As filed with
the Securities and Exchange Commission on October 22,
2010
Registration
No. 333-
UNITED STATES SECURITIES AND
EXCHANGE COMMISSION
Washington, D.C.
20549
Form S-1
REGISTRATION
STATEMENT
UNDER
THE SECURITIES ACT OF
1933
Anthera Pharmaceuticals,
Inc.
(Exact name of registrant as
specified in its charter)
|
|
|
|
|
Delaware
|
|
2834
|
|
20-1852016
|
(State or other jurisdiction
of
incorporation or organization)
|
|
(Primary Standard Industrial
Classification Code Number)
|
|
(I.R.S. Employer
Identification No.)
|
Anthera Pharmaceuticals,
Inc.
25801 Industrial Boulevard,
Suite B
Hayward, California
94545
(510) 856-5600
(Address, including zip code and
telephone number, including area code, of registrants
principal executive offices)
Paul F. Truex
President and Chief Executive
Officer
25801 Industrial Boulevard,
Suite B
Hayward, California
94545
(510) 856-5600
(Name, address, including zip
code and telephone number, including area code, of agent for
service)
Copies to:
Bradley A.
Bugdanowitz, Esq.
Mitzi Chang, Esq.
Goodwin Procter LLP
Three Embarcadero Center, 24th
Floor
San Francisco, California
94111-4003
(415) 733-6000
Approximate date of commencement of proposed sale to
public: From time to time after the effective
date of this registration statement.
If any of the securities being registered on this Form are to
be offered on a delayed or continuous basis pursuant to
Rule 415 under the Securities Act of 1933, check the
following
box. o
If this Form is filed to register additional securities for
an offering pursuant to Rule 462(b) under the Securities Act,
check the following box and list the Securities Act registration
statement number of the earlier effective registration statement
for the same
offering. o
If this Form is a post-effective amendment filed pursuant to
Rule 462(c) under the Securities Act, check the following
box and list the Securities Act registration statement number of
the earlier effective registration statement for the same
offering. o
If this Form is a post-effective amendment filed pursuant to
Rule 462(d) under the Securities Act, check the following
box and list the Securities Act registration statement number of
the earlier effective registration statement for the same
offering. o
Indicate by check mark whether the registrant is a large
accelerated filer, an accelerated filer, a non-accelerated
filer, or a smaller reporting company. See the definitions of
large accelerated filer, accelerated
filer and smaller reporting company in Rule
12b-2 of the
Exchange Act. (Check one):
|
|
|
|
Large
accelerated
filer o |
Accelerated
filer o |
Non-accelerated
filer þ |
Smaller reporting
company o |
(Do not check if a smaller
reporting company)
CALCULATION
OF REGISTRATION FEE
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Proposed Maximum
|
|
|
Proposed Maximum
|
|
|
|
Title of Each Class of
|
|
|
Amount to be
|
|
|
Offering Price Per
|
|
|
Aggregate Offering
|
|
|
Amount of
|
Securities to be Registered
|
|
|
Registered(1)
|
|
|
Unit(2)
|
|
|
Price(2)
|
|
|
Registration Fee
|
Common Stock, par value $0.001 per share
|
|
|
20,380,549
|
|
|
$6.26
|
|
|
$127,582,237
|
|
|
$9,097
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(1)
|
Represents shares of common stock to be sold by the selling
stockholders named in this registration statement and includes
an aggregate of 4,200,000 shares of common stock that may
be issued upon the exercise of warrants held by the selling
stockholders described in this registration statement. Pursuant
to Rule 416 of the Securities Act of 1933, as amended (the
Securities Act), this registration statement also
covers such an indeterminate amount of shares of common stock as
may become issuable to prevent dilution resulting from stock
splits, stock dividends and similar events.
|
(2)
|
Estimated solely for the purpose of calculating the
registration fee, based on the average of the high and low
prices for the registrants common stock on
October 20, 2010, pursuant to Rule 457(c) under the
Securities Act.
|
The Registrant hereby amends this Registration Statement on
such date or dates as may be necessary to delay its effective
date until the registrant shall file a further amendment which
specifically states that this Registration Statement shall
thereafter become effective in accordance with Section 8(a)
of the Securities Act of 1933, as amended, or until the
Registration Statement shall become effective on such date as
the Securities and Exchange Commission, acting pursuant to said
Section 8(a), may determine.
The information in
this prospectus is not complete and may be changed. The selling
stockholders may not sell these securities until the
registration statement filed with the Securities and Exchange
Commission is effective. This prospectus is not an offer to sell
these securities and it is not soliciting offers to buy these
securities in any state where the offer or sale is not
permitted.
|
Subject to completion, dated
October 22, 2010
PROSPECTUS
20,380,549 Shares of
Common Stock
This prospectus covers the sale of an aggregate of
20,380,549 shares of our common stock, $0.001 par
value per share, by the selling stockholders identified in this
prospectus, including their transferees, pledgees, donees or
successors. The common stock covered by this prospectus consists
of (i) 10,500,000 shares of common stock and
(ii) 4,200,000 shares of common stock issuable upon
exercise of outstanding warrants (the Warrants)
issued in a private placement transaction that closed on
September 24, 2010, and (iii) 5,517,349 shares of
common stock and 163,200 shares of common stock issuable
upon outstanding warrants held by existing stockholders.
The selling stockholders may sell their shares of common stock
from time to time at market prices prevailing at the time of
sale, at prices related to the prevailing market price, or at
negotiated prices. We will not receive any proceeds from the
sale of common stock by the selling stockholders, other than as
a result of the exercise of Warrants held by the selling
stockholders for cash.
No underwriter or other person has been engaged to facilitate
the sale of shares of our common stock in this offering. We are
paying the cost of registering the shares of common stock
covered by this prospectus as well as various related expenses.
The selling stockholders are responsible for all selling
commissions, transfer taxes and other costs related to the offer
and sale of their shares of common stock.
Our common stock is traded on the NASDAQ Global Market under the
symbol ANTH. On October 21, 2010, the closing
sale price of our common stock on the NASDAQ Global Market was
$6.68 per share.
This investment involves risks. See Risk Factors
beginning on page 11.
Neither the Securities and Exchange Commission nor any state
securities commission has approved or disapproved of these
securities or determined if this prospectus is truthful or
complete. Any representation to the contrary is a criminal
offense.
The date of this prospectus
is ,
2010.
TABLE OF
CONTENTS
|
|
|
|
|
|
|
Page
|
|
|
|
|
1
|
|
|
|
|
11
|
|
|
|
|
35
|
|
|
|
|
37
|
|
|
|
|
38
|
|
|
|
|
39
|
|
|
|
|
40
|
|
|
|
|
41
|
|
|
|
|
43
|
|
|
|
|
46
|
|
|
|
|
49
|
|
|
|
|
65
|
|
|
|
|
109
|
|
|
|
|
118
|
|
|
|
|
138
|
|
|
|
|
146
|
|
|
|
|
150
|
|
|
|
|
156
|
|
|
|
|
158
|
|
|
|
|
162
|
|
|
|
|
162
|
|
|
|
|
162
|
|
|
|
|
F-1
|
|
EX-5.1 |
EX-10.40 |
EX-23.1 |
You should rely only on the information contained in this
prospectus, any applicable prospectus supplement and the
information incorporated by reference in this prospectus. We
have not authorized anyone to provide you with additional or
different information. This document may only be used where it
is legal to sell these securities. The information in this
prospectus is accurate only as of the date of this prospectus,
regardless of the time of delivery of this prospectus or any
sale of shares of our common stock.
PROSPECTUS
SUMMARY
This summary highlights certain information contained elsewhere
in this prospectus. Because this is only a summary, it does not
contain all of the information you should consider before
investing in our common stock. You should read this entire
prospectus carefully, especially the information set forth under
the headings Risk Factors and
Managements Discussion and Analysis of Financial
Condition and Results of Operations and our financial
statements and the related notes appearing at the end of this
prospectus, before making an investment decision.
Our
Company
We are a biopharmaceutical company focused on developing and
commercializing products to treat serious diseases associated
with inflammation, including cardiovascular and autoimmune
diseases. We currently have one Phase 3 clinical program,
varespladib, and two Phase 2 clinical programs,
A-623 and
A-001.
Varespladib and
A-001
inhibit a novel enzyme target known as secretory phospholipase
A2,
or
sPLA2.
Elevated levels of
sPLA2
have been implicated in a variety of acute inflammatory
conditions, including acute coronary syndrome and acute chest
syndrome, as well as chronic diseases such as stable coronary
artery disease, or CAD. Our Phase 2 product candidate,
A-623,
targets elevated levels of
B-lymphocyte
stimulator, or BLyS, also known as B-Cell Activating Factor, or
BAFF, which has been associated with a variety of B-cell
mediated autoimmune diseases, including systemic lupus
erythematosus, or lupus. We have worldwide rights to our product
candidates, with the exception of Japan, where
Shionogi & Co., Ltd. retains commercial rights to our
sPLA2
product candidates.
Product
Development Programs
We have focused our product development programs on
anti-inflammatory therapeutics for cardiovascular diseases,
lupus and other serious diseases for which we believe current
treatments are either inadequate or non-existent. Our current
product development programs are listed in the figure below.
Varespladib
for the Treatment of Acute Coronary Syndrome
We have commenced a pivotal Phase 3 clinical study named
VISTA-16 (Vascular Inflammation Suppression to Treat Acute
coronary syndrome 16 Weeks) for our lead product
candidate, varespladib, an oral
sPLA2
inhibitor, in combination with Lipitor (atorvastatin), a HMG-CoA
reductase inhibitor, for short-term (16-week) treatment of
patients experiencing an acute coronary syndrome. The American
Heart Association defines acute coronary syndrome as any group
of clinical signs and
1
symptoms related to acute myocardial ischemia, or heart muscle
damage. Patients experiencing an acute coronary syndrome suffer
from significant inflammation and abnormal lipid profiles, which
may lead to further vascular damage and a second cardiovascular
event.
sPLA2
enzymes act to directly amplify inflammation, and adversely
modify lipids. Varespladib, when combined with lipid-lowering
therapies, is one of only a few therapeutics in development with
the potential to offer a unique and synergistic approach
targeting inflammation, elevated lipid levels and
atherosclerosis.
Clinical results from FRANCIS (Fewer Recurrent Acute coronary
events with Near-term Cardiovascular Inflammation Suppression),
our Phase 2b clinical study enrolling 625 acute coronary
syndrome patients, and two Phase 2 clinical studies enrolling
534 stable CAD patients demonstrated statistically significant
reductions in low-density lipoprotein cholesterol, or LDL-C, a
known predictor of cardiovascular risk. Reductions in LDL-C were
greater when used in combination with commonly prescribed statin
therapies. In addition, rapid and sustained anti-inflammatory
activity was also evident as
sPLA2
concentrations were statistically significantly reduced from
baseline levels throughout dosing in all clinical studies. In
our Phase 2b clinical study, C-reactive protein, or CRP, and
interleukin-6, or IL-6, both independent predictors of
cardiovascular risk, were lower at all time points among
varespladib treated patients as compared to placebo. The percent
decrease in CRP at week two in our Phase 2b clinical study was
nearly two-fold greater among varespladib treated patients than
those treated with placebo (p = 0.183) and by week 16, the
difference between the two groups achieved statistical
significance (p = 0.0067). A p-value is a probability with a
value ranging from 0 to 1, which indicates the likelihood that a
clinical study is different between treatment and control
groups. P-values below 0.05 are typically referred to as
statistically significant.
The VISTA-16 acute coronary syndrome study is a multinational,
randomized, double-blind, placebo-controlled Phase 3 clinical
study designed to evaluate short-term (16-week) therapy with
varespladib in combination with Lipitor (atorvastatin) for the
prevention of secondary major adverse coronary events in
patients who have recently experienced an acute coronary
syndrome. As part of our Special Protocol Assessment, or SPA,
agreement with the U.S. Food and Drug Administration, or
FDA, the VISTA-16 study is estimated to enroll up to
6,500 patients with similar characteristics to patients in
FRANCIS. Patients are randomized within 96 hours of an
acute coronary syndrome and will receive 16 weeks of either
once-daily varespladib or placebo in addition to a dose of
Lipitor (atorvastatin). VISTA-16 will continue enrollment until
a minimum of 385 primary endpoint events have occurred. The
primary endpoint of the VISTA-16 study will assess the time to
the first occurrence of the combined endpoint of cardiovascular
death, non-fatal myocardial infarction, non-fatal stroke or
documented unstable angina with objective evidence of ischemia,
which is lack of blood to tissues due to a blockage of a vessel,
requiring hospitalization. Survival status will be obtained for
all patients six months after the completion of dosing. Based
upon our statistical calculations, this number of primary
endpoint events will allow us to detect a treatment effect on
the composite endpoint as low as 18.1% with a p-value of less
than 0.05. As in the FRANCIS study, changes in
sPLA2,
CRP and LDL-C will be measured at baseline, 24 hours, and
at weeks one, two, four, eight and 16. An independent committee
comprised of individuals who are not involved with the VISTA-16
clinical study will conduct a data review of these biomarkers
after at least 1,000 patients have been enrolled in the
clinical study. This biomarker futility analysis is designed to
confirm that relevant biomarkers have met pre-specified
statistical reductions versus placebo at various time-points.
Elevations of each of these biomarkers,
sPLA2,
CRP, LDL-C, and IL-6 are known to be independently correlated
with adverse cardiovascular outcomes. At the same time, our
independent Data Safety Monitoring Board, or DSMB, will review
all available clinical data from VISTA-16 to assess overall
safety.
2
Our
Special Protocol Assessment Agreement with the FDA
We have reached agreement with the FDA on an SPA, for the
VISTA-16 acute coronary syndrome study protocol, including
patient inclusion/exclusion criteria, study size, statistical
considerations, efficacy endpoints, study duration,
randomization and lipid management strategies.
A-623
Our BAFF Antagonism Program for the Treatment of Lupus
BAFF has been associated with a wide range of B-cell mediated
autoimmune diseases, including systemic lupus erythematosus, or
lupus. The beneficial role of BAFF inhibition to improve
clinical outcomes in patients with lupus has been evaluated in
multiple phase 3 studies with another BAFF antagonist. We intend
to advance the development of
A-623, a
BAFF inhibitor, in a number of autoimmune diseases including
lupus and rheumatoid arthritis. Our peptibody is a novel fusion
protein that is distinct from an antibody, binds to both soluble
and membrane-bound BAFF and is manufactured using bacterial
fermentation. Recent clinical and non-clinical studies have
reported that membrane-bound BAFF is a more potent stimuli for
B-cell maturation and survival than the soluble form of BAFF. In
fact, lupus disease severity can be correlated with higher
levels of membrane-bound BAFF. We are actively evaluating a
partnership opportunity with major pharmaceutical companies to
develop and commercialize
A-623. We
licensed
A-623 from
Amgen Inc. in December 2007 and have worldwide product rights in
all indications.
Two randomized, dose-ranging, placebo-controlled Phase 1
clinical studies evaluating
A-623 in
104 patients have been completed. Results from these
studies demonstrated antagonism of BAFF by
A-623 led to
statistically significant reductions in B-cells of approximately
50-70% (p
< 0.001) among lupus patients across multiple subcutaneous
and intravenous formulations. We believe
A-623 could
offer
3
a number of advantages over other BAFF (or BLyS) antagonists as
well as other novel B-cell directed therapies including:
|
|
|
|
|
convenient, at-home, patient-administered subcutaneous dosing;
|
|
|
|
a range of dosing frequencies including monthly and weekly;
|
|
|
|
|
|
binding to both membrane-bound and soluble forms of BAFF;
|
|
|
|
|
|
low cost of goods based on a bacterial fermentation
manufacturing process; and
|
|
|
|
|
|
multiple binding domains achieve highest reported affinity for
inhibition of BAFF.
|
Based on results from 104 patients in our Phase 1a and 1b
clinical studies, we have commenced patient dosing in our Phase
2b clinical study with
A-623 in
lupus patients. PEARL-SC (A Randomized, Double-Blind Phase 2b
Study to Evaluate the Efficacy, Safety, and Tolerability of
A-623
AdministRation in Subjects with Systemic Lupus Erythematosus) is
a randomized, placebo-controlled, phase 2b clinical study that
allows enrollment of up to 600 patients in 60 centers
worldwide. Subjects will be randomized into three active
subcutaneous treatment arms and one subcutaneous placebo
treatment arm for a minimum of 24 weeks and a maximum of
one year. The primary endpoint of the PEARL-SC study will be
clinical improvement at 24 weeks in responder rates of a
systemic lupus erythematosus responder index, or SRI, in the
pooled treatment arms versus placebo. The primary endpoint is
based upon changes in SELENA and SLEDAI disease activity scale,
Physicians Global Assessment scores and British Isles
Lupus Assessment Group scores, which are clinical standards for
the measurement of disease severity in lupus patients. Secondary
endpoints will include safety, improvement in other clinical
assessment scores, clinical response in patients with various
baseline disease severities, resolution of fatigue, steroid
utilization and time to flare. A blinded interim biomarker
analysis to establish the appropriate drug effect on B-Cells is
included early in the study.
4
The following table represents our current PEARL-SC study as
well as the option to extend the study to collect long-term
safety data.
A-001
for the Prevention of Acute Chest Syndrome Associated with
Sickle Cell Disease
Our next product candidate, varespladib sodium,
A-001, is an
intravenously administered inhibitor of
sPLA2,
which is in a Phase 2 clinical study for the prevention of acute
chest syndrome associated with sickle cell disease. Acute chest
syndrome is a form of inflammation-induced lung failure and is
the most common cause of death in patients with sickle cell
disease.
sPLA2
levels increase substantially in the 24 to 48 hours before
the onset of acute chest syndrome. According to the Sickle Cell
Information Center, sickle cell disease is a genetic disorder
afflicting more than 70,000 people in the United States
alone. Given the small patient population and lack of approved
drugs for the prevention of acute chest syndrome, we have
received orphan drug designation and fast track status from the
FDA for
A-001.
A pre-specified interim review of our Phase 2 clinical study
results by a DSMB indicate
A-001, at a
certain dose, reduced
sPLA2
activity by more than 80% from baseline within 48 hours.
Furthermore, the incidence of acute chest syndrome appeared to
be related to the level of
sPLA2
activity.
Other
sPLA2
Inhibitors
We also have an additional novel
sPLA2
inhibitor,
A-003, in
preclinical development for existing target indications as well
as other therapeutic areas.
A-003 has
shown increased potency against
sPLA2
and favorable characteristics in preclinical studies. We plan to
file an investigational new drug application for
A-003 in the
future.
5
Our
Strategy
Our objective is to develop and commercialize our product
candidates to treat serious diseases associated with
inflammation, including cardiovascular and autoimmune diseases.
To achieve these objectives, we intend to initially focus on:
|
|
|
|
|
advancing the development of varespladib through the Phase 3
VISTA-16 clinical study;
|
|
|
|
advancing the development of
A-623
through the Phase 2b PEARL-SC clinical study;
|
|
|
|
leveraging our
sPLA2
expertise to develop products for additional disease
indications; and
|
|
|
|
developing commercial strategies designed to maximize our
product candidates market potential, including securing
corporate partners whose capabilities complement ours.
|
Risks
Related to Our Business
The risks set forth under the section entitled Risk
Factors beginning on page 11 of this prospectus
reflect risks and uncertainties that could significantly and
adversely affect our business and our ability to execute our
business strategy. For example:
|
|
|
|
|
We are a development-stage company with no revenue and no
products approved for marketing. We will need substantial
additional capital to fund our operations and develop our
product candidates.
|
|
|
|
For the six months ended June 30, 2010, we had net losses
of approximately $19.0 million, and as of June 30,
2010, we had an accumulated deficit of approximately
$84.3 million. We expect to incur continued significant
losses for the foreseeable future.
|
|
|
|
We are largely dependent on the success of our development-stage
product candidates, particularly our primary product candidates,
varespladib,
A-623 and
A-001, and
our clinical studies may fail to adequately demonstrate their
safety and efficacy. If a clinical study fails, or if additional
clinical studies are required, our development costs may
increase and we may be unable to continue operations without
raising additional funding.
|
|
|
|
The regulatory approval process is expensive, time-consuming and
uncertain, and our product candidates have not been, and may not
be, approved for sale by regulatory authorities or be
successfully commercialized. Even if approved for sale by the
appropriate regulatory authorities, our products may not achieve
market acceptance and we may never achieve profitability.
|
|
|
|
Our preclinical development programs may not produce any other
viable or marketable product candidates.
|
|
|
|
Our and our licensors patent positions may not adequately
protect our present or future product candidates or permit us to
gain or keep a competitive advantage.
|
Commercialization
Strategy
We have worldwide rights to develop and commercialize our
products in all indications and markets, with the exception of
Japan, where Shionogi & Co., Ltd. retains commercial
rights to our
sPLA2
product candidates. Our current development plans are focused on
acute treatment and orphan
6
indications that may provide an accelerated and cost-efficient
path to regulatory approval and commercialization. We believe
that certain of these markets can be commercialized through a
limited specialty sales force. In addition, we believe that our
product candidates can also address market opportunities in
chronic indications and we may seek development and
commercialization partners to address these non-specialty and
international markets.
Company
Information
We were incorporated in Delaware on September 9, 2004 as
Anthera Pharmaceuticals, Inc. Our corporate headquarters are
located at 25801 Industrial Boulevard, Suite B, Hayward,
California 94545 and our telephone number is
(510) 856-5600.
Our website address is www.anthera.com. The information
contained on our website or that can be accessed through our
website is not incorporated by reference into this prospectus
and is not part of this prospectus.
