasome inhibition; bortezomib; multiple myeloma; hematologic malignancies.
`
`INTRODUCTION
`
`The proteasome is a multicatalytic enzyme complex
`that degrades numerous types of proteins (reviewed
`by Adams in this issue), many of which are regula-
`tory proteins that control the cell cycle or play a role
`in survival pathways. The coordinated expression
`and degradation of these proteins are essential for
`normal cellular function. Consequently, the protea-
`some plays a central role in up- or down-regulation
`of growth signaling pathways by removing key
`signals via protein degradation. Inhibition of the
`proteasome is therefore a promising approach for
`the treatment of cancer, a disease characterized by
`defects in growth signaling pathways that promote
`the hyperproliferation of aberrant cells.
`
`The proteasome inhibitor, bortezomib (VEL-
`CADE; formerly PS-341, LDP-341, MLN341), is a
`novel dipeptide boronic acid small molecule that has
`shown antitumor activity in preclinical studies and
`is the first such agent to have progressed to clinical
`trials. A phase III trial in patients with multiple
`myeloma (MM) is ongoing and several trials in pa-
`tients with other hematologic malignancies or solid
`tumors are in progress. Here, we briefly review the
`preclinical rationale for bortezomib and the clinical
`trial status of bortezomib in patients with hemato-
`logic malignancies, with a focus on MM.
`
`PRECLINICAL RATIONALE FOR
`PROTEASOME INHIBITORS IN
`HEMATOLOGIC MALIGNANCIES
`
`Correspondence to: Paul Richardson MD, Jerome Lipper Multiple
`Myeloma Center, Division of Hematologic Oncology, Dana-Farber
`Cancer Institute, Harvard Medical School, Boston, MA 02115,
`USA. Tel.: 1-617-632-2104; Fax: 1-617-632-6624; E-mail: paul_
`richardson@dfci.harvard.edu
`
`In cell culture and in xenografted tumors, bortezo-
`mib had potent activity, enhanced the sensitivity of
`cancer cells to traditional tumoricidal chemothera-
`peutics (1–4) and appeared to overcome drug resis-
`tance (5). This activity is at least in part mediated by
`
`0305-7372/03/$ - see front matter C 2003 ELSEVIER SCIENCE LTD. ALL RIGHTS RESERVED.
`
`CFAD v. Anacor, IPR2015-01776
`ANACOR EX. 2011 - 1/7
`
`

`
`34
`
`P . R I C H A R D S O N
`
`the activity of bortezomib against the NF-jB path-
`way. The NF-jB pathway is constitutively active in
`some cancer cells and is associated with prolifera-
`tion, cell survival, and protection from chemothera-
`py-induced apoptosis
`(Figure
`1). Proteasome
`inhibition has also been shown to block chemother-
`apy- and radiotherapy-induced activation of NF-jB
`in laboratory studies, resulting in enhanced sensi-
`tivity to these tumoricidal agents and increased ap-
`optosis in cancer cells (1–3,6).
`Activation of NF-jB signaling appears to be par-
`ticularly important for the survival of multiple my-
`eloma cells (MMCs): MMCs isolated from patients
`had constitutive NF-jB activity, MMCs and BMSCs
`showed enhanced NF-jB activity, and chemoresis-
`tant MMCs had increased NF-jB activity compared
`with chemosensitive lines (5,7). Bortezomib ap-
`peared not only to have activity against MMCs, but
`also to downregulate protective interactions with
`bone marrow stromal cells (BMSCs) in the bone
`marrow microenvironment (4), and to inhibit blood
`vessel development
`(8). Thus, bortezomib acts
`against MMCs by antagonizing the activation of
`protective NF-jB functions in not only MMCs but
`BMSCs.
`Myeloma growth and resistance pathways be-
`come activated when MMCs bind to normal BMSCs.