We use various trademarks, service marks and trade names in our
business, including without limitation Anthera
Pharmaceuticals and Anthera. This prospectus
also contains trademarks, services marks and trade names of
other businesses that are the property of their respective
holders.
Unless the context otherwise requires, we use the terms
Anthera Pharmaceuticals, Anthera,
we, us, the Company and
our in this prospectus to refer to Anthera
Pharmaceuticals, Inc. and its sole subsidiary.
7
THE
OFFERING
This prospectus relates to the resale by the selling
stockholders identified in this prospectus of up to
20,380,549 shares of common stock, of which
16,017,349 shares are issued and outstanding as of the date
of this prospectus, and 4,363,200 shares of which are
issuable upon the exercise of certain warrants.
10,500,000 shares of common stock issued and outstanding as
of the date of this prospectus and Warrants exercisable for
4,200,000 shares of common stock were issued in a private
placement transaction that closed on September 24, 2010.
5,517,349 shares of common stock and 163,200 shares of
common stock issuable upon outstanding warrants are held by
existing stockholders. All of the shares, when sold, will be
sold by the selling stockholders. The selling stockholders may
sell their shares from time to time at market prices prevailing
at the time of sale, at prices related to the prevailing market
price, or at negotiated prices. We will not receive any proceeds
from the sale of shares by the selling stockholders, other than
as a result of the exercise of Warrants held by the selling
stockholders for cash.
8
SUMMARY
FINANCIAL DATA
The following summary financial data should be read together
with our financial statements and the related notes and
Managements Discussion and Analysis of Financial
Condition and Results of Operations appearing elsewhere in
this prospectus. The summary financial data in this section is
not intended to replace our financial statements and the related
notes. Our historical results are not necessarily indicative of
the results to be expected for any future period.
We were incorporated on September 9, 2004. The following
statement of operations data, including share data, for the
years ended December 31, 2007, 2008 and 2009 have been
derived from our audited financial statements and related notes
appearing elsewhere in this prospectus. The statement of
operations data, including share data, for the six months ended
June 30, 2009 and 2010 and the balance sheet data as of
June 30, 2010 have been derived from our unaudited interim
financial statements appearing elsewhere in this prospectus. The
unaudited interim financial statements have been prepared on the
same basis as the audited financial statements and reflect all
adjustments necessary to fairly state our financial position as
of June 30, 2010 and results of operations for the six
months ended June 30, 2009 and 2010. The operating results
for any period are not necessarily indicative of financial
results that may be expected for any future period.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Fiscal Year Ended December 31,
|
|
|
Six Months Ended June 30,
|
|
|
|
2007
|
|
|
2008
|
|
|
2009
|
|
|
2009
|
|
|
2010
|
|
|
Statement of Operations Data:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Operating expenses
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Research and development
|
|
$
|
23,921,932
|
|
|
$
|
10,882,322
|
|
|
$
|
8,415,414
|
|
|
$
|
5,201,181
|
|
|
$
|
11,679,963
|
|
General and administrative
|
|
|
2,468,607
|
|
|
|
2,980,170
|
|
|
|
3,425,690
|
|
|
|
1,845,574
|
|
|
|
2,733,979
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total operating expenses
|
|
|
(26,390,539
|
)
|
|
|
(13,862,492
|
)
|
|
|
(11,841,104
|
)
|
|
|
(7,046,755
|
)
|
|
|
(14,413,942
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Other income (expense)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Interest and other income
|
|
|
696,962
|
|
|
|
178,129
|
|
|
|
23,534
|
|
|
|
21,637
|
|
|
|
14,956
|
|
Interest and other expense
|
|
|
|
|
|
|
(296,303
|
)
|
|
|
(385,922
|
)
|
|
|
(96,298
|
)
|
|
|
(4,641,169
|
)
|
Beneficial conversion feature
|
|
|
|
|
|
|
(4,118,544
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total other income (expense)
|
|
|
696,962
|
|
|
|
(4,236,718
|
)
|
|
|
(362,388
|
)
|
|
|
(74,661
|
)
|
|
|
(4,626,213
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss
|
|
$
|
(25,693,577
|
)
|
|
$
|
(18,099,210
|
)
|
|
$
|
(12,203,492
|
)
|
|
$
|
(7,121,416
|
)
|
|
$
|
(19,040,155
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss per share-basic and
diluted(1)
|
|
$
|
(28.15
|
)
|
|
$
|
(13.47
|
)
|
|
$
|
(8.06
|
)
|
|
$
|
(4.80
|
)
|
|
$
|
(1.07
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Weighted-average number of shares used in share
calculation-basic and
diluted(2)
|
|
|
912,668
|
|
|
|
1,343,420
|
|
|
|
1,513,598
|
|
|
|
1,483,524
|
|
|
|
17,843,335
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(1)
|
Diluted earnings per share, or EPS, is identical to basic EPS
since common equivalent and shares are excluded from the
calculation, as their effect is anti-dilutive.
|
(2)
|
For accounting purposes only, the number of issued and
outstanding shares for the years ended December 31, 2007,
2008 and 2009 and the six months ended June 30, 2009 and
2010 do not include weighted-average shares of unvested stock of
261,649, 230,028, 110,079, 133,010 and 53,435, respectively.
These shares are subject to a risk of repurchase by us until
such shares are vested. See Note 10 to our financial
statements for more information.
|
9
|
|
|
|
|
|
|
|
|
|
|
As of June 30, 2010
|
|
|
|
|
|
|
As
|
|
|
|
Actual
|
|
|
Adjusted
|
|
|
Balance Sheet Data:
|
|
|
|
|
|
|
|
|
Cash and cash equivalents
|
|
$
|
35,787,481
|
|
|
$
|
64,864,517
|
|
Short-term investments
|
|
|
15,250,803
|
|
|
|
15,250,803
|
|
Working capital
|
|
|
48,800,562
|
|
|
|
77,877,598
|
|
Total assets
|
|
|
52,058,781
|
|
|
|
81,135,817
|
|
Indebtedness
|
|
|
3,230,861
|
|
|
|
3,230,861
|
|
Deficit accumulated during the development stage
|
|
|
(84,270,107
|
)
|
|
|
(84,270,107
|
)
|
Total stockholders (deficit) equity
|
|
|
48,827,920
|
|
|
|
77,904,956
|
|
The June 30, 2010 as adjusted balance sheet data reflects
the sale of 10,500,000 units, at an offering price of $3.00
per unit, with each unit consisting of one share of common stock
and a Warrant to purchase 0.40 shares of common stock, to
the selling stockholders pursuant to the private placement
transaction that closed on September 24, 2010. The above
table does not reflect any exercise by the selling stockholders
of the Warrants.
10
RISK
FACTORS
Before you decide to invest in our common stock, you should
carefully consider the risks described below, together with the
other information contained in this prospectus, including the
financial statements and the related notes that appear at the
end of this prospectus. We believe the risks described below are
the risks that are material to us as of the date of this
prospectus. If any of the following risks occur, our business,
financial condition, results of operations and future growth
prospects would likely be materially and adversely affected. In
these circumstances, the market price of our common stock could
decline, and you may lose all or part of your investment.
Risks
Related to Our Financial Condition and Capital
Requirements
We have incurred significant losses since our inception
and anticipate that we will incur continued significant losses
for the foreseeable future.
We are a development stage company with only six years of
operating history. We have focused primarily on developing our
three product candidates, varespladib,
A-623 and
varespladib sodium
(A-001). We
have financed our operations exclusively through equity
offerings and private placements of convertible debt and we have
incurred losses in each year since our inception in September
2004. Our net losses were approximately $15,000 in 2004,
$540,000 in 2005, $8.7 million in 2006, $25.7 million
in 2007, $18.1 million in 2008 and $12.2 million in
2009. For the six months ended June 30, 2010, our net loss
was $19.0 million and as of June 30, 2010, we had an
accumulated deficit of approximately $84.3 million.
Substantially all of our losses resulted from costs incurred in
connection with our product development programs and from
general and administrative costs associated with our operations.
We expect to incur additional losses over the next several
years, and these losses may increase if we cannot generate
revenues. These losses, combined with expected future losses,
have had and will continue to have an adverse effect on our
stockholders equity and working capital. We expect our
development expenses, as well as our clinical product
manufacturing expenses, to increase in connection with our
pivotal Phase 3 clinical study named VISTA-16 for varespladib
and our Phase 2b clinical study named PEARL-SC for
A-623. In
addition, we will incur additional costs of operating as a
public company and, if we obtain regulatory approval for any of
our product candidates, we may incur significant sales,
marketing, in-licensing and outsourced manufacturing expenses as
well as continued product development expenses. As a result, we
expect to continue to incur significant and increasing losses
for the foreseeable future.
We have never generated any revenue and may never be
profitable.
Our ability to generate revenue and achieve profitability
depends on our ability, alone or with collaborators, to
successfully complete the development of our product candidates,
conduct preclinical tests in animals and clinical studies in
human beings, obtain the necessary regulatory approvals for our
product candidates and commercialize any approved products. We
have not generated any revenue from our development-stage
product candidates, and we do not know when, or if, we will
generate any revenue. The commercial success of our
development-stage product candidates will depend on a number of
factors, including, but not limited to, our ability to:
|
|
|
|
|
obtain favorable results for and advance the development of our
lead product candidate, varespladib, for the treatment of acute
coronary syndrome, including successfully completing the
VISTA-16 study;
|
|
|
|
obtain favorable results for and advance the development of our
product candidate
A-623, for
the treatment of B-cell mediated autoimmune diseases, including
successfully completing a Phase 2b clinical study in patients
with systemic lupus erythematosus, or lupus;
|
11
|
|
|
|
|
obtain favorable results for and advance the development of our
product candidate
A-001, for
the prevention of acute chest syndrome associated with sickle
cell disease, including completing a multi-center Phase 2
clinical study;
|
|
|
|
successfully execute our planned preclinical studies in animals
and clinical studies in human beings for our other product
candidates;
|
|
|
|
obtain regulatory approval for varespladib,
A-623,
A-001 and
our other product candidates;
|
|
|
|
if regulatory approvals are obtained, begin the commercial
manufacturing of our product candidates with our third-party
manufacturers;
|
|
|
|
launch commercial sales and effectively market our product
candidates, either independently or in strategic collaborations
with third parties; and
|
|
|
|
achieve broad market acceptance of our product candidates in the
medical community and with third-party payors.
|
All of our product candidates are subject to the risks of
failure inherent in the development of therapeutics based on new
technologies. Currently, we have three product candidates in
clinical development: varespladib,
A-623 and
A-001. These
product candidates could fail in clinical studies if we are
unable to demonstrate that they are effective or if they cause
unacceptable adverse effects in the patients we treat. Failure
of our product candidates in clinical studies would have a
material adverse effect on our ability to generate revenue or
become profitable. If we are not successful in achieving
regulatory approval for our product candidates or are
significantly delayed in doing so, our business will be
materially harmed.
Additionally, all of our other product candidates are in
preclinical development. Our drug discovery efforts may not
produce any other viable or marketable product candidates. We do
not expect any of our potential product candidates to be
commercially available until at least 2013.
Even if our product candidates are approved for commercial sale,
the approved product candidate may not gain market acceptance or
achieve commercial success. Physicians, patients, payors or the
medical community in general may be unwilling to accept, utilize
or recommend any of our products. We would anticipate incurring
significant costs associated with commercializing any approved
product. Even if we are able to generate product sales, which we
cannot guarantee, we may not achieve profitability soon
thereafter, if ever. If we are unable to generate product
revenues, we will not become profitable and may be unable to
continue operations without additional funding.
Because we will need substantial additional capital in the
future to fund our operations, our independent registered public
accounting firm included a paragraph regarding concerns about
our ability to continue as a going concern in their report on
our financial statements. If additional capital is not
available, we will have to delay, reduce or cease
operations.
We will need to raise substantial additional capital to fund our
operations and to develop our product candidates. Our future
capital requirements could be substantial and will depend on
many factors including:
|
|
|
|
|
the rate of progress of our VISTA-16 study for varespladib and
our Phase 2b clinical study for
A-623;
|
|
|
|
the scope, size, rate of progress, results and costs of our
preclinical studies, clinical studies and other development
activities for one or more of our other product candidates;
|
|
|
|
the cost, timing and outcomes of regulatory proceedings;
|
12
|
|
|
|
|
payments received under any strategic collaborations;
|
|
|
|
the filing, prosecution and enforcement of patent claims;
|
|
|
|
the costs associated with commercializing our product candidates
if they receive regulatory approval, including the cost and
timing of developing sales and marketing capabilities, or
entering into strategic collaboration with others relating to
the commercialization of our product candidates; and
|
|
|
|
revenues received from approved products, if any, in the future.
|
As of the date of this prospectus, we anticipate that our
existing cash, cash equivalents and short-term investments, will
enable us to maintain our currently planned operations through
at least the next 12 months. Changing circumstances may
cause us to consume capital significantly faster than we
currently anticipate. Additional financing may not be available
when we need it or may not be available on terms that are
favorable to us. If adequate funds are not available to us on a
timely basis, or at all, we may be required to:
|
|
|
|
|
terminate, reduce or delay preclinical studies, clinical studies
or other development activities for one or more of our product
candidates; or
|
|
|
|
terminate, reduce or delay our (i) establishment of sales
and marketing capabilities, (ii) pursuit of strategic
collaborations with others relating to the sales, marketing and
commercialization of our product candidates or (iii) other
activities that may be necessary to commercialize our product
candidates, if approved for sale.
|
The timing of the milestone and royalty payments we are
required to make to each of Eli Lilly and Company,
Shionogi & Co., Ltd. and Amgen Inc. is uncertain and
could adversely affect our cash flows and results of
operations.
In July 2006, we entered into a license agreement with Eli Lilly
and Company, or Eli Lilly, and Shionogi & Co., Ltd. to
develop and commercialize certain secretory phospholipase
A2,
or
sPLA2,
inhibitors for the treatment of cardiovascular disease and other
diseases. Pursuant to our license agreement with them, we have
an obligation to pay to each of Eli Lilly and
Shionogi & Co., Ltd. significant milestone and royalty
payments based upon how we develop and commercialize certain
sPLA2
inhibitors, including varespladib and
A-001, and
our achievement of certain significant corporate, clinical and
financial events. For varespladib, we are required to pay up to
$32.0 million upon achievement of certain approval and
post-approval sales milestones. For
A-001, we
are required to pay up to $3.0 million upon achievement of
certain clinical development milestones and up to
$25.0 million upon achievement of certain approval and
post-approval sales milestones. For other product formulations
that we are not currently developing, we would be required to
pay up to $2.0 million upon achievement of certain clinical
development milestones and up to $35.5 million upon
achievement of certain approval and post- approval sales
milestones. In addition, in December 2007, we entered into a
license agreement with Amgen Inc., or Amgen, pursuant to which
we obtained an exclusive worldwide license to certain technology
and compounds relating to
A-623.
Pursuant to our license agreement with Amgen, we are required to
make various milestone payments upon our achievement of certain
development, regulatory and commercial objectives for any
A-623
formulation. We are required to pay up to $10.0 million
upon achievement of certain pre-approval clinical development
milestones and up to $23.0 million upon achievement of
certain post-approval milestones. We are also required to make
tiered quarterly royalty payments on net sales, which increase
as a percentage from the high single digits to the low double
digits as net sales increase. The timing of our achievement of
these events and corresponding milestone payments becoming due
to Eli Lilly, Shionogi & Co., Ltd. and Amgen is
subject to factors relating to the clinical and regulatory
development and commercialization of certain
sPLA2
inhibitors or
A-623, as
applicable, many of which are beyond our control. We may become
obligated
13
to make a milestone payment during a period in which we do not
have the cash on hand to make such payment, which could require
us to delay our clinical studies, curtail our operations, scale
back our commercialization and marketing efforts or seek funds
to meet these obligations at terms unfavorable to us.
Our limited operating history makes it difficult to
evaluate our business and prospects.
We were incorporated in September 2004. Our operations to date
have been limited to organizing and staffing our company,
acquiring product and technology rights, conducting product
development activities for our primary product candidates,
varespladib,
A-623 and
A-001, and
performing research and development. We have not yet
demonstrated an ability to obtain regulatory approval for or
commercialize a product candidate. Consequently, any predictions
about our future performance may not be as accurate as they
could be if we had a history of successfully developing and
commercializing pharmaceutical products.
Risks
Associated with Development and Commercialization of Our Product
Candidates
We depend substantially on the success of our three
primary product candidates, varespladib,
A-623 and
A-001, which
are still under clinical development. We cannot assure you that
these product candidates or any of our other product candidates
will receive regulatory approval or be successfully
commercialized.
To date, we have not marketed, distributed or sold any product
candidates. The success of our business depends primarily upon
our ability to develop and commercialize our three primary
product candidates successfully. Our lead product candidate is
varespladib, which has completed its Phase 2 clinical studies
and for which we have received (i) an agreement from the
U.S. Food and Drug Administration, or FDA, on a Special
Protocol Assessment, or SPA, for the VISTA-16 Phase 3 study
protocol, and (ii) scientific advice from the European
Medicines Agency on our European development strategy for
varespladib. We initiated the VISTA-16 study for varespladib in
June 2010.
Our next product candidate is
A-623, which
has completed several Phase 1 clinical studies and recently
began enrollment for our Phase 2b clinical study. In July 2010,
we received clearance from the FDA to begin recruitment of lupus
patients into the PEARL-SC phase 2b clinical study.
Our third product candidate is
A-001. Our
product candidates are prone to the risks of failure inherent in
drug development. Before obtaining regulatory approvals for the
commercial sale of any product candidate for a target
indication, we must demonstrate with substantial evidence
gathered in preclinical and well-controlled clinical studies,
and, with respect to approval in the United States, to the
satisfaction of the FDA and, with respect to approval in other
countries, similar regulatory authorities in those countries,
that the product candidate is safe and effective for use for
that target indication and that the manufacturing facilities,
processes and controls are adequate. Despite our efforts, our
product candidates may not:
|
|
|
|
|
offer therapeutic or other improvement over existing, comparable
therapeutics;
|
|
|
|
be proven safe and effective in clinical studies;
|
|
|
|
meet applicable regulatory standards;
|
|
|
|
be capable of being produced in sufficient quantities at
acceptable costs;
|
|
|
|
be successfully commercialized; or
|
|
|
|
obtain favorable reimbursement.
|
We are not permitted to market our varespladib and
A-001
product candidates in the United States until we receive
approval of a new drug application, or NDA, or with respect to
our A-623
product candidate, approval of a biologics license application,
or BLA, from the FDA, or in any foreign
14
countries until we receive the requisite approval from such
countries. We have not submitted an NDA or BLA or received
marketing approval for any of our product candidates.
Preclinical testing and clinical studies are long, expensive and
uncertain processes. We may spend several years completing our
testing for any particular product candidate, and failure can
occur at any stage. Negative or inconclusive results or adverse
medical events during a clinical study could also cause the FDA
or us to terminate a clinical study or require that we repeat it
or conduct additional clinical studies. Additionally, data
obtained from a clinical study are susceptible to varying
interpretations and the FDA or other regulatory authorities may
interpret the results of our clinical studies less favorably
than we do. The FDA and equivalent foreign regulatory agencies
have substantial discretion in the approval process and may
decide that our data are insufficient to support a marketing
application and require additional preclinical, clinical or
other studies.
Any termination or suspension of, or delays in the
commencement or completion of, clinical testing of our product
candidates could result in increased costs to us, delay or limit
our ability to generate revenue and adversely affect our
commercial prospects.
Delays in the commencement or completion of clinical testing
could significantly affect our product development costs. We do
not know whether planned clinical studies will begin on time or
be completed on schedule, if at all. The commencement and
completion of clinical studies can be delayed for a number of
reasons, including delays related to:
|
|
|
|
|
obtaining regulatory approval to commence a clinical study or
complying with conditions imposed by a regulatory authority
regarding the scope or design of a clinical study;
|
|
|
|
reaching agreement on acceptable terms with prospective clinical
research organizations, or CROs, and study sites, the terms of
which can be subject to extensive negotiation and may vary
significantly among different CROs and study sites;
|
|
|
|
manufacturing, including manufacturing sufficient quantities of
a product candidate or other materials for use in clinical
studies;
|
|
|
|
obtaining institutional review board, or IRB, approval or the
approval of other reviewing entities to conduct a clinical study
at a prospective site;
|
|
|
|
recruiting and enrolling patients to participate in clinical
studies for a variety of reasons, including size of patient
population, nature of clinical study protocol, the availability
of approved effective treatments for the relevant disease and
competition from other clinical study programs for similar
indications;
|
|
|
|
severe or unexpected drug-related adverse effects experienced by
patients in a clinical study; and
|
|
|
|
retaining patients who have initiated a clinical study, but may
withdraw due to treatment protocol, adverse effects from the
therapy, lack of efficacy from the treatment, personal issues or
who are lost to further
follow-up.
|
Clinical studies may also be delayed, suspended or terminated as
a result of ambiguous or negative interim results, or results
that are inconsistent with earlier results. For example, the
Data Safety Monitoring Board, or DSMB, may recommend that we
stop our VISTA-16 study for varespladib if certain biomarkers of
inflammation and lipid profiles fail to meet pre-specified
reductions in the first 1,000 or more patients. In addition, a
clinical study may be suspended or terminated by us, the FDA,
15
the IRB or other reviewing entity overseeing the clinical study
at issue, any of our clinical study sites with respect to that
site, or other regulatory authorities due to a number of
factors, including:
|
|
|
|
|
failure to conduct the clinical study in accordance with
regulatory requirements or our clinical protocols;
|
|
|
|
inspection of the clinical study operations or study sites by
the FDA or other regulatory authorities resulting in the
imposition of a clinical hold;
|
|
|
|
unforeseen safety issues or any determination that a clinical
study presents unacceptable health risks; and
|
|
|
|
lack of adequate funding to continue the clinical study,
including the incurrence of unforeseen costs due to enrollment
delays, requirements to conduct additional clinical studies and
increased expenses associated with the services of our CROs and
other third parties.
|
Product development costs to us and our collaborators will
increase if we have delays in testing or approval of our product
candidates or if we need to perform more or larger clinical
studies than planned. For example, we may need to increase our
sample size for our VISTA-16 study for varespladib if the
overall major adverse cardiovascular event, or MACE, rate is
lower than expected. We typically rely on third-party clinical
investigators at medical institutions and health care facilities
to conduct our clinical studies and, as a result, we may face
additional delaying factors outside our control.
Additionally, changes in regulatory requirements and policies
may occur and we may need to amend clinical study protocols to
reflect these changes. Amendments may require us to resubmit our
clinical study protocols to IRBs for reexamination, which may
impact the costs, timing or successful completion of a clinical
study. If we experience delays in completion of, or if we, the
FDA or other regulatory authorities, the IRB or other reviewing
entities, or any of our clinical study sites suspend or
terminate any of our clinical studies, the commercial prospects
for our product candidates may be harmed and our ability to
generate product revenues will be delayed. In addition, many of
the factors that cause, or lead to, termination or suspension
of, or a delay in the commencement or completion of, clinical
studies may also ultimately lead to the denial of regulatory
approval of a product candidate. Also, if one or more clinical
studies are delayed, our competitors may be able to bring
products to market before we do, and the commercial viability of
our product candidates could be significantly reduced.
The results of biomarker assays in earlier clinical
studies in varespladib are not necessarily predictive of future
results, and therefore the results of biomarker assays in the
VISTA-16 study may not be similar to those observed
previously.
Success in our Phase 2 clinical studies in lowering low-density
lipoprotein cholesterol, or LDL-C, C-reactive protein, or CRP,
sPLA2
and interleukin-6, or IL-6, during treatment with varespladib
does not ensure that later clinical studies, such as our
VISTA-16 study, will demonstrate similar reductions in these
biomarkers. Each of these biomarkers has been associated with an
increased risk for secondary MACE following an acute coronary
syndrome. Our inability to demonstrate similar biomarker effects
in our VISTA-16 study may reduce our ability to achieve our
primary endpoint to reduce MACE and to achieve regulatory
approval of varespladib.
16
Because the results of preclinical testing or earlier
clinical studies are not necessarily predictive of future
results, varespladib,
A-623,
A-001 or any
other product candidate we advance into clinical studies may not
have favorable results in later clinical studies or receive
regulatory approval.
Success in preclinical testing and early clinical studies does
not ensure that later clinical studies will generate adequate
data to demonstrate the efficacy and safety of an
investigational drug or biologic. A number of companies in the
pharmaceutical and biotechnology industries, including those
with greater resources and experience, have suffered significant
setbacks in Phase 3 clinical studies, even after seeing
promising results in earlier clinical studies. Despite the
results reported in earlier clinical studies for our product
candidates, including varespladib,
A-623 and
A-001, we do
not know whether any Phase 3 or other clinical studies we may
conduct will demonstrate adequate efficacy and safety to result
in regulatory approval to market any of our product candidates.
If later stage clinical studies do not produce favorable
results, our ability to achieve regulatory approval for any of
our product candidates may be adversely impacted.
If we breach the license agreements for our primary
product candidates, we could lose the ability to continue the
development and commercialization of our primary product
candidates.
We are party to an agreement with Eli Lilly and
Shionogi & Co., Ltd. containing exclusive, worldwide
licenses, except for Japan, of the composition of matter,
methods of making and methods of use for certain
sPLA2
inhibitors. We are also party to an agreement with Amgen
containing exclusive, worldwide licenses of the composition of
matter and methods of use for
A-623. These
agreements require us to make timely milestone and royalty
payments, provide regular information, maintain the
confidentiality of and indemnify Eli Lilly, Shionogi &
Co., Ltd. and Amgen under the terms of the agreements.
If we fail to meet these obligations, our licensors may
terminate our exclusive licenses and may be able to re-obtain
licensed technology and aspects of any intellectual property
controlled by us that relate to the licensed technology that
originated from the licensors. Our licensors could effectively
take control of the development and commercialization of
varespladib,
A-623 and
A-001 after
an uncured, material breach of our license agreements by us or
if we voluntarily terminate the agreements. While we would
expect to exercise all rights and remedies available to us,
including seeking to cure any breach by us, and otherwise seek
to preserve our rights under the patents licensed to us, we may
not be able to do so in a timely manner, at an acceptable cost
or at all. Any uncured, material breach under the licenses could
result in our loss of exclusive rights and may lead to a
complete termination of our product development and any
commercialization efforts for varespladib,
A-623 or
A-001.
Our industry is subject to intense competition. If we are
unable to compete effectively, our product candidates may be
rendered non-competitive or obsolete.
The pharmaceutical industry is highly competitive and subject to
rapid and significant technological change. Our potential
competitors include large pharmaceutical and more established
biotechnology companies, specialty pharmaceutical and generic
drug companies, academic institutions, government agencies and
other public and private research organizations that conduct
research, seek patent protection and establish collaborative
arrangements for research, development, manufacturing and
commercialization. All of these competitors currently engage in,
have engaged in or may engage in the future in the development,
manufacturing, marketing and commercialization of
pharmaceuticals and biotechnologies, some of which may compete
with our present or future product candidates. It is possible
that any of these competitors could develop technologies or
products that would render our product candidates obsolete or
non-competitive, which could adversely affect our revenue
potential. Key competitive factors affecting the commercial
success of our product candidates are likely to be efficacy,
safety profile, reliability, convenience of dosing, price and
reimbursement.
17
The market for inflammatory disease therapeutics is especially
large and competitive. All of the
sPLA2
inhibitor compounds we are currently developing, if approved,
will face intense competition, either as monotherapies or in
combination therapies. We are aware of other companies with
products in development that are being tested for
anti-inflammatory benefits in patients with acute coronary
syndrome, such as Via Pharmaceuticals, Inc. and its
5-lipoxygenase, or 5-LO, inhibitor, which has been evaluated in
Phase 2 clinical studies; and GlaxoSmithKline plc and its
product candidate, darapladib, which is a lipoprotein associated
phospholipase
A2,
or
Lp-PLA2,
inhibitor currently being evaluated in Phase 3 clinical studies.
Although there are no
sPLA2
inhibitor compounds currently approved by the FDA for the
treatment of acute chest syndrome associated with sickle cell
disease, Droxia, or hydroxyurea, is approved for the prevention
of vaso-occlusive crisis, or VOC, in sickle cell disease and
thus could reduce the pool of patients with VOC at risk for
acute chest syndrome. Further, we are aware of companies with
other products in development that are being tested for
potential treatment of lupus, including Human Genome Sciences,
Inc. and GlaxoSmithKline plc, who have a BAFF antagonist
monoclonal antibody product candidate, Benlysta, which recently
reported favorable results from a Phase 3 clinical study in
lupus; ZymoGenetics, Inc. and Merck Serono S.A., whose dual
BAFF/APRIL antagonist fusion protein, Atacicept, is in a Phase 3
clinical study for lupus; and Immunomedics, Inc. and UCB S.A.,
who recently reported favorable results for their CD-22
antagonist humanized antibody, epratuzumab, which completed a
Phase 2b clinical study in lupus.
Many of our potential competitors have substantially greater
financial, technical and human resources than we do and
significantly greater experience in the discovery and
development of drug candidates, obtaining FDA and other
regulatory approvals of products and the commercialization of
those products. Accordingly, our competitors may be more
successful than we may be in obtaining FDA approval for drugs
and achieving widespread market acceptance. Our
competitors drugs may be more effective, have fewer
adverse effects, be less expensive to develop and manufacture or
be more effectively marketed and sold than any product candidate
we may commercialize and may render our product candidates
obsolete or non-competitive before we can recover the expenses
of developing and commercializing any of our product candidates.
We anticipate that we will face intense and increasing
competition as new drugs enter the market and advanced
technologies become available. These entities may also establish
collaborative or licensing relationships with our competitors.
Finally, the development of new treatment methods for the
diseases we are targeting could render our drugs non-competitive
or obsolete. All of these factors could adversely affect our
business.
Our product candidates may cause undesirable adverse
effects or have other properties that could delay or prevent
their regulatory approval or limit the commercial profile of any
approved label.
Undesirable adverse effects caused by our product candidates
could cause us, IRBs or other reviewing entities, clinical study
sites, or regulatory authorities to interrupt, delay or halt
clinical studies and could result in the denial of regulatory
approval by the FDA or other regulatory authorities. Phase 2
clinical studies conducted by us with our product candidates
have generated differences in adverse effects and serious
adverse events. The most common adverse effects seen with any of
our product candidates versus placebo include diarrhea,
headache, nausea and increases in alanine aminotransferase,
which is an enzyme that indicates liver cell injury. The most
common serious adverse events seen with any of our product
candidates include death, VOC and congestive heart failure.
While none of these serious adverse events were considered
related to the administration of our product candidates by the
clinical investigators, if serious adverse events that are
considered related to our product candidates are observed in any
Phase 3 clinical studies, our ability to obtain regulatory
approval for our product candidates may be adversely impacted.
Further, if any of our product candidates receives marketing
approval and we or others later discover, after approval and use
in an increasing number of patients, that our products could
have adverse effect profiles that limit their usefulness or
require their
18
withdrawal (whether or not the therapies showed the adverse
effect profile in Phase 1 through Phase 3 clinical studies), a
number of potentially significant negative consequences could
result, including:
|
|
|
|
|
regulatory authorities may withdraw their approval of the
product;
|
|
|
|
regulatory authorities may require the addition of labeling
statements, such as warnings or contraindications;
|
|
|
|
we may be required to change the way the product is
administered, conduct additional clinical studies or change the
labeling of the product;
|
|
|
|
we could be sued and held liable for harm caused to
patients; and
|
|
|
|
our reputation may suffer.
|
Any of these events could prevent us from achieving or
maintaining market acceptance of the affected product candidate
and could substantially increase the costs of commercializing
our product candidates.
After the completion of our clinical studies, we cannot
predict whether or when we will obtain regulatory approval to
commercialize our product candidates and we cannot, therefore,
predict the timing of any future revenue from these product
candidates.
We cannot commercialize any of our product candidates until the
appropriate regulatory authorities have reviewed and approved
the applications for the product candidates. We cannot assure
you that the regulatory agencies will complete their review
processes in a timely manner or that we will obtain regulatory
approval for any product candidate we develop. Satisfaction of
regulatory requirements typically takes many years, is dependent
upon the type, complexity and novelty of the product and
requires the expenditure of substantial resources. In addition,
we may experience delays or rejections based upon additional
government regulation from future legislation or administrative
action or changes in FDA policy during the period of product
development, clinical studies and FDA regulatory review.
Our agreement with the FDA on an SPA for our VISTA-16
study of varespladib for the potential treatment of acute
coronary syndrome does not guarantee any particular outcome from
regulatory review of the study or the product candidate.
The FDAs SPA process creates a written agreement between
the sponsoring company and the FDA regarding clinical study
design and other clinical study issues that can be used to
support approval of a product candidate. The SPA is intended to
provide assurance that if the agreed upon clinical study
protocols are followed and the clinical study endpoints are
achieved, the data may serve as the primary basis for an
efficacy claim in support of an NDA. However, the SPA agreement
is not a guarantee of an approval of a product or any
permissible claims about the product. In particular, the SPA is
not binding on the FDA if public health concerns unrecognized at
the time of the SPA agreement is entered into become evident,
other new scientific concerns regarding product safety or
efficacy arise or if the sponsor company fails to comply with
the agreed upon clinical study protocols. Although we have an
agreement with the FDA on an SPA for our VISTA-16 clinical study
of varespladib for the potential short-term (16-week) treatment
of acute coronary syndrome, we do not know how the FDA will
interpret the commitments under our agreed upon SPA, how it will
interpret the data and results or whether it will approve our
varespladib product candidate for the short-term (16-week)
treatment of acute coronary syndrome. Regardless of our SPA
agreement, we cannot guarantee any particular outcome from
regulatory review of our VISTA-16 study.
19
Even if our product candidates receive regulatory
approval, they may still face future development and regulatory
difficulties.
Even if U.S. regulatory approval is obtained, the FDA may
still impose significant restrictions on a products
indicated uses or marketing or impose ongoing requirements for
potentially costly post-approval studies or post-market
surveillance. For example, the label ultimately approved for
varespladib, if any, may include restrictions on use. Further,
the FDA has indicated that long-term safety data on varespladib
may need to be obtained as a post-market requirement. Our
product candidates will also be subject to ongoing FDA
requirements governing the labeling, packaging, storage,
distribution, safety surveillance, advertising, promotion,
recordkeeping and reporting of safety and other post-market
information. In addition, manufacturers of drug products and
their facilities are subject to continual review and periodic
inspections by the FDA and other regulatory authorities for
compliance with current good manufacturing practices, or cGMP,
regulations. If we or a regulatory agency discovers previously
unknown problems with a product, such as adverse events of
unanticipated severity or frequency, or problems with the
facility where the product is manufactured, a regulatory agency
may impose restrictions on that product, the manufacturing
facility or us, including requiring recall or withdrawal of the
product from the market or suspension of manufacturing. If we,
our product candidates or the manufacturing facilities for our
product candidates fail to comply with applicable regulatory
requirements, a regulatory agency may:
|
|
|
|
|
issue warning letters or untitled letters;
|
|
|
|
seek an injunction or impose civil or criminal penalties or
monetary fines;
|
|
|
|
suspend or withdraw regulatory approval;
|
|
|
|
suspend any ongoing clinical studies;
|
|
|
|
refuse to approve pending applications or supplements to
applications filed by us;
|
|
|
|
suspend or impose restrictions on operations, including costly
new manufacturing requirements; or
|
|
|
|
seize or detain products, refuse to permit the import or export
of products, or require us to initiate a product recall.
|
The occurrence of any event or penalty described above may
inhibit our ability to commercialize our products and generate
revenue.
New legal and regulatory requirements could make it more
difficult for us to obtain approvals for our product candidates
and could limit or make more burdensome our ability to
commercialize any approved products.
New federal legislation or regulatory requirements could affect
the requirements for obtaining regulatory approvals of our
product candidates or otherwise limit our ability to
commercialize any approved products or subject our products to
more rigorous post-approval requirements. For example, the FDA
Amendments Act of 2007, or FDAAA, granted the FDA new authority
to impose post-approval clinical study requirements, require
safety-related changes to product labeling and require the
adoption of risk management plans, referred to in the
legislation as risk evaluation and mitigation strategies, or
REMS. The REMS may include requirements for special labeling or
medication guides for patients, special communication plans to
health care professionals, and restrictions on distribution and
use. Pursuant to the FDAAA, if the FDA makes the requisite
findings, it might require that a new product be used only
20
by physicians with specified specialized training, only in
specified designated health care settings, or only in
conjunction with special patient testing and monitoring. The
legislation also included the following: requirements for
providing the public information on ongoing clinical studies
through a clinical study registry and for disclosing clinical
study results to the public through such registry; renewed
requirements for conducting clinical studies to generate
information on the use of products in pediatric patients; and
substantial new penalties, for example, for false or misleading
consumer advertisements. Other proposals have been made to
impose additional requirements on drug approvals, further expand
post-approval requirements, and restrict sales and promotional
activities. The new legislation, and the additional proposals if
enacted, may make it more difficult or burdensome for us to
obtain approval of our product candidates, any approvals we
receive may be more restrictive or be subject to onerous
post-approval requirements, our ability to successfully
commercialize approved products may be hindered and our business
may be harmed as a result.
If any of our product candidates for which we receive
regulatory approval does not achieve broad market acceptance,
the revenue that we generate from its sales, if any, will be
limited.
The commercial success of our product candidates for which we
obtain marketing approval from the FDA or other regulatory
authorities will depend upon the acceptance of these products by
the medical community, including physicians, patients and health
care payors. The degree of market acceptance of any of our
approved products will depend on a number of factors, including:
|
|
|
|
|
demonstration of clinical safety and efficacy compared to other
products;
|
|
|
|
the relative convenience, ease of administration and acceptance
by physicians and payors of varespladib in the treatment of
acute coronary syndrome,
A-623 in the
treatment of lupus and
A-001 in the
prevention of acute chest syndrome associated with sickle cell
disease;
|
|
|
|
the prevalence and severity of any adverse effects;
|
|
|
|
limitations or warnings contained in a products
FDA-approved labeling;
|
|
|
|
availability of alternative treatments, including, in the case
of varespladib, a number of competitive products being studied
for anti-inflammatory benefits in patients with acute coronary
syndrome or expected to be commercially launched in the near
future;
|
|
|
|
pricing and cost-effectiveness;
|
|
|
|
the effectiveness of our or any future collaborators sales
and marketing strategies;
|
|
|
|
our ability to obtain and maintain sufficient third-party
coverage or reimbursement from government health care programs,
including Medicare and Medicaid; and
|
|
|
|
the willingness of patients to pay
out-of-pocket
in the absence of third-party coverage.
|
If our product candidates are approved but do not achieve an
adequate level of acceptance by physicians, health care payors
and patients, we may not generate sufficient revenue from these
products, and we may not become or remain profitable. In
addition, our efforts to educate the medical community and
third-party payors on the benefits of our product candidates may
require significant resources and may never be successful.
21
Our future success depends on our ability to retain our
chief executive officer and other key executives and to attract,
retain and motivate qualified personnel.
We are highly dependent on Mr. Paul F. Truex, our President
and Chief Executive Officer, Dr. Colin Hislop, our Senior
Vice President and Chief Medical Officer and the other principal
members of our executive team listed under
Management on page 109. The loss of the
services of any of these persons might impede the achievement of
our research, development and commercialization objectives.
Recruiting and retaining qualified scientific personnel and
possibly sales and marketing personnel will also be critical to
our success. We may not be able to attract and retain these
personnel on acceptable terms given the competition among
numerous pharmaceutical and biotechnology companies for similar
personnel. We also experience competition for the hiring of
scientific personnel from universities and research
institutions. Failure to succeed in clinical studies may make it
more challenging to recruit and retain qualified scientific
personnel. In addition, we rely on consultants and advisors,
including scientific and clinical advisors, to assist us in
formulating our research and development and commercialization
strategy. Our consultants and advisors may be employed by
employers other than us and may have commitments under
consulting or advisory contracts with other entities that may
limit their availability to us.
Recently enacted and future legislation or regulatory
reform of the health care system in the United States and
foreign jurisdictions may affect our ability to sell our
products profitably.
Our ability to commercialize our future products successfully,
alone or with collaborators, will depend in part on the extent
to which reimbursement for the products will be available from
government and health administration authorities, private health
insurers and other third-party payors. The continuing efforts of
the U.S. and foreign governments, insurance companies,
managed care organizations and other payors of health care
services to contain or reduce health care costs may adversely
affect our ability to set prices for our products which we
believe are fair, and our ability to generate revenues and
achieve and maintain profitability.
Specifically, in both the United States and some foreign
jurisdictions, there have been a number of legislative and
regulatory proposals to change the health care system in ways
that could affect our ability to sell our products profitably.
In March 2010, President Obama signed into law the Patient
Protection and Affordable Care Act, as amended by the Health
Care and Education Reconciliation Act, or collectively, the
Health Care Reform Law, a sweeping law intended to broaden
access to health insurance, reduce or constrain the growth of
healthcare spending, enhance remedies against fraud and abuse,
add new transparency requirements for healthcare and health
insurance industries, impose new taxes and fees on the health
industry and impose additional health policy reforms.
We will not know the full effects of the Health Care Reform Law
until applicable federal and state agencies issue regulations or
guidance under the new law. Although it is too early to
determine the effect of the Health Care Reform Law, the new law
appears likely to continue the pressure on pharmaceutical
pricing, especially under the Medicare program, and also may
increase our regulatory burdens and operating costs. We expect
further federal and state proposals and health care reforms to
continue to be proposed by legislators, which could limit the
prices that can be charged for the products we develop and may
limit our commercial opportunity.
Also in the United States, the Medicare Prescription Drug,
Improvement, and Modernization Act of 2003, also called the
Medicare Modernization Act, or MMA, changed the way Medicare
covers and pays for pharmaceutical products. The legislation
expanded Medicare coverage for drug purchases by the elderly and
introduced a new reimbursement methodology based on average
sales prices for drugs. In addition, this legislation authorized
Medicare Part D prescription drug plans to use formularies
where they can limit the number of drugs that will be covered in
any therapeutic class. As a result of this legislation and the
expansion of federal coverage of drug products, we expect that
there will be
22
additional pressure to contain and reduce costs. These cost
reduction initiatives and other provisions of this legislation
could decrease the coverage and price that we receive for any
approved products and could seriously harm our business. While
the MMA applies only to drug benefits for Medicare
beneficiaries, private payors often follow Medicare coverage
policy and payment limitations in setting their own
reimbursement rates, and any reduction in reimbursement that
results from the MMA may result in a similar reduction in
payments from private payors.
The continuing efforts of government and other third-party
payors to contain or reduce the costs of health care through
various means may limit our commercial opportunity. It will be
time-consuming and expensive for us to go through the process of
seeking reimbursement from Medicare and private payors. Our
products may not be considered cost-effective, and government
and third-party private health insurance coverage and
reimbursement may not be available to patients for any of our
future products or sufficient to allow us to sell our products
on a competitive and profitable basis. Our results of operations
could be adversely affected by the MMA, the Health Care Reform
Law, and additional prescription drug coverage legislation, by
the possible effect of this legislation on amounts that private
insurers will pay and by other health care reforms that may be
enacted or adopted in the future. In addition, increasing
emphasis on managed care in the United States will continue to
put pressure on the pricing of pharmaceutical products. Cost
control initiatives could decrease the price that we or any
potential collaborators could receive for any of our future
products and could adversely affect our profitability.