`Myeloma cells express VLA-4 and VLA-5, which are
`receptors that allow myeloma to bind to the VCAM-
`1 receptor on BMSCs. Damiano and colleagues
`
`demonstrated that binding VLA receptors with fi-
`bronectin conferred protection against apoptosis,
`and Chauhan and colleagues have shown that the
`binding of myeloma cells to BMSCs resulted in the
`NF-jB-dependent secretion of
`IL-6 from BMSCs
`(9,10). These studies suggested that the interactions
`between myeloma cells and normal cells in the
`bone marrow microenvironment are important for
`the activation of resistance or growth-promoting
`mechanisms.
`Preclinical studies with bortezomib showed direct
`cytotoxic activity against MMCs in vitro. In addition,
`bortezomib inhibited growth of dexamethasone-,
`doxorubicin-, and melphalan-resistant myeloma
`cells lines (4), reduced the expression of the BMSC
`adhesion receptor VCAM-1 (11), decreased adher-
`ence of myeloma cells to BMSCs (presumably due to
`downregulation of adhesion molecules), suppressed
`the NF-jB-dependent expression of IL-6 (a myeloma
`growth factor) (4), and had in vivo activity against
`human myeloma xenografts
`(8). Synergy with
`doxorubicin and melphalan but not dexametha-
`sonewas observed in sensitive MMCs since low
`concentrations of bortezomib significantly reduced
`the LD50 of either of these chemotherapies (12). In
`addition, MMCs from a heavily treated patient prior
`regimens included conventional and high dose
`chemotherapy, interferon-a, thalidomide, liposomal
`doxorubicin, and bortezomib as a single agent and in
`combination with dexamethasonedid not respond
`
`Figure 1 Downregulation of the NF-jB pathway by bortezomib. NF-jB activation leads to phosphorylation and ubiquitination of IjB.
`Once ubiquitinated, IjB is bound by the proteasome and degraded, releasing NF-jB. Free NFjB readily enters the nucleus where it
`activates the transcription of promoters containing jB response elements. Some of the genes activated by NF-jB encode anti-apoptotic
`factors, cell adhesion molecules, cytokines, and cell cycle regulators. Bortezomib prevents IjB degradation, maintaining NF-jB in an
`inactive complex with IjB and preventing the downstream effects of NF-jB activation. Consequently, cancer cells that require NF-jB for
`survival or to prevent apoptosis following cytotoxic treatments experience a downregulation in NF-jB-activated gene expression.
`
`ANACOR EX. 2011 - 2/7
`
`

`
`P R O T E A S O M E I N H I B I T I O N I N H E M A T O L O G I C M A L I G N A N C I E S
`
`35
`
`well to bortezomib or doxorubicin monotherapy in
`vitro. However, combined treatment led to a 4–5-fold
`increase in cell killing. Further studies from this lab
`show that bortezomib can interfere with DNA repair
`pathways by promoting the cleavage of DNA PKcs
`(DNA protein kinase catalytic subunit) and/or ATM
`(13). In addition, bortezomib could increase MDM2
`expression, thereby reducing the degradation of p53.
`Thus, while NF-jB may be a key target for bort-
`ezomib,
`inhibition of the proteasome affects nu-
`merous cellular pathways, thereby sensitizing the
`cell to apoptosis through several mechanisms.
`Proteasome inhibitors have also demonstrated
`activity against other B-cell malignancies in labora-
`tory studies, including mantle cell lymphoma (MCL)
`and Hodgkin’s disease. An in vitro study of MCL
`cells treated with bortezomib resulted in stabiliza-
`tion of IjB and reduced binding of activated NF-jB
`to its promoter. In addition, bortezomib treatment in
`vitro led to cell growth inhibition and rapid induc-
`tion of apoptosis of MCL cells. Bortezomib also
`exhibited activity in a xenograft model with MCL-
`xenografted, severe combined immunodeficiency
`mice evincing little or no gross MCL tumor in-
`volvement post-treatment compared to controls (14).