In some foreign countries, including major markets in the
European Union and Japan, the pricing of prescription
pharmaceuticals is subject to governmental control. In these
countries, pricing negotiations with governmental authorities
can take six to 12 months or longer after the receipt of
regulatory marketing approval for a product. To obtain
reimbursement or pricing approval in some countries, we may be
required to conduct a clinical study that compares the
cost-effectiveness of our product candidates to other available
therapies. Such pharmacoeconomic studies can be costly and the
results uncertain. Our business could be harmed if reimbursement
of our products is unavailable or limited in scope or amount or
if pricing is set at unsatisfactory levels.
We face potential product liability exposure, and, if
successful claims are brought against us, we may incur
substantial liability.
The use of our product candidates in clinical studies and the
sale of any products for which we obtain marketing approval
expose us to the risk of product liability claims. Product
liability claims might be brought against us by consumers,
health care providers, pharmaceutical companies or others
selling or otherwise coming into contact with our products. If
we cannot successfully defend ourselves against product
liability claims, we could incur substantial liabilities. In
addition, regardless of merit or eventual outcome, product
liability claims may result in:
|
|
|
|
|
impairment of our business reputation;
|
|
|
|
withdrawal of clinical study participants;
|
|
|
|
costs of related litigation;
|
|
|
|
distraction of managements attention from our primary
business;
|
|
|
|
substantial monetary awards to patients or other claimants;
|
|
|
|
the inability to commercialize our product candidates; and
|
|
|
|
decreased demand for our product candidates, if approved for
commercial sale.
|
23
Our product liability insurance coverage, with a
$5.0 million annual aggregate coverage limit, for our
clinical studies may not be sufficient to reimburse us for any
expenses or losses we may suffer. Moreover, insurance coverage
is becoming increasingly expensive, and, in the future, we may
not be able to maintain insurance coverage at a reasonable cost
or in sufficient amounts to protect us against losses due to
liability. If and when we obtain marketing approval for any of
our product candidates, we intend to expand our insurance
coverage to include the sale of commercial products; however, we
may be unable to obtain this product liability insurance on
commercially reasonable terms. On occasion, large judgments have
been awarded in class action lawsuits based on drugs that had
unanticipated adverse effects. A successful product liability
claim or series of claims brought against us could cause our
stock price to decline and, if judgments exceed our insurance
coverage, could decrease our cash and adversely affect our
business.
If we use hazardous and biological materials in a manner
that causes injury or violates applicable law, we may be liable
for damages.
Our research and development activities involve the controlled
use of potentially hazardous substances, including toxic
chemical and biological materials. We could be held liable for
any contamination, injury or other damages resulting from these
hazardous substances. In addition, our operations produce
hazardous waste products. While third parties are responsible
for disposal of our hazardous waste, we could be liable under
environmental laws for any required cleanup of sites at which
our waste is disposed. Federal, state, foreign and local laws
and regulations govern the use, manufacture, storage, handling
and disposal of these hazardous materials. If we fail to comply
with these laws and regulations at any time, or if they change,
we may be subject to criminal sanctions and substantial civil
liabilities, which may harm our business. Even if we continue to
comply with all applicable laws and regulations regarding
hazardous materials, we cannot eliminate the risk of accidental
contamination or discharge and our resultant liability for any
injuries or other damages caused by these accidents.
We rely on third parties to conduct, supervise and monitor
our clinical studies, and those third parties may perform in an
unsatisfactory manner, such as by failing to meet established
deadlines for the completion of these clinical studies, or may
harm our business if they suffer a catastrophic
event.
We rely on third parties such as CROs, medical institutions and
clinical investigators to enroll qualified patients and conduct,
supervise and monitor our clinical studies. Our reliance on
these third parties for clinical development activities reduces
our control over these activities. Our reliance on these third
parties, however, does not relieve us of our regulatory
responsibilities, including ensuring that our clinical studies
are conducted in accordance with good clinical practices and the
investigational plan and protocols contained in the relevant
regulatory application, such as the investigational new drug
application, or IND. In addition, the CROs with which we
contract may not complete activities on schedule, or may not
conduct our preclinical studies or clinical studies in
accordance with regulatory requirements or our clinical study
design. If these third parties do not successfully carry out
their contractual duties or meet expected deadlines, our efforts
to obtain regulatory approvals for, and to commercialize, our
product candidates may be delayed or prevented. In addition, if
a catastrophe such as an earthquake, fire, flood or power loss
should affect one of the third parties on which we rely, our
business prospects could be harmed. For example, if a central
laboratory holding all of our clinical study samples were to
suffer a catastrophic loss of their facility, we would lose all
of our samples and would have to repeat our studies.
24
Any failure by our third-party manufacturers on which we
rely to produce our preclinical and clinical drug supplies and
on which we intend to rely to produce commercial supplies of any
approved product candidates may delay or impair our ability to
commercialize our product candidates.
We have relied upon a small number of third-party manufacturers
and active pharmaceutical ingredient formulators for the
manufacture of our material for preclinical and clinical testing
purposes and intend to continue to do so in the future. We also
expect to rely upon third parties to produce materials required
for the commercial production of our product candidates if we
succeed in obtaining necessary regulatory approvals. If we are
unable to arrange for third-party manufacturing sources, or to
do so on commercially reasonable terms, we may not be able to
complete development of our product candidates or market them.
Reliance on third-party manufacturers entails risks to which we
would not be subject if we manufactured product candidates
ourselves, including reliance on the third party for regulatory
compliance and quality assurance, the possibility of breach of
the manufacturing agreement by the third party because of
factors beyond our control (including a failure to synthesize
and manufacture our product candidates in accordance with our
product specifications) and the possibility of termination or
nonrenewal of the agreement by the third party, based on its own
business priorities, at a time that is costly or damaging to us.
In addition, the FDA and other regulatory authorities require
that our product candidates be manufactured according to cGMP
and similar foreign standards. Any failure by our third-party
manufacturers to comply with cGMP or failure to scale up
manufacturing processes, including any failure to deliver
sufficient quantities of product candidates in a timely manner,
could lead to a delay in, or failure to obtain, regulatory
approval of any of our product candidates. In addition, such
failure could be the basis for action by the FDA to withdraw
approvals for product candidates previously granted to us and
for other regulatory action, including recall or seizure, total
or partial suspension of production or injunction.
We received a request from the FDA for additional information
regarding the characterization and qualification of the
manufactured vials of A-623 we intend to use in our PEARL-SC
clinical study. In addition, the FDA has asked for a proposal to
establish comparability of future manufactured A-623 to be
included in clinical studies. Any inability to use A-623 in our
inventory would require manufacture of additional A-623 for use
in our clinical study and would result in additional expense and
potential delay of our clinical development plans.
We rely on our manufacturers to purchase from third-party
suppliers the materials necessary to produce our product
candidates for our clinical studies. There are a small number of
suppliers for certain capital equipment and raw materials that
we use to manufacture our drugs. Such suppliers may not sell
these raw materials to our manufacturers at the times we need
them or on commercially reasonable terms. We do not have any
control over the process or timing of the acquisition of these
raw materials by our manufacturers. Moreover, we currently do
not have any agreements for the commercial production of these
raw materials. Although we generally do not begin a clinical
study unless we believe we have a sufficient supply of a product
candidate to complete the clinical study, any significant delay
in the supply of a product candidate or the raw material
components thereof for an ongoing clinical study due to the need
to replace a third-party manufacturer could considerably delay
completion of our clinical studies, product testing and
potential regulatory approval of our product candidates. If our
manufacturers or we are unable to purchase these raw materials
after regulatory approval has been obtained for our product
candidates, the commercial launch of our product candidates
would be delayed or there would be a shortage in supply, which
would impair our ability to generate revenues from the sale of
our product candidates.
Because of the complex nature of our compounds, our
manufacturers may not be able to manufacture our compounds at a
cost or in quantities or in a timely manner necessary to make
commercially
25
successful products. If we successfully commercialize any of our
drugs, we may be required to establish large-scale commercial
manufacturing capabilities. In addition, as our drug development
pipeline increases and matures, we will have a greater need for
clinical study and commercial manufacturing capacity. We have no
experience manufacturing pharmaceutical products on a commercial
scale and some of these suppliers will need to increase their
scale of production to meet our projected needs for commercial
manufacturing, the satisfaction of which on a timely basis may
not be met.
If we are unable to establish sales and marketing
capabilities or enter into agreements with third parties to
market and sell our product candidates, we may be unable to
generate any revenue.
We do not currently have an organization for the sales,
marketing and distribution of pharmaceutical products and the
cost of establishing and maintaining such an organization may
exceed the cost-effectiveness of doing so. In order to market
any products that may be approved by the FDA, we must build our
sales, marketing, managerial and other non-technical
capabilities or make arrangements with third parties to perform
these services. If we are unable to establish adequate sales,
marketing and distribution capabilities, whether independently
or with third parties, we may not be able to generate product
revenue and may not become profitable. We will be competing with
many companies that currently have extensive and well-funded
marketing and sales operations. Without an internal team or the
support of a third party to perform marketing and sales
functions, we may be unable to compete successfully against
these more established companies.
Guidelines and recommendations published by various
organizations may adversely affect the use of any products for
which we may receive regulatory approval.
Government agencies issue regulations and guidelines directly
applicable to us and to our product candidates. In addition,
professional societies, practice management groups, private
health or science foundations and organizations involved in
various diseases from time to time publish guidelines or
recommendations to the medical and patient communities. These
various sorts of recommendations may relate to such matters as
product usage and use of related or competing therapies. For
example, organizations like the American Heart Association have
made recommendations about therapies in the cardiovascular
therapeutics market. Changes to these recommendations or other
guidelines advocating alternative therapies could result in
decreased use of any products for which we may receive
regulatory approval, which may adversely affect our results of
operations.
Risks
Related to Our Intellectual Property
If our or our licensors patent positions do not
adequately protect our product candidates or any future
products, others could compete with us more directly, which
would harm our business.
As of the date of this prospectus and as described in the
section entitled Business Intellectual
Property on page 92, we hold a total of four pending
U.S. non-provisional patent applications, three pending
U.S. provisional patent applications and two pending Patent
Cooperation Treaty, or PCT, patent applications. Another PCT
application has entered the national phase in the European
Patent Office, the Eurasian Patent Organization and 17 other
countries. We have also entered into license agreements for
certain composition of matter, method of use and method of
making patents and patent applications for certain of our
development compounds. These license agreements encompass
(i) 13 U.S. patents, one pending
U.S. non-provisional patent application, five European, or
EP, patents, one pending EP patent application, 18 non-EP
foreign patents and four pending non-EP foreign patent
applications relating to varespladib and
A-001;
(ii) more than 30 U.S. patents, one pending
U.S. non-provisional patent application, six EP patents,
one pending EP patent application, 13 issued non-EP foreign
patents and two pending non-EP foreign patent applications
relating to other
sPLA2
inhibiting compounds including
A-003; and
(iii) two U.S. patents, one pending
U.S. non-provisional patent
26
application, one EP patent, two pending EP patent applications,
ten non-EP foreign patents and 14 non-EP foreign patent
applications relating to
A-623. Our
commercial success will depend in part on our and our
licensors ability to obtain additional patents and protect
our existing patent positions, particularly those patents for
which we have secured exclusive rights, as well as our ability
to maintain adequate protection of other intellectual property
for our technologies, product candidates and any future products
in the United States and other countries. If we or our licensors
do not adequately protect our intellectual property, competitors
may be able to use our technologies and erode or negate any
competitive advantage we may have, which could materially harm
our business, negatively affect our position in the marketplace,
limit our ability to commercialize our product candidates and
delay or render impossible our achievement of profitability. The
laws of some foreign countries do not protect our proprietary
rights to the same extent as the laws of the United States, and
we may encounter significant problems in protecting our
proprietary rights in these countries.
The patent positions of biotechnology and pharmaceutical
companies, including our patent position, involve complex legal
and factual questions, and, therefore, validity and
enforceability cannot be predicted with certainty. Patents may
be challenged, deemed unenforceable, invalidated or
circumvented. We and our licensors will be able to protect our
proprietary rights from unauthorized use by third parties only
to the extent that our proprietary technologies, product
candidates and any future products are covered by valid and
enforceable patents or are effectively maintained as trade
secrets.
The degree of future protection for our proprietary rights is
uncertain, and we cannot ensure that:
|
|
|
|
|
we or our licensors were the first to make the inventions
covered by each of our pending patent applications;
|
|
|
|
we or our licensors were the first to file patent applications
for these inventions;
|
|
|
|
others will not independently develop similar or alternative
technologies or duplicate any of our technologies;
|
|
|
|
any of our or our licensors pending patent applications
will result in issued patents;
|
|
|
|
any of our or our licensors patents will be valid or
enforceable;
|
|
|
|
any patents issued to us or our licensors and collaborators will
provide a basis for commercially viable products, will provide
us with any competitive advantages or will not be challenged by
third parties;
|
|
|
|
we will develop additional proprietary technologies or product
candidates that are patentable; or
|
|
|
|
the patents of others will not have an adverse effect on our
business.
|
We may be unable to adequately prevent disclosure of trade
secrets and other proprietary information.
We rely on trade secrets to protect our proprietary know-how and
technological advances, especially where we do not believe
patent protection is appropriate or obtainable. However, trade
secrets are difficult to protect. We rely in part on
confidentiality agreements with our employees, consultants,
outside scientific collaborators, sponsored researchers and
other advisors to protect our trade secrets and other
proprietary information. These agreements may not effectively
prevent disclosure of confidential information and may not
provide an adequate remedy in the event of unauthorized
disclosure of confidential information. In addition, others may
independently discover our trade secrets and
27
proprietary information. Costly and time-consuming litigation
could be necessary to enforce and determine the scope of our
proprietary rights. Failure to obtain or maintain trade secret
protection could enable competitors to use our proprietary
information to develop products that compete with our products
or cause additional, material adverse effects upon our
competitive business position.
We license patent rights from third-party owners. If we,
or such owners, do not properly maintain or enforce the patents
underlying such licenses, our competitive position and business
prospects will be harmed.
We have obtained exclusive, worldwide licenses, except for
Japan, of the composition of matter, methods of making and
methods of use for certain
sPLA2
compounds from Eli Lilly and Shionogi & Co., Ltd. In
addition, we are party to a license agreement with Amgen for the
exclusive and worldwide rights to develop and commercialize
A-623, a
novel BAFF inhibitor. We may enter into additional licenses to
third-party intellectual property in the future.
We depend in part on our licensors to protect the proprietary
rights covering our in-licensed
sPLA2
compounds and
A-623,
respectively. Our licensors are responsible for maintaining
certain issued patents and prosecuting certain patent
applications. We have limited, if any, control over the amount
or timing of resources that our licensors devote on our behalf
or the priority they place on maintaining these patent rights
and prosecuting these patent applications to our advantage. Our
licensors may also be notified of alleged infringement and be
sued for infringement of third-party patents or other
proprietary rights. We may have limited, if any, control or
involvement over the defense of these claims, and our licensors
could be subject to injunctions and temporary or permanent
exclusionary orders in the United States or other countries. Our
licensors are not obligated to defend or assist in our defense
against third-party claims of infringement. We have limited, if
any, control over the amount or timing of resources, if any,
that our licensors devote on our behalf or the priority they
place on defense of such third-party claims of infringement.
Our success will depend in part on the ability of us or our
licensors to obtain, maintain and enforce patent protection for
their intellectual property, in particular, those patents to
which we have secured exclusive rights. We or our licensors may
not successfully prosecute the patent applications which we have
licensed. Even if patents issue in respect of these patent
applications, we or our licensors may fail to maintain these
patents, may determine not to pursue litigation against other
companies that are infringing these patents or may pursue such
litigation less aggressively than we would. Without protection
for the intellectual property we license, other companies might
be able to offer substantially identical products for sale,
which could adversely affect our competitive business position
and harm our business prospects.
If we do not obtain protection under the Hatch-Waxman Act
and similar foreign legislation to extend our licensed patent
terms and to obtain market exclusivity for our product
candidates, our business will be materially harmed.
The United States Drug Price Competition and Patent Term
Restoration Act of 1984, more commonly known as the
Hatch-Waxman Act, provides for an extension of
patent terms for drug compounds for a period of up to five years
to compensate for time spent in development. Assuming we gain a
five-year patent term extension for each of our current product
candidates in clinical development, and that we continue to have
rights under our license agreements with respect to these
product candidates, we would have exclusive rights to
varespladibs U.S. new chemical entity
patent (the primary patent covering the compound as a new
composition of matter) until 2019 and to
A-623s
U.S. new chemical entity patent until 2027. In Europe,
similar legislative enactments allow patent terms in the
European Union to be extended for up to five years through the
grant of a Supplementary Protection Certificate. Assuming we
gain such a five-year extension for each of our current product
candidates in clinical
28
development, and that we continue to have rights under our
license agreements with respect to these product candidates, we
would have exclusive rights to varespladibs European new
chemical entity patents until 2020 and to
A-623s
European new chemical entity patents until 2027. In addition,
since varespladib has not been previously approved in the United
States, varespladib could be eligible for up to five years of
New Chemical Entity, or NCE, exclusivity from the FDA. NCE
exclusivity would prevent the FDA from accepting any generic
competition following NDA approval independent of the patent
status of varespladib. Further, since
A-623 has
not been previously approved,
A-623 could
be eligible for 12 years of data exclusivity from the FDA.
During the data exclusivity period, competitors are barred from
relying on the innovator biologics safety and efficacy
data to gain approval. Similarly, the European Union provides
that companies who receive regulatory approval for a new small
molecule compound or biologic will have a
10-year
period of data exclusivity for that compound or biologic (with
the possibility of a further one-year extension) in most EU
countries, beginning on the date of such European regulatory
approval, regardless of when the European new chemical entity
patent covering such compound expires. A generic version of the
approved drug may not be marketed or sold during such market
exclusivity period. However, there is no assurance that we will
receive the extensions of our patents or other exclusive rights
available under the Hatch-Waxman Act or similar foreign
legislation. If we fail to receive such Hatch-Waxman extensions
or marketing exclusivity rights or if we receive extensions that
are materially shorter than expected, our ability to prevent
competitors from manufacturing, marketing and selling generic
versions of our products will be materially harmed.
Our current patent positions and license portfolio may not
include all patent rights needed for the full development and
commercialization of our product candidates. We cannot be sure
that patent rights we may need in the future will be available
for license to us on commercially reasonable terms, or at
all.
We typically develop our product candidates using compounds for
which we have in-licensed and original composition of matter
patents and patents that claim the activities and methods for
such compounds production and use to the extent known at
that time. As we learn more about the mechanisms of action and
new methods of manufacture and use of these product candidates,
we may file additional patent applications for these new
inventions or we may need to ask our licensors to file them. We
may also need to license additional patent rights or other
rights on compounds, treatment methods or manufacturing
processes because we learn that we need such rights during the
continuing development of our product candidates.
Although our in-licensed and original patents may prevent others
from making, using or selling similar products, they do not
ensure that we will not infringe the patent rights of third
parties. We may not be aware of all patents or patent
applications that may impact our ability to make, use or sell
any of our product candidates or proposed product candidates.
For example, because we sometimes identify the mechanism of
action or molecular target of a given product candidate after
identifying its composition of matter and therapeutic use, we
may not be aware until the mechanism or target is further
elucidated that a third party has an issued or pending patent
claiming biological activities or targets that may cover our
product candidate. U.S. patent applications filed after
November 29, 2000 are confidential in the U.S. Patent
and Trademark Office for the first 18 months after such
applications earliest priority date, and patent offices in
non-U.S. countries
often publish patent applications for the first time six months
or more after filing. Furthermore, we may not be aware of
published or granted conflicting patent rights. Any conflicts
resulting from patent applications and patents of others could
significantly reduce the coverage of our patents and limit our
ability to obtain meaningful patent protection. If others obtain
patents with conflicting claims, we may need to obtain licenses
to these patents or to develop or obtain alternative technology.
We may not be able to obtain any licenses or other rights to
patents, technology or know-how from third parties necessary to
conduct our business as described in this prospectus and such
licenses, if
29
available at all, may not be available on commercially
reasonable terms. Any failure to obtain such licenses could
delay or prevent us from developing or commercializing our drug
candidates or proposed product candidates, which would harm our
business. Litigation or patent interference proceedings may be
necessarily brought against third parties, as discussed below,
to enforce any of our patents or other proprietary rights or to
determine the scope and validity or enforceability of the
proprietary rights of such third parties.
Litigation regarding patents, patent applications and
other proprietary rights may be expensive and time consuming. If
we are involved in such litigation, it could cause delays in
bringing product candidates to market and harm our ability to
operate.
Our commercial success will depend in part on our ability to
manufacture, use, sell and offer to sell our product candidates
and proposed product candidates without infringing patents or
other proprietary rights of third parties. Although we are not
currently aware of any litigation or other proceedings or
third-party claims of intellectual property infringement related
to our product candidates, the pharmaceutical industry is
characterized by extensive litigation regarding patents and
other intellectual property rights. Other parties may obtain
patents in the future and allege that the use of our
technologies infringes these patent claims or that we are
employing their proprietary technology without authorization.
Likewise, third parties may challenge or infringe upon our or
our licensors existing or future patents.
Proceedings involving our patents or patent applications or
those of others could result in adverse decisions regarding the
patentability of our inventions relating to our product
candidates or the enforceability, validity or scope of
protection offered by our patents relating to our product
candidates.