`Other preclinical studies in diffuse large B-cell
`lymphoma (DLBCL) provide indirect evidence that
`proteasome inhibition may be a potential treatment
`approach in this tumor type as well (15). Research
`has indicated that DLBCL is composed of two sub-
`groups – germinal center B-like (GCB) and activated
`B cell-like (ABC) – each of which present distinct
`pathogenetic mechanisms and clinical outcomes.
`Patients with ABC DLBLC have a poorer prognosis
`and ABC-type DLBCL is frequently refractory to
`chemotherapy. In vitro, ABC DLBCL cell lines had
`higher NF-jB activation, constitutive IjB kinase
`(IKK) activity, and IjBa degradation compared to
`GCB lines. Furthermore, ABC DLBCL cell
`lines
`treated with a ‘‘super-repressor’’ IjBa (unphosph-
`orylatable by IKK and therefore non-degradable)
`showed cell death and G1-phase growth arrest.
`These results suggest that proteasome inhibition, via
`its downregulation of the NF-jB pathway, is a po-
`tential mechanism of action in DLBCL treatment (16).
`While NF-jB is clearly important in bortezomib’s
`mechanism of action, several studies suggest that
`other mechanisms act in parallel to NF-B to induce
`apoptosis in MMCs and other cancer cells. In early
`experiments, cell culture experiments uncovered
`evidence that proteasome inhibition altered the ex-
`pression of proteins that control cell cycle and ap-
`optosis (4,17,18). More recently, Hideshima et al. (12)
`used a specific inhibitor of the NF-jB pathway to
`investigate non-NF-jB-mediated mechanisms of cell
`killing by proteasome inhibitors.
`In contrast
`to
`bortezomib, PS-1145 specifically prevents NF-jB
`
`activation by preventing the phosphorylation of IjB.
`Specific pathways activated by NF-j B were blocked
`by PS-1145: ICAM-1 and VCAM-1 expression were
`prevented in MMCs and IL-6 expression in BMSCs
`was eliminated. However, DNA synthesis in MMCs
`was incompletely blocked and PS-1145 was only
`capable of inhibiting cell growth in 20–50% of trea-
`ted MMCs (compared to nearly 100% of bortezomib-
`treated cells). Since a specific NF-jB-inhibitor is not
`as potent an inducer of apoptosis as bortezomib,
`other mechanisms tied to proteasome inhibition
`must be involved in bortezomib-induced apoptosis.
`
`PHASE I: TRIAL OF BORTEZOMIB IN
`PATIENTS WITH HEMATOLOGIC
`MALIGNANCIES
`
`Orlowski et al. conducted a phase I trial to determine
`the maximum-tolerated dose (MTD), dose-limiting
`toxicity (DLT), and phamacodynamics (PD) of bort-
`ezomib in patients with refractory hematologic ma-
`lignancies (19) (Table 1). Patients (N ¼ 27) were
`enrolled at 0.40 mg/m2 (n ¼ 3), 1.04 mg/m2 (n ¼ 12),
`1.20 mg/m2 (n ¼ 7), or 1.38 mg/m2 (n ¼ 5). Bortezo-
`mib was administered twice weekly for 4 weeks
`followed by a 2-week rest. Participants received a
`total of 293 doses of bortezomib, including 24 com-
`plete cycles.
`The MTD was determined to be 1.04 mg/m2. DLTs
`possibly related to bortezomib included thrombo-
`ctyopenia, malaise and fatigue, and electrolyte dis-
`turbances such as hyponatremia and hypokalemia.
`Ten patients developed grade 3 thrombocytopenia
`during cycle 1. Although the study protocol did not
`define thrombocytopenia as dose-limiting, it did in-
`fluence dosing behavior. In the majority of cases,
`grade 3 thrombocytopenia occurred in patients who
`entered the trial with thrombocytopenia. Only 1 of 9
`patients who entered the trial with normal platelet
`counts had dose withheld because of grade 3
`thrombocytopenia. No episodes of febrile neutrope-
`nia were reported. Nor were any major bleeding
`events reported during this trial. Five patients de-
`veloped a treatment-emergent peripheral neuropa-
`thy. Of the four grade 2 cases, three were thought to
`be related to bortezomib but were not dose-limiting;
`the sole grade 3 event was due to progressive disease.
`Adverse events were reported in patients at all
`dose levels with cytopenias as the most common
`events (Table 2); these included thrombocytopenia
`(74% of patients), anemia and leukopenia (48%), and
`neutropenia (37%). Gastrointestinal events were also
`seen in many patients including nausea (52%) and
`constipation (30%). Fatigue was reported in 59% of
`patients.
`
`ANACOR EX. 2011 - 3/7
`
`

`
`36
`
`
`
`TA B L EA B L E 1 Phase I patient demographics
`
`Characteristics
`
`Number of patients
`MSKCC
`UNC
`
`Sex
`Male
`Female
`
`Age (years)
`Mean
`Range
`
`Race
`Native American
`African American
`White
`
`Diagnoses
`Hodgkin’s disease
`Non-Hodgkin’s lymphoma
`SLL/CLL
`DLCL
`Mantle cell
`Plasma cell dycrasias
`Waldenstrom’s
`Multiple myeloma
`MDS with excess blasts and POEMS
`
`ECOG performance scale
`0
`1
`2
`
`Number
`
`Percent
`
`27
`7
`20
`
`17
`10
`
`56
`22–77
`
`1
`7
`19
`
`4
`10
`2
`2
`3
`12
`1
`11
`1
`
`6
`16
`5
`
`63
`37
`
`4
`26
`70
`
`22
`59
`19
`
`Prior therapy
`Chemotherapy
`Median no. regimens
`Range
`Radiation therapy
`Marrow or stem-cell transplantation
`
`27
`3
`1–12
`13
`10
`
`Reprinted with permission from: Orlowski RZ, Stinchcombe TE,
`Mitchell BS et al. Phase I trial of the proteasome inhibitor PS-341 in
`patients with refractory hematologic malignancies.
`J Clin Oncol
`2002; 20: 4420–4427.
` Memorial Sloan-Kettering Cancer Center.
` University of North Carolina.
`
`Twelve patients with heavily pre-treated plasma
`cell dyscrasias were enrolled in the study, nine of
`whom completed at least one full cycle and were
`assessable for response. One patient who received a
`total of 4 cycles showed by the end of cycle 3 an
`immunofixation-negative complete response. There
`was also evidence of antitumor activity among the
`eight remaining patients, with reduction of para-
`protein levels and/or marrow plasmacytosis.
`In
`addition, one patient with mantle cell lymphoma
`and another with refractory follicular lymphoma
`achieved a partial response with treatment at the
`1.38 mg/m2 level.
`
`P . R I C H A R D S O N
`
`Orlowski et al. concluded that bortezomib can be
`administered at 1.04 mg/m2, using a dose-intensive,
`twice-weekly administration for 4 weeks, followed a
`by a 2-week rest with concomitant monitoring for
`electrolyte abnormalities and late toxicities (19).
`However, induction of toxicity requiring the inter-
`ruption of therapy in patients during their first cycle
`of therapy most commonly occurred during the
`third week of treatment. Twice-weekly therapy for
`2-weeks with a third week off, delivering the same
`total dosage of over 6 weeks, was predicted to be
`better
`tolerated. Pharmacodynamic studies sug-
`gested that bortezomib induced 20S proteasome in-
`hibition in a time- and dose-dependent manner. This
`study provided the first evidence of potential activ-
`ity in myeloma and established the rationale for
`more comprehensive assessment in phase II (19).
`
`PHASE II: TRIALS OF BORTEZOMIB IN
`PATIENTS WITH MULTIPLE MYELOMA
`
`The safety and efficacy of bortezomib have been
`assessed in two phase II trials of patients with re-
`lapsed and refractory MM. Patients in Study 025
`(SUMMIT) were relapsed and refractory to their
`most recent therapy; those in Study 024 (CREST)
`were relapsed or refractory after front-line therapy.