Even if we are successful in these proceedings, we may incur
substantial costs and divert management time and attention in
pursuing these proceedings. If we are unable to avoid infringing
the patent rights of others, we may be required to seek a
license, defend an infringement action or challenge the validity
of the patents in court. Patent litigation is costly and
time-consuming. We may not have sufficient resources to bring
these actions to a successful conclusion. In addition, if we do
not obtain a license, develop or obtain non-infringing
technology, fail to defend an infringement action successfully
or have infringed patents declared invalid, we may incur
substantial monetary damages; encounter significant delays in
bringing our product candidates to market; or be precluded from
participating in the manufacture, use or sale of our product
candidates or methods of treatment requiring licenses.
Risks
Related to the Securities Markets and Investment in Our Common
Stock
Market volatility may affect our stock price and the value
of your investment.
The market price for our common stock has been, and is likely to
continue to be, volatile. In addition, the market price of our
common stock may fluctuate significantly in response to a number
of factors, most of which we cannot predict or control,
including:
|
|
|
|
|
plans for, progress in and results from clinical studies for
varespladib,
A-623,
A-001 and
our other product candidates;
|
|
|
|
announcements of new products, services or technologies,
commercial relationships, acquisitions or other events by us or
our competitors;
|
30
|
|
|
|
|
developments concerning proprietary rights, including those
pertaining to patents held by Eli Lilly and Shionogi &
Co., Ltd. concerning our
sPLA2
inhibitors and Amgen concerning
A-623;
|
|
|
|
failure of any of our product candidates, if approved, to
achieve commercial success;
|
|
|
|
fluctuations in stock market prices and trading volumes of
securities of similar companies;
|
|
|
|
general market conditions and overall fluctuations in
U.S. equity markets;
|
|
|
|
variations in our operating results, or the operating results of
our competitors;
|
|
|
|
changes in our financial guidance or securities analysts
estimates of our financial performance;
|
|
|
|
changes in accounting principles;
|
|
|
|
sales of large blocks of our common stock, including sales by
our executive officers, directors and significant stockholders;
|
|
|
|
additions or departures of any of our key personnel;
|
|
|
|
announcements related to litigation;
|
|
|
|
changing legal or regulatory developments in the United States
and other countries; and
|
|
|
|
discussion of us or our stock price by the financial press and
in online investor communities.
|
In addition, the stock market in general, and The NASDAQ Global
Market in particular, have experienced substantial price and
volume volatility that is often seemingly unrelated to the
operating performance of particular companies. These broad
market fluctuations may cause the trading price of our common
stock to decline. In the past, securities class action
litigation has often been brought against a company after a
period of volatility in the market price of its common stock. We
may become involved in this type of litigation in the future.
Any securities litigation claims brought against us could result
in substantial expenses and the diversion of our
managements attention from our business.
Because a small number of our existing stockholders own a
majority of our voting stock, your ability to influence
corporate matters will be limited.
Our executive officers, directors and greater than 5%
stockholders, in the aggregate, own approximately 79% of our
outstanding common stock. As a result, such persons, acting
together, will have the ability to control our management and
affairs and substantially all matters submitted to our
stockholders for approval, including the election and removal of
directors and approval of any significant transaction. These
persons will also have the ability to control our management and
business affairs. This concentration of ownership may have the
effect of delaying, deferring or preventing a change in control,
impeding a merger, consolidation, takeover or other business
combination involving us, or discouraging a potential acquirer
from making a tender offer or otherwise attempting to obtain
control of our business, even if such a transaction would
benefit other stockholders.
31
Future sales of our common stock may cause our stock price
to decline.
As of September 30, 2010, there were 32,835,437 shares of
our common stock outstanding. Of these, 16,017,349 shares
of common stock and 4,363,200 shares of common stock
underlying certain warrants are being sold in this offering by
the selling stockholders and will be freely tradable immediately
after this offering (assuming exercise of the warrants and
except for shares purchased by affiliates) and 1,897,728 of the
32,835,437 shares may be sold upon expiration of
lock-up
agreements 30 days after the effective date of this
registration statement, of which this prospectus forms a part
(subject in some cases to volume limitations). In addition, as
of June 30, 2010, we had outstanding options to purchase
1,239,129 shares of common stock that, if exercised, will
result in these additional shares becoming available for sale. A
large portion of these shares and options are held by a small
number of persons and investment funds. Sales by these
stockholders or optionholders of a substantial number of shares
after this offering could significantly reduce the market price
of our common stock. Moreover, certain holders of shares of
common stock will have rights, subject to some conditions, to
require us to file registration statements covering the shares
they currently hold, or to include these shares in registration
statements that we may file for ourselves or other stockholders.
We have registered all common stock that we may issue under our
Amended and Restated 2010 Stock Option and Incentive Plan, or
the 2010 Plan, and our Employee Stock Purchase Plan, or the
ESPP. An aggregate of 433,644 shares of our common stock
has been reserved for future issuance under the 2010 Plan, plus
any shares reserved and unissued under our 2005 Equity Incentive
Plan, and an aggregate of 100,000 shares has been reserved
for future issuance under our ESPP. These shares can be freely
sold in the public market upon issuance, subject to the
lock-up
agreements referred to above. If a large number of these shares
are sold in the public market, the sales could reduce the
trading price of our common stock. See the section entitled
Shares Eligible for Future Sale on
page 156 for a more detailed description of sales that may
occur in the future.
We may need to raise additional capital to fund our
operations, which may cause dilution to our existing
stockholders, restrict our operations or require us to
relinquish rights.
We may seek additional capital through a combination of private
and public equity offerings, debt financings and collaboration,
strategic and licensing arrangements. To the extent that we
raise additional capital through the sale of equity or
convertible debt securities, your ownership interest will be
diluted, and the terms may include liquidation or other
preferences that adversely affect your rights as a stockholder.
Debt financing, if available, may involve agreements that
include covenants limiting or restricting our ability to take
specific actions such as incurring debt, making capital
expenditures or declaring dividends. If we raise additional
funds through collaboration, strategic alliance and licensing
arrangements with third parties, we may have to relinquish
valuable rights to our technologies or product candidates or
grant licenses on terms that are not favorable to us.
Being a public company increases our expenses and
administrative burden.
As a public company, we incur significant legal, accounting and
other expenses that we did not incur as a private company. In
addition, our administrative staff will be required to perform
additional tasks. For example, the Sarbanes-Oxley Act of 2002,
or the Sarbanes-Oxley Act, as well as rules subsequently
implemented by the Securities and Exchange Commission, or SEC,
and The NASDAQ Global Market, impose various requirements on
public companies, including establishment and maintenance of
effective disclosure and financial controls and changes in
corporate governance practices. We must also bear all of the
internal and external costs of preparing and distributing
periodic public reports in compliance with our obligations under
the securities laws.
32
In particular, the Sarbanes-Oxley Act requires, among other
things, that we maintain effective internal control over
financial reporting and disclosure controls and procedures.
Commencing in 2011, we must perform system and process
evaluation and testing of our internal control over financial
reporting to allow management and our independent registered
public accounting firm to report on the effectiveness of our
internal control over financial reporting, as required by
Section 404 of the Sarbanes-Oxley Act. Our compliance with
Section 404 will require that we incur substantial
accounting expense and expend significant management time on
compliance-related issues. Moreover, if we are not able to
comply with the requirements of Section 404 in a timely
manner, our stock price could decline, and we could face
sanctions, delisting or investigations by The NASDAQ Global
Market, or other material adverse effects on our business,
reputation, results of operations, financial condition or
liquidity.
We do not intend to pay dividends on our common stock so
any returns will be limited to the value of our stock.
We have never declared or paid any cash dividend on our common
stock. We currently anticipate that we will retain future
earnings for the development, operation and expansion of our
business and do not anticipate declaring or paying any cash
dividends for the foreseeable future. Any return to stockholders
will therefore be limited to the value of their stock.
Anti-takeover provisions in our charter documents and
under Delaware law could make an acquisition of us, which may be
beneficial to our stockholders, more difficult and may prevent
attempts by our stockholders to replace or remove our current
management.
Provisions in our amended and restated certificate of
incorporation and amended and restated bylaws may delay or
prevent an acquisition of us or a change in our management.
These provisions include:
|
|
|
|
|
a classified and staggered board of directors whose members can
only be dismissed for cause;
|
|
|
|
the prohibition on actions by written consent of our
stockholders;
|
|
|
|
the limitation on who may call a special meeting of stockholders;
|
|
|
|
the establishment of advance notice requirements for nominations
for election to our board of directors or for proposing matters
that can be acted upon at stockholder meetings;
|
|
|
|
the ability of our board of directors to issue preferred stock
without stockholder approval, which would increase the number of
outstanding shares and could thwart a takeover attempt; and
|
|
|
|
the requirement of at least 75% of the outstanding common stock
to amend any of the foregoing provisions.
|
In addition, because we are incorporated in Delaware, we are
governed by the provisions of Section 203 of the Delaware
General Corporation Law, which limits the ability of
stockholders owning in excess of 15% of our outstanding voting
stock to merge or combine with us. Although we believe these
provisions collectively provide for an opportunity to obtain
greater value for stockholders by requiring potential acquirors
to negotiate with our board of directors, they would apply even
if an offer rejected by our board were considered beneficial by
some stockholders. In addition, these provisions may frustrate
or prevent any attempts by our stockholders to replace or remove
our current management by making it more difficult for
stockholders to replace members of our board of directors, which
is responsible for appointing the members of our management.
33
Our ability to use our net operating loss carryforwards
may be subject to limitation and may result in increased future
tax liability to us.
Generally, a change of more than 50% in the ownership of a
corporations stock, by value, over a three-year period
constitutes an ownership change for U.S. federal income tax
purposes. An ownership change may limit a companys ability
to use its net operating loss carryforwards attributable to the
period prior to such change. We have not performed a detailed
analysis to determine whether an ownership change under
Section 382 of the Internal Revenue Code has occurred after
each of our previous private placements of preferred stock and
convertible debt, or our previous issuances of common stock,
which if sufficient, taking into account prior or future shifts
in our ownership over a three-year period, could cause us to
undergo an ownership change. As a result, if we earn net taxable
income, our ability to use our pre-change net operating loss
carryforwards to offset U.S. federal taxable income may
become subject to limitations, which could potentially result in
increased future tax liability to us.
34
SPECIAL
NOTE REGARDING FORWARD-LOOKING STATEMENTS
This prospectus contains forward-looking statements.
Forward-looking statements relate to future events or our future
financial performance. We generally identify forward-looking
statements by terminology such as may,
will, would, should,
expects, plans, anticipates,
could, intends, target,
projects, contemplates,
believes, estimates,
predicts, assume, intend,
potential, continue or other similar
words or the negative of these terms. These statements are only
predictions. We have based these forward-looking statements
largely on our current expectations and projections about future
events and financial trends that we believe may affect our
business, financial condition and results of operations. The
outcome of the events described in these forward-looking
statements is subject to risks, uncertainties and other factors
described in Risk Factors and elsewhere in this
prospectus. Accordingly, you should not place undue reliance
upon these forward-looking statements. We cannot assure you that
the events and circumstances reflected in the forward-looking
statements will be achieved or occur, the timing of events and
circumstances and actual results could differ materially from
those projected in the forward looking statements.
Forward-looking statements contained in this prospectus include,
but are not limited to, statements about:
|
|
|
|
|
our expectations related to the use of proceeds, if any, from
this offering;
|
|
|
|
|
|
the progress of, timing of and amount of expenses associated
with our research, development and commercialization activities;
|
|
|
|
the timing, conduct and success of our clinical studies for our
product candidates;
|
|
|
|
our ability to obtain U.S. and foreign regulatory approval
for our product candidates and the ability of our product
candidates to meet existing or future regulatory standards;
|
|
|
|
our expectations regarding federal, state and foreign regulatory
requirements;
|
|
|
|
the therapeutic benefits and effectiveness of our product
candidates;
|
|
|
|
the accuracy of our estimates of the size and characteristics of
the markets that may be addressed by our product candidates;
|
|
|
|
our ability to manufacture sufficient amounts of our product
candidates for clinical studies and products for
commercialization activities;
|
|
|
|
our intention to seek to establish strategic collaborations or
partnerships for the development or sale of our product
candidates;
|
|
|
|
our expectations as to future financial performance, expense
levels and liquidity sources;
|
|
|
|
the timing of commercializing our product candidates;
|
|
|
|
our ability to compete with other companies that are or may be
developing or selling products that are competitive with our
product candidates;
|
|
|
|
anticipated trends and challenges in our potential markets;
|
|
|
|
our ability to attract and retain key personnel; and
|
|
|
|
other factors discussed elsewhere in this prospectus.
|
35
The forward-looking statements made in this prospectus relate
only to events as of the date on which the statements are made.
We have included important factors in the cautionary statements
included in this prospectus, particularly in the section
entitled Risk Factors that we believe could cause
actual results or events to differ materially from the
forward-looking statements that we make. Our forward-looking
statements do not reflect the potential impact of any future
acquisitions, mergers, dispositions, joint ventures or
investments we may make. Except as required by law, we do not
assume any intent to update any forward-looking statements after
the date on which the statement is made, whether as a result of
new information, future events or circumstances or otherwise.
36
USE OF
PROCEEDS
The proceeds from the resale of the shares of common stock under
this prospectus are solely for the account of the selling
stockholders identified in this prospectus. We may indirectly
receive proceeds of up to an aggregate of $15,002,400 to the
extent that any selling stockholders exercise Warrants to
purchase shares of common stock for cash, which shares may then
be resold under this prospectus; however, we will not directly
receive any proceeds from the sale of shares under this
prospectus. We intend to use the net proceeds generated by
Warrant exercises, if any, for general corporate purposes. We
cannot estimate how many, if any, of the Warrants will be
exercised as a result of this offering.
37
PRICE
RANGE OF COMMON STOCK
Our common stock has been listed on The NASDAQ Global Market
under the symbol ANTH since our initial public
offering. Prior to that offering, there was no public market for
our common stock. The following table sets forth, for the
periods indicated, the high and low intraday sales prices of our
common stock as reported by The NASDAQ Global Market:
|
|
|
|
|
|
|
|
|
|
|
High
|
|
|
Low
|
|
|
First Quarter 2010 (beginning March 2, 2010)
|
|
$
|
7.19
|
|
|
$
|
6.88
|
|
Second Quarter 2010
|
|
$
|
8.55
|
|
|
$
|
5.07
|
|
Third Quarter 2010
|
|
$
|
5.99
|
|
|
$
|
2.82
|
|
Fourth Quarter 2010 (through October 21, 2010)
|
|
$
|
6.84
|
|
|
$
|
4.12
|
|
On October 21, 2010, the closing price as reported on The
NASDAQ Global Market of our common stock was $6.68. As of
October 15, 2010, we had approximately 750 holders of
record and beneficial holders of our common stock.
38
DIVIDEND
POLICY
We have never declared or paid any cash dividends on our common
stock. We currently intend to retain any future earnings to
finance the growth and development of our business. Therefore,
we do not anticipate declaring or paying any cash dividends in
the foreseeable future. Any future determination as to the
declaration and payment of dividends, if any, will be at the
discretion of our board of directors and will depend on then
existing conditions, including our financial condition,
operating results, contractual restrictions, capital
requirements, business prospects and other factors our board of
directors may deem relevant.
39
CAPITALIZATION
The following table sets forth our capitalization as of
June 30, 2010:
|
|
|
|
|
on an actual basis; and
|
|
|
|
|
|
on an as adjusted basis to give effect to the receipt by us of
net proceeds of $29.1 million from the sale of
10,500,000 units, with each unit consisting of one share of
common stock and a Warrant to purchase 0.40 shares of
common stock, to the selling stockholders pursuant to the
private placement transaction that closed on September 24,
2010.
|
The following table does not reflect any exercise by the selling
stockholders of the Warrants.
You should read the following table in conjunction with our
financial statements and related notes, Selected Financial
Data and Managements Discussion and Analysis
of Financial Condition and Results of Operations appearing
elsewhere in this prospectus.
|
|
|
|
|
|
|
|
|
|
|
As of June 30, 2010
|
|
|
|
|
|
|
As
|
|
|
|
Actual
|
|
|
Adjusted
|
|
|
Cash, cash equivalents and short-term investments
|
|
$
|
51,038,284
|
|
|
$
|
80,115,320
|
|
|
|
|
|
|
|
|
|
|
Common stock, $0.001 par value, 95,000,000 shares
authorized; 22,266,055 shares issued and outstanding,
actual; and 32,766,055 shares outstanding, as adjusted
|
|
|
22,266
|
|
|
|
32,766
|
|
Preferred stock, $0.001 par value, 5,000,000 shares
authorized; 0 shares issued and outstanding, actual and as
adjusted
|
|
|
|
|
|
|
|
|
Additional paid-in capital
|
|
|
133,107,495
|
|
|
|
162,174,031
|
|
Other comprehensive (loss)
|
|
|
(31,734
|
)
|
|
|
(31,734
|
)
|
Deficit accumulated during the development stage
|
|
|
(84,270,107
|
)
|
|
|
(84,270,107
|
)
|
|
|
|
|
|
|
|
|
|
Total stockholders equity
|
|
|
48,827,920
|
|
|
|
77,904,956
|
|
|
|
|
|
|
|
|
|
|
Total capitalization
|
|
$
|
48,827,920
|
|
|
$
|
77,904,956
|
|
|
|
|
|
|
|
|
|
|
40
SELECTED
FINANCIAL DATA
The following selected financial data should be read together
with our financial statements and the related notes and
Managements Discussion and Analysis of Financial
Condition and Results of Operations appearing elsewhere in
this prospectus. The selected financial data in this section is
not intended to replace our financial statements and the related
notes. Our historical results are not necessarily indicative of
the results to be expected for any future period.
We were incorporated on September 9, 2004. The following
statement of operations data, including share data, for the
years ended December 31, 2007, 2008 and 2009 and for the
cumulative period from September 9, 2004 to
December 31, 2009, and the balance sheet data as of
December 31, 2008 and 2009 have been derived from our
audited financial statements and related notes appearing
elsewhere in this prospectus. The statement of operations data
for the years ended December 31, 2005 and 2006 and the
balance sheet data as of December 31, 2005, 2006 and 2007
have been derived from our audited financial statements not
included in this prospectus. The statement of operations data,
including share data, for the six months ended June 30,
2009 and 2010, and the period from September 9, 2004 (date
of inception) through June 30, 2010, and the balance sheet
data as of June 30, 2010 have been derived from our
unaudited interim financial statements appearing elsewhere in
this prospectus. The unaudited interim financial statements have
been prepared on the same basis as the audited financial
statements and reflect all adjustments necessary to fairly state
our financial position as of June 30, 2010 and the results
of operations for the six months ended June 30, 2009 and
2010, and for the cumulative period from September 9, 2004
to June 30, 2010. The operating results for any period are
not necessarily indicative of financial results that may be
expected for any future period.