`Patients in Study 025 thus had more advanced
`disease.
`This study enrolled patients in two cohorts: the
`first cohort filled rapidly ðN ¼ 78Þ, and, at the re-
`quest of the investigators, enrollment continued to
`eventually enroll a total of 202 patients. The first
`phase I trials (19,20) were designed to test several
`dosing schedules for bortezomib of varying dose
`intensities (Table 3). These studies were still in pro-
`gress at the initiation of Study 025 and preliminary
`indications from the study testing the 3-week cycle
`suggested this regimen offered a reasonable balance
`between toxicity and adequate dosing. At the time,
`dose-limiting diarrhea and peripheral neuropathy
`had been observed at 1.56 mg/m2 on the 3-week
`cycle. Thus, 1.3 mg/m2 bortezomib the previous
`dose level was administered by IV push on days 1, 4,
`8, and 11 of a 21 day treatment cycle in Study 025. A
`total of eight treatment cycles were allowed. Patients
`with a suboptimal response were allowed to add
`dexamethasone to bortezomib after 2 (patients with
`progressive disease) or 4 cycles (patients with stable
`disease).
`response criteria
`trial employed strict
`This
`[adapted from SWOG criteria and Blade et al. (21)]
`assessed by an independent review committee, and
`the population was heavily pretreated (the median
`number of prior treatments was 6) (22). For a com-
`
`ANACOR EX. 2011 - 4/7
`
`

`
`P R O T E A S O M E I N H I B I T I O N I N H E M A T O L O G I C M A L I G N A N C I E S
`
`37
`
`
`
`TA B L EA B L E 2 Bortezomib-related adverse events (Pgrade 3) during cycle 1
`
`Adverse event
`
`Bortezomib dose level (mg/m2)
`
`0.40
`(n ¼ 3)
`
`1.04
`(n ¼ 12)
`
`1.20
`(n ¼ 7)
`
`1.38
`(n ¼ 5)
`
`Total
`(N ¼ 27)
`
`P 1 adverse event
`Blood and lymphatic system
`Thrombocytopenia
`Anemia
`Neutropenia
`Leukopenia
`Metabolism and nutrition
`Hyponatremia
`Hypokalemia
`Other
`Malaise
`
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`7
`5
`4
`1
`2
`2
`4
`2
`1
`0
`0
`
`6
`6
`4
`3
`1
`1
`1
`0
`1
`1
`1
`
`4
`4
`2
`1
`1
`0
`2
`2
`0
`0
`0
`
`17
`15
`10
`5
`4
`3
`7
`4
`2
`1
`1
`
`Reprinted with permission from: Orlowski RZ, Stinchcombe TE, Mitchell BS et al. Phase I trial of the proteasome inhibitor PS-341 in
`patients with refractory hematologic malignancies. J Clin Oncol 2002; 20: 4420–4427.
`
`
`
`TA B L EA B L E 3 Dosing schedules in phase I bortezomib trials
`
`Treatment schedule
`1/wk 4
`2/wk 2
`2/wk 4
`
`Cycle length (days)
`
`Dose-limiting toxicities (mg/m2)
`
`Maximum tolerated dose (mg/m2)
`
`35
`21
`42
`
`NDa
`1.56b
`1.38b
`
`NDa (25)
`1.3 (20)
`1.04 (19)
`
`Reprinted with permission from Adams J. Potential for proteasome inhibition in the treatment of cancer. Drug Discovery Today, 2003; in
`press.
`a Not determined. MTD not reached at time of report.
`b Enrollment suspended due to DLT; maximum dose reached.