The pro forma basic and diluted net loss per share and pro forma
weighted-average number of shares gives effect to the conversion
of all our outstanding preferred stock into shares of common
stock as if the conversion occurred on the date of issuance.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Period from
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
September 9
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
2004 (Date of
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Six Months Ended
|
|
|
Inception)
|
|
|
|
Year Ended December 31,
|
|
|
June 30,
|
|
|
To June 30,
|
|
|
|
2005
|
|
|
2006
|
|
|
2007
|
|
|
2008
|
|
|
2009
|
|
|
2009
|
|
|
2010
|
|
|
2009
|
|
|
Statement of Operations Data:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Operating expenses
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Research and development
|
|
$
|
345,208
|
|
|
$
|
7,759,106
|
|
|
$
|
23,921,932
|
|
|
$
|
10,882,322
|
|
|
$
|
8,415,414
|
|
|
$
|
5,201,181
|
|
|
$
|
11,679,963
|
|
|
$
|
63,003,944
|
|
General and administrative
|
|
|
205,527
|
|
|
|
822,732
|
|
|
|
2,468,607
|
|
|
|
2,980,170
|
|
|
|
3,425,690
|
|
|
|
1,845,574
|
|
|
|
2,733,979
|
|
|
|
12,651,546
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total operating expenses
|
|
|
(550,735
|
)
|
|
|
(8,581,838
|
)
|
|
|
(26,390,539
|
)
|
|
|
(13,862,492
|
)
|
|
|
(11,841,104
|
)
|
|
|
(7,046,755
|
)
|
|
|
(14,413,942
|
)
|
|
|
(75,655,490
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Other Income (Expense)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Interest and other income
|
|
|
11,148
|
|
|
|
109,987
|
|
|
|
696,962
|
|
|
|
178,129
|
|
|
|
23,534
|
|
|
|
21,637
|
|
|
|
14,956
|
|
|
|
1,034,716
|
|
Interest and other expense
|
|
|
|
|
|
|
(17,395
|
)
|
|
|
|
|
|
|
(296,303
|
)
|
|
|
(385,922
|
)
|
|
|
(96,298
|
)
|
|
|
(4,641,169
|
)
|
|
|
(5,340,789
|
)
|
Beneficial conversion feature
|
|
|
|
|
|
|
(190,000
|
)
|
|
|
|
|
|
|
(4,118,544
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(4,308,544
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total other income (expense)
|
|
|
11,148
|
|
|
|
(97,408
|
)
|
|
|
696,962
|
|
|
|
(4,236,718
|
)
|
|
|
(362,388
|
)
|
|
|
(74,661
|
)
|
|
|
(4,626,213
|
)
|
|
|
(8,614,617
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss
|
|
$
|
(539,587
|
)
|
|
$
|
(8,679,246
|
)
|
|
$
|
(25,693,577
|
)
|
|
$
|
(18,099,210
|
)
|
|
$
|
(12,203,492
|
)
|
|
$
|
(7,121,416
|
)
|
|
$
|
(19,040,155
|
)
|
|
$
|
(84,270,107
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss per share basic and
diluted(1)
|
|
$
|
(1.38
|
)
|
|
$
|
(13.82
|
)
|
|
$
|
(28.15
|
)
|
|
$
|
(13.47
|
)
|
|
$
|
(8.06
|
)
|
|
$
|
(4.80
|
)
|
|
$
|
(1.07
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Weighted-average number of shares used in per share
calculation basic and
diluted(2)
|
|
|
390,279
|
|
|
|
627,904
|
|
|
|
912,668
|
|
|
|
1,343,420
|
|
|
|
1,513,598
|
|
|
|
1,483,524
|
|
|
|
17,843,335
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Pro forma net loss per share basic and
diluted(1)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
$
|
(1.24
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Pro forma weighted-average number of shares used in per share
calculation basic and
diluted(2)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
9,854,380
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
footnotes on following page
41
|
|
(1)
|
Diluted earnings per share, or EPS, is identical to basic EPS
since common equivalent shares are excluded from the
calculation, as their effect is anti-dilutive.
|
(2)
|
For accounting purposes only, the number of issued and
outstanding shares for the years ended December 31, 2005,
2006, 2007, 2008 and 2009 and the six months ended June 30,
2009 and 2010 do not include weighted-average shares of unvested
stock of 478,799, 297,596, 261,649, 230,028, 110,079, 133,010
and 53,435, respectively. These shares are subject to a risk of
repurchase by us until such shares are vested. See Note 8
to our financial statements for more information.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
As of December 31,
|
|
|
As of June 30,
|
|
|
|
2005
|
|
|
2006
|
|
|
2007
|
|
|
2008
|
|
|
2009
|
|
|
2010
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(unaudited)
|
|
|
Balance Sheet Data:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Cash and cash equivalents
|
|
$
|
381,964
|
|
|
$
|
20,781,916
|
|
|
$
|
152,744
|
|
|
$
|
7,895,113
|
|
|
$
|
3,803,384
|
|
|
$
|
35,787,481
|
|
Short-term investments
|
|
|
|
|
|
|
|
|
|
|
5,825,000
|
|
|
|
|
|
|
|
|
|
|
|
15,250,803
|
|
Working capital
|
|
|
232,136
|
|
|
|
19,629,639
|
|
|
|
(2,907,995
|
)
|
|
|
(495,836
|
)
|
|
|
(14,344,436
|
)
|
|
|
48,800,562
|
|
Total assets
|
|
|
404,091
|
|
|
|
20,856,892
|
|
|
|
6,193,213
|
|
|
|
8,034,154
|
|
|
|
5,888,789
|
|
|
|
52,058,781
|
|
Indebtedness
|
|
|
150,790
|
|
|
|
1,174,621
|
|
|
|
12,058,184
|
|
|
|
8,494,417
|
|
|
|
18,167,645
|
|
|
|
3,230,861
|
|
Convertible preferred stock
|
|
|
804,951
|
|
|
|
28,892,004
|
|
|
|
28,892,004
|
|
|
|
52,123,859
|
|
|
|
52,123,859
|
|
|
|
|
|
Deficit accumulated during the development stage
|
|
|
(554,427
|
)
|
|
|
(9,233,673
|
)
|
|
|
(34,927,250
|
)
|
|
|
(53,026,460
|
)
|
|
|
(65,229,952
|
)
|
|
|
(84,270,107
|
)
|
Total stockholders (deficit) equity
|
|
|
253,301
|
|
|
|
19,682,271
|
|
|
|
(5,864,971
|
)
|
|
|
(460,263
|
)
|
|
|
(12,278,856
|
)
|
|
|
48,827,920
|
|
42
SELLING
SECURITY HOLDERS
On September 24, 2010, we closed a private placement
transaction with certain accredited investors pursuant to which
we sold an aggregate of 10,500,000 units at a purchase
price of $3.00 per unit, with each unit consisting of one share
of common stock and a Warrant to purchase an additional
0.40 shares of common stock. Each Warrant is exercisable in
whole or in part at any time until September 24, 2015 at a
per share exercise price of $3.30, subject to certain
adjustments as specified in the Warrant.
In connection with the private placement, we entered into a
registration rights agreement, pursuant to which we agreed to
register the resale of the shares of common stock and the common
stock underlying the Warrants. This prospectus covers the resale
of such shares, as well as 5,517,349 shares of common stock
and 163,200 shares of common stock issuable upon
outstanding warrants held by existing stockholders who have
piggyback registration rights, by the selling
stockholders and their trustees, pledges, doners or successors.
The following table sets forth certain information regarding the
selling stockholders and the shares of common stock beneficially
owned by them and issuable to the selling stockholders upon a
cash exercise of the warrants, which information is available to
us as of October 21, 2010. The selling stockholders may
offer the shares under this prospectus from time to time and may
elect to sell some, all or none of the shares set forth next to
their name. As a result, we cannot estimate the number of shares
of common stock that a selling stockholder will beneficially own
after termination of sales under this prospectus. However, for
the purposes of the table below, we have assumed that, after
completion of the offering, none of the shares covered by this
prospectus will be held by the selling stockholders. In
addition, a selling stockholder may have sold, transferred or
otherwise disposed of all or a portion of that holders
shares of common stock since the date on which they provided
information for this table. We have not made independent
inquiries about this. We are relying on written commitments from
the selling stockholders to notify us of any changes in their
beneficial ownership after the date they originally provided
this information. See section entitled Plan of
Distribution beginning on page 46.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
# of Shares
|
|
|
|
|
|
# of Shares
|
|
|
# of Shares
|
|
|
% of Shares
|
|
|
|
Beneficially
|
|
|
# of
|
|
|
Underlying
|
|
|
Beneficially
|
|
|
Beneficially
|
|
|
|
Owned Before
|
|
|
Shares
|
|
|
Warrants
|
|
|
Owned After
|
|
|
Owned After
|
|
Selling Stockholder
|
|
Offering(1)
|
|
|
Offered
|
|
|
Offered
|
|
|
Offering(1)
|
|
|
Offering(1)
|
|
|
Caduceus Private Investments IV,
LP(2)
|
|
|
4,783,068
|
|
|
|
3,333,334
|
|
|
|
1,333,334
|
|
|
|
116,400
|
|
|
|
|
*
|
Visium Balanced Master Fund,
Ltd.(3)
|
|
|
1,750,000
|
|
|
|
1,250,000
|
|
|
|
500,000
|
|
|
|
|
|
|
|
|
|
MPM Bioventures IV-QP,
L.P.(4)
|
|
|
1,350,539
|
|
|
|
937,241
|
|
|
|
374,897
|
|
|
|
38,401
|
|
|
|
|
*
|
MPM Bioventures IV GMBH & Co. Beteiligungs
KG(4)
|
|
|
52,031
|
|
|
|
36,108
|
|
|
|
14,443
|
|
|
|
1,480
|
|
|
|
|
*
|
MPM Asset Management Investors BV4
LLC(4)
|
|
|
38,402
|
|
|
|
26,651
|
|
|
|
10,660
|
|
|
|
1,091
|
|
|
|
|
*
|
Camber Capital Master Fund,
L.P.(5)
|
|
|
1,274,980
|
|
|
|
910,700
|
|
|
|
364,280
|
|
|
|
|
|
|
|
|
|
Camber Capital Fund II,
L.P.(5)
|
|
|
34,440
|
|
|
|
24,600
|
|
|
|
9,840
|
|
|
|
|
|
|
|
|
|
Arthur J. Remillard Jr.
Trust(5)
|
|
|
90,580
|
|
|
|
64,700
|
|
|
|
25,880
|
|
|
|
|
|
|
|
|
|
Redmile Capital Fund,
L.P.(6)
|
|
|
539,067
|
|
|
|
306,600
|
|
|
|
122,640
|
|
|
|
109,827
|
|
|
|
|
*
|
Redmile Capital Offshore Fund,
Ltd.(6)
|
|
|
467,561
|
|
|
|
272,572
|
|
|
|
109,029
|
|
|
|
85,960
|
|
|
|
|
*
|
Redmile Capital Offshore Fund II,
Ltd.(6)
|
|
|
401,052
|
|
|
|
224,161
|
|
|
|
89,664
|
|
|
|
87,227
|
|
|
|
|
*
|
Redmile Ventures,
Ltd.(6)
|
|
|
42,000
|
|
|
|
30,000
|
|
|
|
12,000
|
|
|
|
|
|
|
|
|
|
Baker Brothers Life Sciences,
L.P.(7)
|
|
|
1,089,900
|
|
|
|
778,500
|
|
|
|
311,400
|
|
|
|
|
|
|
|
|
|
14159,
L.P.(7)
|
|
|
30,100
|
|
|
|
21,500
|
|
|
|
8,600
|
|
|
|
|
|
|
|
|
|
HBM BioCapital (USD),
L.P.(8)
|
|
|
298,276
|
(9)
|
|
|
50,000
|
|
|
|
20,000
|
|
|
|
228,276
|
|
|
|
|
*
|
HBM BioCapital (EUR),
L.P.(8)
|
|
|
1,690,241
|
(10)
|
|
|
283,333
|
|
|
|
113,333
|
|
|
|
1,293,575
|
|
|
|
3.94
|
%
|
A.M. Pappas Life Science Ventures III,
L.P.(11)
|
|
|
1,707,032
|
(12)
|
|
|
439,352
|
|
|
|
175,741
|
|
|
|
1,091,939
|
|
|
|
3.32
|
%
|
PV III CEO Fund,
L.P.(11)
|
|
|
106,108
|
(13)
|
|
|
27,315
|
|
|
|
10,926
|
|
|
|
67,867
|
|
|
|
|
*
|
43
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
# of Shares
|
|
|
|
|
|
# of Shares
|
|
|
# of Shares
|
|
|
% of Shares
|
|
|
|
Beneficially
|
|
|
# of
|
|
|
Underlying
|
|
|
Beneficially
|
|
|
Beneficially
|
|
|
|
Owned Before
|
|
|
Shares
|
|
|
Warrants
|
|
|
Owned After
|
|
|
Owned After
|
|
Selling Stockholder
|
|
Offering(1)
|
|
|
Offered
|
|
|
Offered
|
|
|
Offering(1)
|
|
|
Offering(1)
|
|
|
Caxton Advantage Life Sciences Fund,
L.P.(14)
|
|
|
1,560,509
|
|
|
|
250,000
|
|
|
|
100,000
|
|
|
|
1,210,509
|
|
|
|
3.68
|
%
|
New Emerging Medical Opportunities Fund L.P.
|
|
|
876,197
|
|
|
|
166,667
|
|
|
|
66,667
|
|
|
|
642,863
|
|
|
|
1.96
|
%
|
DAFNA LifeScience
Ltd(15)
|
|
|
11,666
|
|
|
|
8,333
|
|
|
|
3,333
|
|
|
|
|
|
|
|
|
|
DAFNA LifeScience Market Neutral
Ltd(15)
|
|
|
8,866
|
|
|
|
6,333
|
|
|
|
2,533
|
|
|
|
|
|
|
|
|
|
DAFNA LifeScience Select
Ltd(15)
|
|
|
26,134
|
|
|
|
18,667
|
|
|
|
7,467
|
|
|
|
|
|
|
|
|
|
Kingsbrook Opportunities Master
Fund LP(16)
|
|
|
46,666
|
|
|
|
33,333
|
|
|
|
13,333
|
|
|
|
|
|
|
|
|
|
Deerfield Special Situations Fund,
L.P.(17)
|
|
|
734,300
|
|
|
|
382,000
|
|
|
|
152,800
|
|
|
|
199,500
|
|
|
|
|
*
|
Deerfield Special Situations Fund International,
Limited(17)
|
|
|
1,190,700
|
|
|
|
618,000
|
|
|
|
247,200
|
|
|
|
325,500
|
|
|
|
|
*
|
VantagePoint Venture Partners IV, L.P. and affiliated
entities(18)
|
|
|
6,466,942
|
|
|
|
5,517,349
|
|
|
|
163,200
|
|
|
|
786,393
|
|
|
|
2.39
|
%
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total
|
|
|
26,667,357
|
|
|
|
16,017,349
|
|
|
|
4,363,200
|
|
|
|
6,286,808
|
|
|
|
19.08
|
%
|
|
|
(1)
|
Beneficial ownership is determined in accordance with the rules
of the SEC and includes voting or investment power with respect
to the shares indicated in the table. Percentage ownership
calculations are based on 32,835,437 shares outstanding as
of September 30, 2010.
|
(2)
|
OrbiMed Advisors LLC, a registered investment adviser under the
Investment Advisers Act of 1940, as amended, is the sole
managing member of OrbiMed Capital GP IV LLC, which is the sole
general partner of Caduceus Private Investments IV, LP. Samuel
D. Isaly is the owner of a controlling interest in OrbiMed
Advisors LLC. As such, OrbiMed Advisors LLC, OrbiMed Capital GP
IV LLC and Mr. Isaly may be deemed to have voting and
investment power with respect to such shares.
|
(3)
|
Jacob Gottlieb, Managing Member of JG Asset, LLC, which is the
General Partner of Visium Asset Management, L.P., which is the
investment managed to pooled investment funds, may be deemed to
have voting and investment power with respect to such shares.
|
(4)
|
Ansbert Gadicke, Luke Evnin, John Vander Vort, Ashley
Dombkowski, William Greene, Vaughn Kailian, Steven St. Peter and
James Scope may be deemed to have voting and investment power
with respect to such shares.
|
(5)
|
Stephen Du Bois may be deemed to have voting and investment
power with respect to such shares.
|
(6)
|
Redmile Group, LLC, investment manager to the selling
stockholder, may be deemed to have voting and investment power
with respect to such shares.
|
(7)
|
Julian C. Baker, Managing Member of Baker Brothers Life
Sciences, L.P. and 14159, L.P., may be deemed to have voting and
investment power with respect to such shares.
|
(8)
|
The board of directors of HBM BioCapital Ltd., the general
partner of HBM BioCapital (USD), L.P. and HBM BioCapital (EUR),
L.P., has sole voting and dispositive power with respect to such
shares. The board of directors of HBM BioCapital Ltd. consists
of John Arnold, Sophia Harris, Richard Coles, Dr. Andreas
Wicki and John Urquhart, none of whom has individual voting or
investment power with respect to the shares.
|
(9)
|
Includes 4,575 shares issuable upon exercise of a common
stock purchase warrant not being offered for resale under this
prospectus.
|
|
|
(10)
|
Includes 25,930 shares issuable upon exercise of a common
stock purchase warrant not being offered for resale under this
prospectus.
|
(11)
|
Arthur M. Pappas, in his role as chairman of the investment
committee of AMP&A Management III, LLC, the general partner
of A.M. Pappas Life Science Ventures III, L.P. and PV III
CEO Fund, L.P., has voting and investment authority over these
shares. Mr. Pappas disclaims beneficial ownership of these
shares except to the extent of his pecuniary interest arising
therein.
|
(12)
|
Includes 25,897 shares issuable upon exercise of a common
stock purchase warrant not being offered for resale under this
prospectus.
|
(13)
|
Includes 1,610 shares issuable upon exercise of a common
stock purchase warrant not being offered for resale under this
prospectus.
|
footnotes continued on following page
44
|
|
(14) |
Includes 30,506 shares issuable upon exercise of a common
stock purchase warrant not being offered for resale under this
prospectus and options to purchase an additional
6,107 shares of common stock that are exercisable within
60 days of September 30, 2010 that are owned of record
by Dr. A. Rachel Leheny over which Caxton Advantage Life
Sciences Fund, L.P. may be deemed to hold voting power. Caxton
Advantage Venture Partners, L.P. has voting and investment power
with respect to such shares. Decisions by Caxton Advantage
Venture Partners, L.P. with respect to such shares are made by
Advantage Life Sciences Partners, LLC, the Managing General
Partner of Caxton Advantage Venture Partners, L.P., together
with the investment committee of Caxton Advantage Venture
Partners, L.P. Dr. Leheny and Eric Roberts have authority
to take action on behalf of Advantage Life Sciences Partners,
LLC as members of Advantage Life Sciences Partners, LLC. The
investment committee of Caxton Advantage Venture Partners, L.P.
as of the date hereof is comprised of (i) Mr. Roberts,
(ii) Dr. Leheny, (iii) Bruce Kovner and
(iv) Peter DAngelo and the consent of four members is
required with respect to any decision by the Investment
Committee. Dr. Leheny is a director of Anthera, is
(i) a Managing Director of Caxton Advantage Venture
Partners, L.P., which is the General Partner of Caxton Advantage
Life Sciences Fund, L.P., a life-sciences venture capital fund
that she co-founded in 2006 and is (ii) a member of
Advantage Life Sciences Partners LLC. Mr. Roberts and
Dr. Leheny and the members of the Caxton Advantage Venture
Partners, L.P. investment committee disclaim beneficial
ownership, except to the extent of their proportionate pecuniary
interests, either directly, or indirectly through Caxton
Advantage Venture Partners, L.P. (or through any other entity
which is a limited partner in Caxton Advantage Life Sciences
Fund, L.P.), in Caxton Advantage Life Sciences Fund, L.P.
|
|
|
(15) |
Nathan Fischel, MD, CFA and Fariba Ghodsian, Ph.D., may be
deemed to have voting and investment power with respect to such
shares.
|
|
|
(16) |
Kingsbrook Partners LP (Kingsbrook Partners) is the
investment manager of Kingsbrook Opportunities Master
Fund LP (Kingsbrook Opportunities) and
consequently has voting and investment discretion over
securities held by Kingsbrook Opportunities. Kingbrook
Opportunities GP LLC (Opportunities GP) is the
general partner of Kingbrook Opportunities and may be considered
the beneficial owner of any securities deemed to be beneficially
owned by Kingsbrook Opportunities. KB GP LLC (GP
LLC) is the general partner of Kingsbrook Partners. Ari J.
Storch, Adam J. Chill and Scott M. Wallace are the sole managing
members of Opportunities GP and GP LLC and as a result may be
considered beneficial owners of any securities deemed
beneficially owned by Opportunities GP and GP LLC. Each of
Kingsbrook Partners, Opportunities GP, GP LLC and
Messrs. Storch, Chill and Wallace disclaim beneficial
ownership of these securities.
|
|
|
(17) |
James Flynn, General Partner of Deerfield Special Situations
Fund, L.P. and Deerfield Special Situations
Fund International, Limited, may be deemed to have voting
and investment power with respect to such shares.
|
|
|
(18) |
Includes (i) 5,695,228 shares of common stock
(4,998,725 shares of which are registered for resale under
this prospectus) and 147,861 shares of common stock
issuable upon exercise of warrants (all of which are registered
for resale under this prospectus), all owned of record by
VantagePoint Venture Partners IV (Q), L.P.,
(ii) 570,147 shares of common stock
(500,421 shares of which are registered for resale under
this prospectus) and 14,801 shares of common stock issuable
upon exercise of warrants (all of which are registered for
resale under this prospectus), all owned of record by
VantagePoint Venture Partners IV, L.P.,
(iii) 20,739 shares of common stock
(18,203 shares of which are registered for resale under
this prospectus) and 538 shares of common stock issuable
upon exercise of warrants (all of which are registered for
resale under this prospectus), all owned of record by
VantagePoint Venture Partners IV Principals Fund, L.P., and
(iv) options to purchase an additional 17,628 shares
of common stock that are exercisable within 60 days of
September 30, 2010 that are owned of record by Annette
Bianchi, over which VantagePoint has sole voting and investment
power. Ms. Bianchi, a director of Anthera, is a Managing
Director at VantagePoint. Alan E. Salzman, through his authority
to cause the general partner of the limited partnerships that
directly hold such shares to act, may be deemed to have voting
and investment power with respect to such shares.
Mr. Salzman disclaims beneficial ownership with respect to
such shares except to the extent of his pecuniary interest
therein.
|
45
PLAN OF
DISTRIBUTION
We are registering an aggregate of 20,380,549 shares of
common stock issued to the selling stockholders and issuable
upon exercise of certain warrants issued to the selling
stockholders to permit the resale of such shares of common stock
by the holders thereof from time to time after the date of this
prospectus. We will not receive any of the proceeds from the
sale by the selling stockholders of the shares of common stock.
We will bear all fees and expenses incident to our obligation to
register the shares of common stock.
The selling stockholders may sell all or a portion of the shares
of common stock beneficially owned by them and offered hereby
from time to time directly or through one or more underwriters,
broker-dealers or agents. If the shares of common stock are sold
through underwriters or broker-dealers, the selling stockholders
will be responsible for underwriting discounts or commissions or
agents commissions. The shares of common stock may be sold
on any national securities exchange or quotation service on
which the securities may be listed or quoted at the time of
sale, in the
over-the-counter
market or in transactions otherwise than on these exchanges or
systems or in the
over-the-counter
market and in one or more transactions at fixed prices, at
prevailing market prices at the time of the sale, at varying
prices determined at the time of sale, or at negotiated prices.