`
`plete response, no M protein was visible by immu-
`nofixation, fewer than 5% plasma cells were present
`in the bone marrow, and no new bone disease or
`plasmacytomas were present; responses were con-
`firmed after 6 weeks. Preliminary data from the first
`cohort showed significant biologic activity with
`manageable toxicities, as demonstrated by reduc-
`tions in M protein (23). A preliminary analysis of the
`full cohort ðn ¼ 202Þ confirming and extending these
`results was recently presented and is being prepared
`for publication (22). An integral component of Study
`025 was the collection of bone marrow samples for
`pharmacogenomic analysis. Prior to treatment, bone
`marrow samples were collected from 126 consenting
`patients, and RNA from myeloma cells was purified
`from 64 of these. The gene expression profiles of
`responding patients are now being compared to the
`expression profiles of non-responders to identify
`expression patterns that are predictive of a response
`to bortezomib.
`Study 024 allowed the assessment of a patient
`population with earlier stage disease (median prior
`therapies was 3) and applied the same treatment
`schedule and response criteria as in Study 025. A
`total of 54 patients were randomized to receive ei-
`
`ther 1.0 (28 patients) or 1.3 (26 patients) mg/m2
`bortezomib. Dexamethasone was also allowed for
`patients who had progressive (after cycle 2) or stable
`disease (after cycle 4). Preliminary results of this trial
`were recently reported and the analysis is ongoing
`(24).
`
`FUTURE DIRECTIONS
`
`A phase III study comparing response to bortezomib
`versus high-dose dexamethasone is in progress. The
`multicenter Assessment of Proteasome Inhibition for
`Extending Remissions (APEX)
`trial will be con-
`ducted at more than 60 centers in the United States,
`Canada, and Europe. Over 600 patients with re-
`lapsed or refractory MM will be enrolled and ran-
`domize to either bortezomib or dexamethasone
`treatment arms. Response will be assessed by pro-
`longation of
`time-to-disease progression and se-
`lected measures of clinical benefit. APEX will also
`study the potential of bortezomib as maintenance
`therapy. Survival, response rates, quality of life, and
`safety and tolerability in the two treatment arms will
`
`ANACOR EX. 2011 - 5/7
`
`

`
`38
`
`P . R I C H A R D S O N
`
`Figure 2 Treatment regimens in the phase III APEX trial. Bortezomib is being compared to high dose dexamethasone treatment in
`an international phase III study. Both arms have a treatment phase followed by a maintenance phase.
`
`also be compared. In addition, bone marrow sam-
`ples are being collected for validation of candidate
`drug response markers identified in Study 024 and
`Study 025.
`In the APEX trial, bortezomib 1.3 mg/m2 will be
`administered as injection for eight 3-week induction
`cycles followed by three 5-week maintenance cycles
`(Figure 2). The 3-week cycles include twice-weekly
`administration (Days 1, 4, 8, and 11) for 2 weeks
`followed by a 10-day rest period (Days 12–21); the
`5-week cycles include once-weekly administration
`(Days 1, 8, 15, and 22) for 4 weeks followed by a
`13-day rest period (Days 23–35). In the comparator
`arm, patients will receive oral administration of
`dexamethasone 40 mg/day for four 5-week cycles
`followed by three 4-week cycles. Dosing will be
`administered in the 5-week cycles on Days 1–4, 9–12,
`and 17–20 followed by a 15-day rest period. The
`4-week cycles include doses on Days 1–4 followed
`by a 24-day rest period.
`Depending on response, patients in either treat-
`ment arm may receive up to 10 months of treatment.
`In addition, patients in the dexamethasone cohort
`who experience progressive disease may be eligible
`for participation in a bortezomib extension trial
`(Study 040). The objective of Study 040 is to deter-
`mine rate and duration of response in patients with
`relapsed or refractory MM.
`Additional clinical trials for hematologic malig-
`nancies are currently under consideration. These
`trials will assess bortezomib’s safety and efficacy
`when used as a single agent or in combination with
`other chemotherapeutics. There are currently ten
`additional clinical trials either open or in develop-
`ment to further explore the role of bortezomib in
`myeloma. In addition, there are ten early clinical
`trials open or in development that will evaluate
`bortezomib therapy for lymphomas (non-Hodgkins’
`lymphoma and mantle cell lymphoma) and eight
`clinical
`trials open or in development for other
`hematologic malignancies
`(myelodysplastic syn-
`drome, Waldenstrom’s macroglobulinaemia, acute
`myelogenous leukemia, and chronic myelogenous
`leukemia).