These sales may be effected in transactions, which may involve
crosses or block transactions. The selling stockholders may use
any one or more of the following methods when selling shares:
|
|
|
|
|
ordinary brokerage transactions and transactions in which the
broker-dealer solicits purchasers;
|
|
|
|
|
|
block trades in which the broker-dealer will attempt to sell the
shares as agent but may position and resell a portion of the
block as principal to facilitate the transaction;
|
|
|
|
|
|
purchases by a broker-dealer as principal and resale by the
broker-dealer for its account;
|
|
|
|
|
|
an exchange distribution in accordance with the rules of the
applicable exchange;
|
|
|
|
|
|
privately negotiated transactions;
|
|
|
|
|
|
settlement of short sales entered into after the effective date
of the registration statement of which this prospectus is a part;
|
|
|
|
|
|
broker-dealers may agree with the selling stockholders to sell a
specified number of such shares at a stipulated price per share;
|
|
|
|
|
|
through the writing or settlement of options or other hedging
transactions, whether such options are listed on an options
exchange or otherwise;
|
|
|
|
|
|
a combination of any such methods of sale; and
|
|
|
|
|
|
any other method permitted pursuant to applicable law.
|
The selling stockholders also may resell all or a portion of the
shares in open market transactions in reliance upon
Rule 144 under the Securities Act, as permitted by that
rule, or Section 4(1) under the Securities Act, if
available, rather than under this prospectus, provided that they
meet the criteria and conform to the requirements of those
provisions.
46
Broker-dealers engaged by the selling stockholders may arrange
for other broker-dealers to participate in sales. If the selling
stockholders effect such transactions by selling shares of
common stock to or through underwriters, broker-dealers or
agents, such underwriters, broker-dealers or agents may receive
commissions in the form of discounts, concessions or commissions
from the selling stockholders or commissions from purchasers of
the shares of common stock for whom they may act as agent or to
whom they may sell as principal. Such commissions will be in
amounts to be negotiated, but, except as set forth in a
supplement to this prospectus, in the case of an agency
transaction will not be in excess of a customary brokerage
commission in compliance with the Financial Industry Regulatory
Authority or FINRA, Rule 2440; and in the case of a
principal transaction a markup or markdown in compliance with
FINRA IM-2440.
In connection with sales of the shares of common stock or
otherwise, the selling stockholders may enter into hedging
transactions with broker-dealers or other financial
institutions, which may in turn engage in short sales of the
shares of common stock in the course of hedging in positions
they assume. The selling stockholders may also sell shares of
common stock short and if such short sale shall take place after
the date that this registration statement is declared effective
by the SEC, the selling stockholders may deliver shares of
common stock covered by this prospectus to close out short
positions and to return borrowed shares in connection with such
short sales. The selling stockholders may also loan or pledge
shares of common stock to broker-dealers that in turn may sell
such shares, to the extent permitted by applicable law. The
selling stockholders may also enter into option or other
transactions with broker-dealers or other financial institutions
or the creation of one or more derivative securities which
require the delivery to such broker-dealer or other financial
institution of shares offered by this prospectus, which shares
such broker-dealer or other financial institution may resell
pursuant to this prospectus (as supplemented or amended to
reflect such transaction). Notwithstanding the foregoing, the
selling stockholders have been advised that they may not use
shares registered on this registration statement to cover short
sales of our common stock made prior to the date the
registration statement, of which this prospectus forms a part,
has been declared effective by the SEC.
The selling stockholders may, from time to time, pledge or grant
a security interest in some or all of the warrants or shares of
common stock owned by them and, if they default in the
performance of their secured obligations, the pledgees or
secured parties may offer and sell the shares of common stock
from time to time pursuant to this prospectus or any amendment
to this prospectus under Rule 424(b)(3) or other applicable
provision of the Securities Act, amending, if necessary, the
list of selling stockholders to include the pledgee, transferee
or other successors in interest as selling stockholders under
this prospectus. The selling stockholders also may transfer and
donate the shares of common stock in other circumstances in
which case the transferees, donees, pledgees or other successors
in interest will be the selling beneficial owners for purposes
of this prospectus.
In addition, as used in this prospectus, the selling
stockholders shall include individuals and/or entities to whom
VantagePoint may assign its registration rights, including other
funds under common control with VantagePoint, such funds
limited or general partners, limited liability company members
and/or individuals or entities who have been granted by a
limited partnership or limited liability company the right to
receive such distributions. The selling stockholders may from
time to time offer and sell pursuant to this prospectus any or
all of the common stock offered hereby.
The selling stockholders and any broker-dealer or agents
participating in the distribution of the shares of common stock
may be deemed to be underwriters within the meaning
of Section 2(11) of the Securities Act in connection with
such sales. In such event, any commissions paid, or any
discounts or concessions allowed to, any such broker-dealer or
agent and any profit on the resale of the shares purchased by
them may be deemed to be underwriting commissions or discounts
under the Securities Act. selling stockholders who are
underwriters within the meaning of
Section 2(11) of the Securities Act will be subject to the
applicable prospectus delivery requirements of the Securities
Act including
47
Rule 172 thereunder and may be subject to certain
statutory liabilities of, including but not limited to,
Sections 11, 12 and 17 of the Securities Act and
Rule 10b-5
under the Securities Exchange Act of 1934, as amended, or the
Exchange Act.
Each selling stockholder has informed the Company that it is not
a registered broker-dealer and does not have any written or oral
agreement or understanding, directly or indirectly, with any
person to distribute the common stock. Upon the Company being
notified in writing by a selling stockholder that any material
arrangement has been entered into with a broker-dealer for the
sale of common stock through a block trade, special offering,
exchange distribution or secondary distribution or a purchase by
a broker or dealer, a supplement to this prospectus will be
filed, if required, pursuant to Rule 424(b) under the
Securities Act, disclosing (i) the name of each such
selling stockholder and of the participating broker-dealer(s),
(ii) the number of shares involved, (iii) the price at
which such the shares of common stock were sold, (iv) the
commissions paid or discounts or concessions allowed to such
broker-dealer(s), where applicable, (v) that such
broker-dealer(s) did not conduct any investigation to verify the
information set out or incorporated by reference in this
prospectus, and (vi) other facts material to the
transaction. In no event shall any broker-dealer receive fees,
commissions and markups, which, in the aggregate, would exceed
eight percent (8.0%).
Under the securities laws of some states, the shares of common
stock may be sold in such states only through registered or
licensed brokers or dealers. In addition, in some states the
shares of common stock may not be sold unless such shares have
been registered or qualified for sale in such state or an
exemption from registration or qualification is available and is
complied with.
There can be no assurance that any selling stockholder will sell
any or all of the shares of common stock registered pursuant to
the registration statement, of which this prospectus forms a
part.
Each selling stockholder and any other person participating in
such distribution will be subject to applicable provisions of
the Exchange Act and the rules and regulations thereunder,
including, without limitation, to the extent applicable,
Regulation M of the Exchange Act, which may limit the
timing of purchases and sales of any of the shares of common
stock by the selling stockholder and any other participating
person. To the extent applicable, Regulation M may also
restrict the ability of any person engaged in the distribution
of the shares of common stock to engage in market-making
activities with respect to the shares of common stock. All of
the foregoing may affect the marketability of the shares of
common stock and the ability of any person or entity to engage
in market-making activities with respect to the shares of common
stock.
We will pay all expenses of the registration of the shares of
common stock pursuant to the registration rights agreement,
including, without limitation, SEC filing fees and expenses of
compliance with state securities or blue sky laws;
provided, however, that each selling stockholder
will pay all underwriting discounts and selling commissions, if
any and any related legal expenses incurred by it. We will
indemnify the selling stockholders against certain liabilities,
including some liabilities under the Securities Act, in
accordance with the registration rights agreement, or the
selling stockholders will be entitled to contribution. We may be
indemnified by the selling stockholders against civil
liabilities, including liabilities under the Securities Act,
that may arise from any written information furnished to us by
the selling stockholders specifically for use in this
prospectus, in accordance with the related registration rights
agreements, or we may be entitled to contribution.
48
MANAGEMENTS
DISCUSSION AND ANALYSIS OF
FINANCIAL CONDITION AND RESULTS OF OPERATIONS
The following discussion and analysis should be read together
with our financial statements and the related notes appearing
elsewhere in this prospectus. This discussion contains
forward-looking statements reflecting our current expectations
that involve risks and uncertainties. See Special Note
Regarding Forward-Looking Statements for a discussion of
the uncertainties, risks and assumptions associated with these
statements. Actual results and the timing of events could differ
materially from those discussed in our forward-looking
statements as a result of many factors, including those set
forth under Risk Factors and elsewhere in this
prospectus.
Overview
We are a biopharmaceutical company focused on developing and
commercializing products to treat serious diseases associated
with inflammation, including cardiovascular and autoimmune
diseases. We currently have one Phase 3 clinical program,
varespladib, and two Phase 2 clinical programs,
A-623 and
A-001. Two
of our product candidates, varespladib and
A-001, are
designed to inhibit a novel enzyme target known as secretory
phospholipase
A2,
or
sPLA2.
Elevated levels of
sPLA2
have been implicated in a variety of acute inflammatory
conditions, including acute coronary syndrome and acute chest
syndrome associated with sickle cell disease, as well as in
chronic diseases, including stable coronary artery disease. In
addition, our Phase 2 product candidate,
A-623,
targets elevated levels of B-Cell activating factor, or BAFF,
which has been associated with a variety of B-cell mediated
autoimmune diseases, including systemic lupus erythematosus, or
lupus, lupus nephritis, rheumatoid arthritis, multiple
sclerosis, Sjögrens Syndrome, Graves Disease
and others.
We were incorporated and commenced operations in September 2004.
Since our inception, we have generated significant losses. As of
June 30, 2010, we had an accumulated deficit of
approximately $84.3 million. As of the date of this
prospectus, we have never generated any revenue and have
generated only interest income from cash and cash equivalents
and short-term investments. We expect to incur substantial and
increasing losses for at least the next several years as we
pursue the development and commercialization of our product
candidates. In their report on our financial statements for the
year ended December 31, 2009, our independent auditors
included an explanatory paragraph regarding concerns about our
ability to continue as a going concern. Our financial statements
contain additional note disclosures describing the circumstances
that led to this disclosure.
To date, we have funded our operations through equity offerings
and private placements of convertible debt, raising an aggregate
of approximately $116.8 million. We will need substantial
additional financing to continue to develop our product
candidates, obtain regulatory approvals and to fund operating
expenses, which we will seek to raise through public or private
equity or debt financings, collaborative or other arrangements
with third parties or through other sources of financing. We
cannot assure you that such funds will be available on terms
favorable to us, if at all. In addition to the normal risks
associated with development-stage companies, we may never
successfully complete development of any of our product
candidates, obtain adequate patent protection for our
technology, obtain necessary government regulatory approval for
our product candidates or achieve commercial viability for any
approved product candidates. In addition, we may not be
profitable even if we succeed in commercializing any of our
product candidates.
Revenue
To date, we have not generated any revenue. We do not expect to
generate revenue unless or until we obtain regulatory approval
of, and commercialize, our product candidates or in- license
additional products that generate revenue. We intend to seek to
generate revenue from a combination of product sales, up-front
fees and milestone payments in connection with collaborative or
strategic relationships
49
and royalties resulting from the licensing of the commercial
rights to our intellectual property. We expect that any revenue
we generate will fluctuate from quarter to quarter as a result
of the nature, timing and amount of milestone payments we may
receive upon the sale of our products, to the extent any are
successfully commercialized, as well as any revenue we may
receive from our collaborative or strategic relationships.
Research
and Development Expenses
Since our inception, we have focused our activities on our
product candidate development programs. We expense research and
development costs as they are incurred. Research and development
expenses consist of personnel costs, including salaries,
benefits and stock-based compensation, clinical studies
performed by contract research organizations, or CROs, materials
and supplies, licenses and fees and overhead allocations
consisting of various administrative and facilities-related
costs. Research and development activities are also separated
into three main categories: licensing, clinical development and
pharmaceutical development. Licensing costs consist primarily of
fees paid pursuant to license agreements. Historically, our
clinical development costs have included costs for preclinical
and clinical studies. We expect to incur substantial clinical
development costs for our Phase 3 clinical study named VISTA- 16
for varespladib and for our Phase 2b clinical study named
PEARL-SC for
A-623, as
well as for the development of our other product candidates.
Pharmaceutical development costs consist of expenses incurred
relating to clinical studies and product formulation and
manufacturing.
We expense both internal and external research and development
costs as incurred. We are developing our product candidates in
parallel, and we typically use our employee and infrastructure
resources across several projects. Thus, some of our research
and development costs are not attributable to an individually
named project, but rather are allocated across our clinical
stage programs. These unallocated costs include salaries,
stock-based compensation charges and related fringe benefit
costs for our employees, consulting fees and travel.
The following table shows our total research and development
expenses for the years ended December 31, 2007, 2008 and
2009, the six months ended June 30, 2009 and 2010, and for
the period from September 9, 2004 (Date of Inception)
through June 30, 2010:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
For the Period
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
September 9,
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
2004 (Date of
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Inception)
|
|
|
|
Year Ended December 31,
|
|
|
Six Months Ended June 30,
|
|
|
to June 30,
|
|
|
|
2007
|
|
|
2008
|
|
|
2009
|
|
|
2009
|
|
|
2010
|
|
|
2010
|
|
|
Allocated costs:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
A-001
|
|
$
|
2,302,454
|
|
|
$
|
456,633
|
|
|
$
|
192,979
|
|
|
$
|
121,460
|
|
|
$
|
113,107
|
|
|
$
|
6,633,153
|
|
Varespladib
|
|
|
12,053,943
|
|
|
|
7,370,850
|
|
|
|
5,535,529
|
|
|
|
3,829,160
|
|
|
|
7,962,469
|
(2)
|
|
|
35,823,114
|
(3)
|
A-623
|
|
|
6,004,667
|
(1)
|
|
|
100,851
|
|
|
|
34,179
|
|
|
|
9,958
|
|
|
|
2,158,120
|
|
|
|
8,301,537
|
|
Unallocated costs
|
|
|
3,560,868
|
|
|
|
2,953,988
|
|
|
|
2,652,727
|
|
|
|
1,240,603
|
|
|
|
1,446,267
|
|
|
|
12,246,140
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Total development
|
|
$
|
23,921,932
|
|
|
$
|
10,882,322
|
|
|
$
|
8,415,414
|
|
|
$
|
5,201,181
|
|
|
$
|
11,679,963
|
|
|
$
|
63,003,944
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(1)
|
Includes a one-time license initiation fee of $6.0 million
pursuant to a license agreement with Amgen.
|
(2)
|
Includes milestone payments of $3.5 million pursuant to
amendments to the license agreements with each of Eli Lilly and
Shionogi & Co. Ltd., which were paid in the form of
shares of common stock.
|
(3)
|
Includes a one-time license fee initiation of $4.0 million
pursuance to a license agreement with each of Eli Lilly and
Shionogi & Co. Ltd., which were paid in the form of
shares of preferred stock.
|
50
We expect our research and development expenses to increase
significantly as we continue to develop our product candidates.
We began enrollment of patients in the VISTA-16 study of
varespladib for the treatment of patients experiencing acute
coronary syndrome in June 2010. We also initiated the
PEARL-SC
study of
A-623 in
July 2010. We intend to fund our clinical studies with existing
cash and future offerings.
We expect that a large percentage of our research and
development expenses in the future will be incurred in support
of our current and future clinical development programs. These
expenditures are subject to numerous uncertainties in timing and
cost to completion. As we obtain results from clinical studies,
we may elect to discontinue or delay clinical studies for
certain product candidates or programs in order to focus our
resources on more promising product candidates or programs.
Completion of clinical studies may take several years or more,
but the length of time generally varies according to the type,
complexity, novelty and intended use of a product candidate. The
cost of clinical studies may vary significantly over the life of
a program as a result of differences arising during clinical
development, including:
|
|
|
|
|
the number of sites included in the studies;
|
|
|
|
the length of time required to enroll suitable patient subjects;
|
|
|
|
the number of patients that participate in the studies;
|
|
|
|
the number of doses that patients receive;
|
|
|
|
the drop-out or discontinuation rates of patients; and
|
|
|
|
the duration of patient
follow-up.
|
Our expenses related to clinical studies are based on estimates
of the services received and efforts expended pursuant to
contracts with multiple research institutions and clinical
research organizations that conduct and manage clinical studies
on our behalf. The financial terms of these agreements are
subject to negotiation and vary from contract to contract and
may result in uneven payment flows. Generally, these agreements
set forth the scope of work to be performed at a fixed fee or
unit price. Payments under the contracts depend on factors such
as the successful enrollment of patients or the completion of
clinical study milestones. Expenses related to clinical studies
generally are accrued based on contracted amounts and the
achievement of milestones such as number of patients enrolled.
If timelines or contracts are modified based upon changes to the
clinical study design or scope of work to be performed, we
modify our estimates of accrued expenses accordingly on a
prospective basis.
None of our product candidates has received U.S. Food and
Drug Administration, or FDA, or foreign regulatory marketing
approval. In order to grant marketing approval, the FDA or
foreign regulatory agencies must conclude that clinical data
establishes the safety and efficacy of our product candidates
and that the manufacturing facilities, processes and controls
are adequate. Despite our efforts, our product candidates may
not offer therapeutic or other improvement over existing,
comparable drugs, be proven safe and effective in clinical
studies, or meet applicable regulatory standards.
As a result of the uncertainties discussed above, we are unable
to determine the duration and completion costs of our
development projects or when and to what extent we will receive
cash inflows from the commercialization and sale of an approved
product candidate, if ever.
51
General
and Administrative Expenses
General and administrative expenses consist primarily of
compensation for employees in executive and operational
functions, including clinical, chemical manufacturing,
regulatory, finance and business development. Other significant
costs include professional fees for legal services, including
legal services associated with obtaining and maintaining
patents. We will continue to incur significant general and
administrative expenses as a public company, including costs for
insurance, costs related to the hiring of additional personnel,
payment to outside consultants, lawyers and accountants and
complying with the corporate governance, internal controls and
similar requirements applicable to public companies.
Critical
Accounting Policies and Estimates
Our managements discussion and analysis of our financial
condition and results of operations is based on our financial
statements, which have been prepared in accordance with
accounting principles generally accepted in the United States,
or GAAP. The preparation of these financial statements requires
us to make estimates and judgments that affect the reported
amounts of assets, liabilities and expenses. On an ongoing
basis, we evaluate these estimates and judgments, including
those described below. We base our estimates on our historical
experience and on various other assumptions that we believe to
be reasonable under the circumstances. These estimates and
assumptions form the basis for making judgments about the
carrying values of assets and liabilities that are not readily
apparent from other sources. Actual results and experiences may
differ materially from these estimates.
While our significant accounting policies are more fully
described in Note 2 to our financial statements included at
the end of this prospectus, we believe that the following
accounting policies are the most critical to aid you in fully
understanding and evaluating our reported financial results and
affect the more significant judgments and estimates that we use
in the preparation of our financial statements.
Accrued
Clinical Expenses
As part of the process of preparing our financial statements, we
are required to estimate our accrued expenses. This process
involves reviewing open contracts and purchase orders,
communicating with our applicable personnel to identify services
that have been performed on our behalf and estimating the level
of service performed and the associated cost incurred for the
service when we have not yet been invoiced or otherwise notified
of actual cost. The majority of our service providers invoice us
monthly in arrears for services performed. We make estimates of
our accrued expenses as of each balance sheet date in our
financial statements based on facts and circumstances known to
us at that time. We periodically confirm the accuracy of our
estimates with the service providers and make adjustments if
necessary. Examples of estimated accrued clinical expenses
include:
|
|
|
|
|
fees paid to CROs in connection with clinical studies;
|
|
|
|
fees paid to investigative sites in connection with clinical
studies;
|
|
|
|
fees paid to contract manufacturers in connection with the
production of clinical study materials; and
|
|
|
|
fees paid to vendors in connection with the preclinical
development activities.
|
We base our expenses related to clinical studies on our
estimates of the services received and efforts expended pursuant
to contracts with multiple research institutions and CROs that
conduct and manage clinical studies on our behalf. The financial
terms of these agreements are subject to negotiation, vary from
contract to contract and may result in uneven payment flows.
Payments under some of these contracts depend on factors such as
the successful enrollment of patients and the completion of
clinical
52
study milestones. In accruing service fees, we estimate the time
period over which services will be performed and the level of
effort to be expended in each period. If the actual timing of
the performance of services or the level of effort varies from
our estimate, we adjust the accrual accordingly. If we do not
identify costs that we have begun to incur or if we
underestimate or overestimate the level of services performed or
the costs of these services, our actual expenses could differ
from our estimates.
Stock-Based
Compensation
Effective January 1, 2006, we adopted the provisions of
FASB ASC 718, Compensation Stock
Compensation, using the modified prospective method.
Compensation costs related to all equity instruments granted
after January 1, 2006 are recognized at the grant-date fair
value of the awards. Additionally, we are required to include an
estimate of the number of awards that will be forfeited in
calculating compensation costs, which are recognized over the
requisite service period of the awards on a straight-line basis.
We estimate the fair value of our share-based payment awards on
the date of grant using an option-pricing model.
We recognized employee stock-based compensation expense of
approximately $75,000 in 2007, $143,000 in 2008, $254,000 in
2009, and $106,000 for the six months ended June 30, 2010.
As of June 30, 2010, we had $2.0 million in total
unrecognized compensation cost related to non-vested employee
stock-based compensation arrangements, $1.5 million of
which is related to unvested restricted stock units and the
remaining $0.5 million of which is related to unvested
stock options. The intrinsic value of all outstanding vested and
non-vested stock-based compensation arrangements, based on $4.19
per share, which is the closing sale price of our common stock
as reported on The NASDAQ Global Market on September 30,
2010, is $3.9 million, based on 1,307,066 shares of
our common stock issuable upon exercise of stock options at a
weighted-average exercise price of $1.27 per share and
333,000 shares of unvested restricted stock units at a
weighted-average purchase price of $5.15 per share.