`
`CONCLUSIONS
`
`The NF-jB signaling pathway appears to be a
`promising target for the treatment of hematologic
`malignancies. Activation of NF-jB is an important
`step in transformation and can provide protection
`from cytotoxic treatments, while inhibition of this
`pathway with proteasome inhibitors results in ap-
`optosis or sensitizes cancer cells to cytotoxic drugs.
`Phase I trials demonstrated responses in several
`hematologic malignancies,
`including MM; other
`phase I trials showed that this agent was relatively
`well tolerated. Several phase II studies have further
`examined the safety and activity of bortezomib in
`MM, and the findings in these trials have been en-
`couraging. Bortezomib appears to have antitumor
`activity in MM and the adverse effects associated
`with treatment are manageable. A phase III trial is
`now in progress comparing the safety and efficacy of
`bortezomib treatment to high dose dexamethasone
`in patients with advanced MM.
`
`REFERENCES
`
`1. Shah SA, Potter MW, McDade TP, et al. 26S proteasome
`inhibition induces apoptosis and limits growth of human
`pancreatic cancer. J Cell Biochem 2001; 82(1): 110–122.
`2. Bold RJ, Virudachalam S, McConkey DJ. Chemosensitization
`of pancreatic cancer by inhibition of the 26S proteasome. J
`Surg Res 2001; 100(1): 11–17.
`al. Enhanced
`et
`3. Cusack Jr
`JC, Liu R, Houston M,
`chemosensitivity to CPT-11 with proteasome inhibitor PS-
`341: Implications for systemic nuclear factor-jB inhibition.
`Cancer Res 2001; 61(9): 3535–3540.
`4. Hideshima T, Richardson P, Chauhan D, et al. The proteasome
`inhibitor PS-341 inhibits growth,
`induces apoptosis, and
`overcomes drug resistance in human multiple myeloma
`cells. Cancer Res 2001; 61(7): 3071–3076.
`5. Ma MH, Parker KM, Manyak S, et al. Proteasome inhibitor PS-
`341 markedly enhances sensitivity of multiple myeloma cells
`to chemotherapeutic agents and overcomes chemoresistance
`through inhibition of the NF-jB pathway. Blood 2001; 98(11):
`437a.
`6. Russo SM, Tepper JE, Baldwin AS, et al. Enhancement of
`radiosensitivity by proteasome inhibition: Implications for a
`
`ANACOR EX. 2011 - 6/7
`
`

`
`P R O T E A S O M E I N H I B I T I O N I N H E M A T O L O G I C M A L I G N A N C I E S
`
`39
`
`Int
`
`J Radiat Oncol Biol Phys 2001; 50(1):
`
`role of NF-jB.
`183–193.
`7. Berenson JR, Ma HM, Vescio R. The role of nuclear factor-jB
`in the biology and treatment of multiple myeloma. Semin
`Oncol 2001; 28(6): 626–633.
`8. LeBlanc R, Catley LP, Hideshima T, et al. Proteasome inhibitor
`PS-341 inhibits human myeloma cell growth in vivo and
`prolongs survival in a murine model. Cancer Res 2002; 62(17):
`4996–5000.
`9. Damiano JS, Cress AE, Hazlehurst LA, Shtil AA, Dalton WS.
`Cell adhesion mediated drug resistance (CAM-DR): role of
`integrins and resistance to apoptosis in human myeloma cell
`lines. Blood 1999; 93(5): 1658–1667.
`10. Chauhan D, Uchiyama H, Akbarali Y, et al. Multiple myeloma
`cell adhesion-induced interleukin-6 expression in bone
`marrow stromal cells involves activation of NF-jB. Blood
`1996; 87(3): 1104–1112.
`11. Read MA, Neish AS, Luscinskas FW, Palombella VJ, Maniatis
`T, Collins T. The proteasome pathway is required for
`cytokine-induced endothelial–leukocyte adhesion molecule
`expression. Immunity 1995; 2(5): 493–506.
`12. Hideshima T, Mitsiades C, Akiyama M, et al. Molecular
`mechanisms mediating anti-myeloma activity of proteasome
`inhibitor PS-341. Blood 2003; 101(4): 1530–1534.
`et al. The
`13. Mitsiades N, Mitsiades CS, Richardson PG,
`proteasome
`inhibitor PS-341 potentiates
`sensitivity of
`multiple myeloma cells to conventional chemotherapeutic
`agents: therapeutic applications. Blood 2002; published online.
`14. Pham L, Tamayo A, Lo P, Yoshimura L, Ford RJ. Antitumor
`activity of the proteasome inhibitor PS-341 in mantle cell
`lymphoma B cells. Blood 2001; 98(11): 465a.
`15. Schenkein D. Proteasome inhibitors in the treatment of B-cell
`malignancies. Clin Lymphoma 2002; 3(1): 49–55.
`16. Davis RE, Brown KD, Siebenlist U, Staudt LM. Constitutive
`nuclear factor jB activity is required for survival of activated
`B cell-like diffuse large B cell lymphoma cells. J Exp Med 2001;
`194(12): 1861–1874.
`
`17. Desai SD, Li TK, Rodriguez-Bauman A, Rubin EH, Liu LF.
`Ubiquitin/26S proteasome-mediated degradation of topoisom-
`erase I as a resistance mechanism to camptothecin in tumor
`cells. Cancer Res 2001; 61(15): 5926–5932.
`18. Adams J, Palombella VJ, Sausville EA, et al. Proteasome
`inhibitors: a novel class of potent and effective antitumor
`agents. Cancer Res 1999; 59(11): 2615–2622.
`19. Orlowski RZ, Stinchcombe TE, Mitchell BS, et al. Phase I trial of
`the proteasome inhibitor PS-341 in patients with refractory
`hematologic malignancies. J Clin Oncol 2002; 20(21): 4420–4427.
`20. Aghajanian C, Soignet S, Dizon DS, et al. A phase I trial of the
`novel proteasome inhibitor PS341 in advanced solid tumor
`malignancies. Clin Cancer Res 2002; 8(8): 2505–2511.
`21. Blade J, Samson D, Reece D, et al. Criteria for evaluating
`disease response and progression in patients with multiple
`myeloma treated by high-dose therapy and haemopoietic
`stem cell
`transplantation. Myeloma Subcommittee of
`the
`EBMT. European Group for Blood and Marrow Transplant.
`Br J Haematol 1998; 102(5): 1115–1123.
`22. Richardson P, Barlogie B, Berenson J, et al. A phase II
`multicenter study of the proteasome inhibitor bortezomib
`(Velcadee formerly PS-341) in multiple myeloma patients
`(pts) with relapsed/refractory disease. Blood 2002; 100(11): 104a.
`23. Richardson PG, Barlogie B, Berenson J, et al. Phase II study of
`the proteasome inhibitor PS-341 in multiple myeloma patients
`(pts) with relapsed/refractory disease: results from cohort 1
`analysis. Proceedings of the American Society of Clinical Oncology-
`38th Annual Meeting 2002; 21: 11a.
`24. Jagannath S, Barlogie B, Berenson J, et al. A phase II
`multicenter randomized study of the proteasome inhibitor
`bortezomib (VELCADEe,
`formerly PS-341)
`in multiple
`myeloma (MM) patients
`(pts)
`relapsed after
`front-line
`therapy. Blood 2002; 100(11): 812a.
`25. Papandreou C, Daliani D, Millikan RE, et al. Phase I study of
`intravenous (I.V.) proteasome inhibitor PS-341 in patients (Pts)
`the American
`with advanced malignancies. Proceedings of
`Society of Clinical Oncology-37th Annual Meeting 2001; 20: 86a.
`
`ANACOR EX. 2011 - 7/7

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