We calculate the fair value of stock-based compensation awards
using the Black-Scholes option-pricing model. For the years
ended December 31, 2007, 2008 and 2009, and the six months
ended June 30, 2010, the weighted-average assumptions used
in the Black-Scholes model were 6.25 years for the expected
terms, 81%, 81%, 74% and 89% for the expected volatility, 4.54%,
3.08%, 2.10% and 3.02% for the risk free rate and 0.0% for
dividend yield, respectively. Expense amounts for future awards
for any particular quarterly or annual period could be affected
by changes in our assumptions. The weighted-average expected
option terms for 2007, 2008, 2009 and the six months ended
June 30, 2010 reflect the application of the simplified
method set out in FASB
ASC 718-10.
The simplified method defines the life as the average of the
contractual term of the stock-based compensation award and the
weighted-average vesting period for all tranches. Estimated
volatility for fiscal 2007, 2008, 2009 and six months ended
June 30, 2010 also reflects the application of interpretive
guidance provided in FASB
ASC 718-10
and, accordingly, incorporates historical volatility of similar
public entities.
The exercise price of options to purchase our common stock
granted to our employees, directors and consultants was the fair
value of our common stock on the date of grant. Prior to our
initial public offering in March 2010, the fair value of our
common stock was determined by our board of directors as there
was no public market for our common stock at that time. Our
board of directors determined the fair value of our common stock
based on several factors, including:
|
|
|
|
|
the rights, preferences and privileges of our preferred stock
(which was then outstanding) relative to our common stock;
|
|
|
|
our performance and stage of development;
|
53
|
|
|
|
|
the likelihood of achieving a liquidity event for the shares of
our common stock underlying these stock options, such as an
initial public offering or sale of our company, given prevailing
market conditions;
|
|
|
|
the trading value of common stock of public companies comparable
to our company;
|
|
|
|
the sale prices of comparable acquisition transactions of public
companies comparable to ours; and
|
|
|
|
the available data resulting from our clinical studies and
development to date.
|
In considering the rights, preferences and privileges of our
preferred stock relative to our common stock, our board of
directors considered the following rights, preferences and
privileges of our
Series B-1
and
Series B-2
preferred stock:
|
|
|
|
|
a senior liquidation preference of $7.28 per share in the event
of any sale of our company or similar liquidity event;
|
|
|
|
a right to participate alongside our common stock in the event
of any sale or similar liquidity event with a 3.5x cap on such
participation;
|
|
|
|
a senior non-cumulative dividend of 7.0% of the original issue
price;
|
|
|
|
protection against dilutive issuances of new shares;
|
|
|
|
a right to convert each share of preferred stock into common
stock;
|
|
|
|
a right to receive quarterly unaudited and annual audited
financial statements, to inspect our books and records and to
meet with our management team;
|
|
|
|
a right to vote with other holders of preferred and common stock
to elect members of our board of directors; and
|
|
|
|
a right to vote separately on issues such as changes in capital
structure, interested party transactions, mergers, sales and
acquisitions.
|
Specifically, with respect to liquidation preference and
participation features, each share of our
Series B-1
and
Series B-2
preferred stock had a liquidation preference equal to the price
per share at which such share was sold, and in addition, would
have participated with the common stock on proceeds available
for distribution in a buy-out or sale of our company until such
preferred shares received
three-and-one-half
times the original price per share. As a result of these
participation rights and preferences, the preferred shareholders
would have received substantially more of our companys
value in the event of the dissolution or liquidation of our
company, such as in a buy-out or sale of our company, or on the
payment of the dividends. For example, on a buy-out or sale of
our company, the
Series B-1
and
B-2 shareholders
were each entitled to receive liquidation preferences of $7.28
per share, before then participating equally with the common
shareholders in the remaining value of our company until they
have received $25.46 per share.
In addition, we obtained the reports of independent valuation
firms with respect to their estimates of the fair values of our
common stock. We obtained reports of the fair value of our
common stock as of October 31, 2006 on December 4,
2006, as of December 31, 2007 on February 12, 2008,
and as of October 15, 2008 on October 24, 2008. In
estimating the fair value of our common stock, the
54
independent firms used the income approach. The income approach
is an estimate of the present value of the future monetary
benefits expected to flow to the owners of a business. It
requires a projection of the cash flows that the business
expected to generate over a forecast period and an estimate of
the present value of cash flows beyond that period, which is
referred to as residual value. These cash flows are converted to
present value by means of discounting, using a rate of return
that accounts for the time value of money and the appropriate
degree of risks inherent in the business. After calculation of
the companys enterprise value using this approach, the
value of a share of common stock is then discounted for lack of
marketability, or the inability to readily sell shares, which
increases the owners exposure to changing market
conditions and increases the risk of ownership.
In its report as of December 31, 2007, the independent
valuation firm estimated our enterprise value using discounted
cash flows, a terminal value based on comparable publicly traded
company revenue multiples and a risk-adjusted discount rate of
39.1%. Our enterprise value was estimated to be approximately
$34.0 million. This enterprise value was then allocated
among the various classes of our securities, including preferred
stock, common stock and options to purchase common stock using
the Black-Scholes option-pricing model, which yielded an
estimated value per share of our common stock of $1.76, which
was in turn reduced by a discount for lack of marketability of
24.0% using a protective put analysis and an estimated time to
liquidity of two years, which resulted in an estimated value per
share of $1.34.
In its report as of October 15, 2008, the independent
valuation firm estimated our enterprise value using discounted
cash flows, a terminal value based on comparable publicly traded
company revenue multiples and a risk-adjusted discount rate of
32.6%. Our enterprise value was estimated to be approximately
$60.6 million. This enterprise value was then allocated
among the various classes of our securities, including preferred
stock, common stock and options to purchase common stock using
the Black-Scholes option-pricing model, which yielded an
estimated value per share of our common stock of $2.02, which
was in turn reduced by a discount for lack of marketability of
25.0% using a protective put analysis and an estimated time to
liquidity of two years, which resulted in an estimated value per
share of $1.51.
On October 13, 2009, our board of directors determined an
estimated fair value per share of $7.70 for our common stock.
Our board of directors examined the enterprise values of 10 peer
companies in the life sciences industry and used the mean
enterprise value to approximate our anticipated enterprise value
upon completion of a public offering. Our board of directors
used the mean enterprise value, rather than a multiple of
earnings or revenue, since we have no earnings or revenue, nor
do any of the companies in the peer group. We selected the peer
group based on the following criteria: publicly traded drug
development companies that have one or more pharmaceutical
compounds targeted at patient markets of approximately the same
size as the target market for our compounds in Phase 2 or Phase
3 clinical studies and no compounds yet approved for general
use. To estimate our enterprise value, our board of directors
discounted the enterprise value by 15% to reflect a lack of
marketability. Our board of directors then further discounted
the estimated enterprise value by an additional 25% to reflect
the time our board of directors estimated would be necessary to
complete our initial public offering as well as the risk that
such offering would not be completed. 100% of this enterprise
value was then allocated to our common stock, assuming the
conversion of all shares of preferred stock outstanding and the
exercise of all outstanding options and warrants, which yielded
an estimated fair value of our common stock of $7.70 per share.
In determining the valuation of our common stock, our board of
directors did not take into account (i) the expected timing
of commercialization of our varespladib product candidate, other
than 2012 being the earliest possible time of commercialization,
which is already reflected in the discount for lack of
marketability and liquidity, or (ii) any future revenues
and operating profits expected to be generated from sales of
varespladib.
55
Based on the factors listed above, our board of directors
determined the fair value of our common stock for option grants
made in October 2009 to be $7.70 per share, for option grants
made in February and April 2009 to be $1.51 per share, and for
option grants made in 2008 to be $1.34 per share. The following
table summarizes by grant date the number of shares of common
stock subject to options granted in 2008 and 2009 through the
date of our initial public offering and the associated per-share
exercise price. The exercise prices were set by our board of
directors at prices believed to equal the fair value of our
common stock at each of the grant dates.
|
|
|
|
|
|
|
|
|
Grant Date
|
|
Number of Options
|
|
Per Share Exercise Price
|
|
2/21/2008
|
|
|
287,086
|
|
|
$
|
1.34
|
|
6/26/2008
|
|
|
40,887
|
|
|
$
|
1.34
|
|
2/18/2009
|
|
|
367,395
|
|
|
$
|
1.51
|
|
4/15/2009
|
|
|
26,281
|
|
|
$
|
1.51
|
|
10/13/2009
|
|
|
11,682
|
|
|
$
|
7.70
|
|
The estimated fair value common stock from June 2008 to February
2009 increased from $1.34 per share to $1.51 per share. The
change in estimated fair value was primarily the result of an
increase in the estimated enterprise value of the company from
$34.0 million to $60.6 million, and reflected the
following positive factors:
|
|
|
|
|
successful completion of enrollment of our Phase 2b FRANCIS
study; and
|
|
|
|
the conclusion in February 2009 of a DSMB evaluation that our
IMPACTS study was well-tolerated and should continue.
|
The positive factors set forth above were partially offset by:
|
|
|
|
|
a sharp deterioration in financial markets with accompanying
decrease in market capitalization of companies comparable to
ours;
|
|
|
|
increased difficulty in raising equity financing with
accompanying financing uncertainty; and
|
|
|
|
increased risk of failure to achieve an initial public offering,
sale of the company or other similar liquidity event.
|
While no single factor listed above was specifically quantified
or weighted greater than another in estimating the
companys enterprise value, each was taken into account in
calculating the discount rate for the discounted cash flow
analysis, estimating the time to liquidity and the expense that
would be required to achieve liquidity.
The estimated fair value of our common stock from April 2009 to
October 2009 increased from $1.51 per share to $7.70 per share.
The change in estimated fair value primarily reflected the
following factors:
|
|
|
|
|
we successfully achieved the primary endpoint of our Phase 2b
FRANCIS study in July 2009;
|
|
|
|
an analysis of secondary endpoints from FRANCIS revealed
generally favorable efficacy trends in August 2009;
|
|
|
|
a successful initial public offering of a company in our
industry; and
|
|
|
|
progress towards our initial public offering.
|
56
While no single factor listed above was specifically quantified
or weighted greater than another in estimating the
companys enterprise value, each was taken into account in
estimating the time to liquidity and the expense that would be
required to achieve liquidity.
The initial public offering price of our common stock was $7.00
per share. The difference between the estimated fair value of
our common stock of $7.70 per share in October 2009 and the
initial public offering price took into account several factors
considered by our board of directors and the underwriters:
|
|
|
|
|
an analysis of the typical valuation ranges seen in initial
public offerings for companies in our industry with similar
market capitalization for the last five years;
|
|
|
|
a review of then current market conditions and the results of
operations, competitive position and the stock performance of
our competitors; and
|
|
|
|
consideration of our history as a private company and previous
valuation reports received by independent valuation firms.
|
As of June 30, 2010, 1,239,129 shares of our common
stock were issuable upon exercise of stock options.
Results
of Operations
Comparison
of the Six Months Ended June 30, 2010 and
2009
Research and Development
Expenses. Research and development expenses
were $11.7 million for the six months ended June 30,
2010, compared with $5.2 million for the six months ended
June 30, 2009. The $6.5 million increase in our
research and development expenses was primarily attributable to
the recognition of a $3.5 million non-cash charge related
to milestone payments recorded in connection with the initiation
of our Phase 3 clinical study for varespladib, which we paid by
issuing 531,914 shares of common stock, and increased CRO
and manufacturing costs related to the launch of our Phase 3
clinical study for varespladib and Phase 2b clinical study for
A-623, as
well as increased headcount to support these clinical studies.
General and Administrative
Expenses. General and administrative expenses
were $2.7 million for the six months ended June 30,
2010, compared with $1.8 million for the six months ended
June 30, 2009. The $0.9 million increase was primarily
attributable to increased headcount and professional services
incurred in connection with our financial audit and other costs
associated with operating as a public company.
Interest and Other Income. Interest and
other income was $15,000 for the six months ended June 30,
2010, compared with $22,000 for the six months ended
June 30, 2009. The decrease in interest and other income
was due to a lower rate of return on cash equivalents and
short-term investments as compared to the prior year.
Interest and Other Expense. Interest
and other expense was $4.6 million for the six months ended
June 30, 2010, compared with $75,000 for the six months
ended June 30, 2009. Interest and other expense recorded
during the six months ended June 30, 2010 included a
$4.5 million non-cash charge recorded as part of interest
and other expense related to the amortization of discounts on
our convertible promissory notes and the
mark-to-market
adjustment relating to warrants and embedded derivative
connected to our convertible promissory notes. Interest and
other expense recorded during the comparable period in 2009
consisted of interest accrued on past due license fee
obligations.
57
Comparison
of the Years Ended December 31, 2009 and 2008
Research and Development
Expenses. Research and development expenses
were $8.4 million for the year ended December 31,
2009, compared with $10.9 million for the year ended
December 31, 2008. The $2.5 million decrease in our
research and development expenses was due to the decreased
activity in our Phase 2 clinical study designed to examine the
impact of varespladib when administered to patients within
96 hours of an acute coronary syndrome event in the third
quarter of 2009 as the study progressed toward completion.
General and Administrative
Expenses. General and administrative expenses
were $3.4 million for the year ended December 31,
2009, compared with $3.0 million for the year ended
December 31, 2008. The $0.4 million increase was
primarily attributable to expenses relating to the expansion of
our intellectual property portfolio.
Interest and Other Income. Interest and
other income was $24,000 for the year ended December 31,
2009, compared with $178,000 for the year ended
December 31, 2008. The decrease in interest and other
income was due to lower average cash balances.
Interest and Other Expense. Interest
and other expense was $386,000 for the year ended
December 31, 2009, compared with $296,000 for the year
ended December 31, 2008. Interest and other expense
recorded in 2009 consisted of interest accrued for convertible
promissory notes and amortization of note discount and debt
issuance cost. Interest and other expense recorded in 2008
consisted of interest accrued on past due license fee
obligations.
Beneficial Conversion Feature. In
connection with the issuance of convertible promissory notes in
2008, we recorded expense related to the beneficial conversion
feature of the notes in the amount of $4.1 million for the
year ended December 31, 2008. The expense was amortized
from the issuance date of the notes to the date of their
conversion into shares of
Series B-2
convertible preferred stock in August 2008. The convertible
promissory notes issued in 2009 included a beneficial conversion
feature that would be measured and recorded upon a triggering
event as defined in the agreement.
Comparison
of the Years Ended December 31, 2008 and December 31,
2007
Research and Development
Expenses. Research and development expenses
were $10.9 million for the year ended December 31,
2008, compared with $23.9 million for the year ended
December 31, 2007. The $13.0 million decrease in our
research and development expenses reflects a one-time license
initiation fee of $6.0 million recognized in 2007 in
connection with a worldwide, exclusive license agreement we
entered into with Amgen (see Note 5 to our financial
statements for further details). The remaining decrease of
$7.0 million was primarily attributable to reduced clinical
costs associated with our Phase 2 clinical studies for the
development of varespladib. In 2007, we initiated and completed
two Phase 2 clinical studies for varespladib, while in 2008, we
initiated a single Phase 2b clinical study for varespladib.
General and Administrative
Expenses. General and administrative expenses
were $3.0 million for the year ended December 31,
2008, compared with $2.5 million for the year ended
December 31, 2007. The $0.5 million increase was
primarily attributable to our implementation of our vacation
policy, professional fees relating to the expansion of our
intellectual property portfolio and travel relating to business
development activities primarily consisting of scientific and
industry conferences and symposiums.
Interest and Other Income. Interest and
other income was $178,000 for the year ended December 31,
2008, compared with $697,000 for the year ended
December 31, 2007. The decrease in interest and
58
other income of approximately $519,000 was primarily
attributable to lower average cash balances and lower average
interest rates during 2008.
Interest Expense. Interest expense was
$296,000 for the year ended December 31, 2008, compared
with no interest expense for the year ended December 31,
2007. The interest expense during the year ended
December 31, 2008 was due to interest recognized in
connection with issuance of convertible promissory notes in
February and May 2008, which were converted into shares of our
Series B-2
convertible preferred stock in connection with our
Series B-2
financing consummated in August 2008 and interest accrued in
connection with a license fee payable due to Amgen.
Beneficial Conversion Features. For the
year ended December 31, 2008, we recorded $4.1 million
in expense related to the beneficial conversion features of our
convertible promissory notes, which were convertible into shares
of our
Series B-2
convertible preferred stock at a discount of 25% from the
original issue price of our
Series B-2
convertible preferred stock. There were no outstanding notes
with similar terms during 2007.
Liquidity
and Capital Resources
To date, we have funded our operations primarily through private
placements of preferred stock, common stock and convertible debt
and our initial public offering. As of June 30, 2010, we
had received net proceeds of approximately $90.5 million
from the sale of equity securities and net proceeds of
approximately $26.3 million from the issuance of
convertible promissory notes. As of June 30, 2010, we had
cash, cash equivalents and short-term investments of
approximately $51.0 million.
Cash
Flows
Six
Months Ended June 30, 2010
For the six months ended June 30, 2010, we incurred a net
loss of approximately $19.0 million.
Net cash used in operating activities was approximately
$11.1 million. The net loss is higher than cash used in
operating activities by $7.9 million. The primary drivers
for the difference are adjustments for non-cash charges such as
stock-based compensation of approximately $114,000, amortization
of note discount and debt issuance cost of approximately
$769,000, issuance of $3.5 million shares of common stock
in lieu of cash milestone payments due to Eli Lilly and
Shionogi & Co., Ltd., the conversion of approximately
$300,000 of accrued interest into shares of common stock upon
conversion of certain convertible promissory notes and mark to
market adjustments relating to warrant and derivative liability
of $3.8 million, offset by a decrease in operating assets
and liabilities of approximately $468,000.
Net cash used by investing activities was $15.3 million and
was primarily driven by the purchase of short-term investments
during the period.
Net cash provided by financing activities was approximately
$58.4 million and consisted of proceeds of
$61.2 million received from the issuance of common stock in
our initial public offering, the exercise of the overallotment
option by our underwriters in connection with our initial public
offering and the release of funds held in an escrow account from
the sale of shares of our common stock in a private placement
concurrent with the closing of our initial public offering,
offset by approximately $2.9 million of issuance cost paid
during the period.
Six
Months Ended June 30, 2009
For the six months ended June 30, 2009, we incurred a net
loss of approximately $7.1 million.
59
Net cash used in operating activities was approximately
$6.6 million. The net loss is higher than cash used in
operating activities by $500,000. The primary drivers for the
difference are adjustments for non-cash charges such as
depreciation and amortization of $9,500, stock-based
compensation of $135,000 due to increased headcount and
corresponding equity grants made to new and existing employees,
an increase in current liabilities of $841,000 due to increased
expense relating to our Phase 2 clinical study activity and a
decrease in license fee payable by $500,000, offset by a
decrease in current assets of $31,000.
No cash was used or provided by investing or financing
activities during the six months ended June 30, 2009.
Year
Ended December 31, 2009
For the year ended December 31, 2009, we incurred a net
loss of approximately $12.2 million.
Net cash used in operating activities was approximately
$17.2 million. The net loss is higher than cash used in
operating activities by $5.0 million. The primary drivers
for the difference are adjustments for non-cash charges such as
depreciation of $18,000, stock-based compensation of
approximately $342,000 and amortization of note discount and
debt issuance cost of approximately $216,000, a decrease in
current liabilities of approximately $598,000 primarily due to
payments made to CROs for the achievement of clinical milestones
and a $5.0 million license fee payment made to Amgen.
Net cash provided by financing activities was approximately
$13.0 million and consisted of net proceeds of
$13.3 million received from the issuance of convertible
promissory notes and escrow notes, partially offset by
approximately $274,000 in expense paid in connection with our
initial public offering.
Year
Ended December 31, 2008
For the year ended December 31, 2008, we incurred a net
loss of $18.1 million.
Net cash used in operating activities was approximately
$17.1 million. The net loss is higher than cash used in
operating activities by $1.0 million. The primary drivers
for the difference are adjustments for non-cash charges such as
depreciation and amortization of $22,000 and stock-based
compensation of $195,000 due to increased headcount and
corresponding equity grants made to new and existing employees,
issuance of convertible preferred stock in lieu of interest
payments of $156,000, beneficial conversion feature of
$4.1 million and a decrease in current assets of $31,000,
offset by a decrease in current liabilities of $2.6 million
due to payments made to vendors for Phase 2 clinical study
activities previously completed and a decrease in license fee
payable of $1.0 million due to payments made.
Net cash provided by investing activities was approximately
$5.8 million and consisted of proceeds received from the
sale or maturity of short-term investments.
Net cash provided by financing activities was approximately
$19.0 million and consisted primarily of private placements
of our convertible preferred stock, through which we received
net proceeds of $6.8 million, and issuance of convertible
promissory notes for $12.2 million, which were converted
into
Series B-2
convertible preferred stock during 2008.
Year
Ended December 31, 2007
For the year ended December 31, 2007, we incurred a net
loss of $25.7 million.
Net cash used in operating activities was approximately
$15.0 million. The net loss is higher than cash used in
operating activities by $10.7 million. The primary drivers
for the difference are adjustments for
60
non-cash charges such as depreciation and amortization of
$19,000, amortization of discount on short-term investments of
$130,000 and stock-based compensation of $87,000, offset by an
increase in current liabilities of $4.8 million as a result
of increased Phase 2 clinical study expenses, an increase of
license fee payable of $6.0 million due the completion of a
licensing agreement with Amgen to acquire the rights to
A-623 and an
increase in current assets of $62,000.
Net cash used in investing activities was approximately
$5.8 million, consisting primarily of purchases of
short-term investments of $14.8 million, offset by proceeds
from the sale or maturity of these investments totaling
$9.1 million.
Net cash provided by financing activities was approximately
$119,000, which consisted of cash proceeds from the exercise of
stock options.
Selected
Quarterly Financial Data (Unaudited)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Quarter Ended
|
|
|
|
March 31
|
|
|
June 30
|
|
|
September 30
|
|
|
December 31
|
|
|
2008
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
OPERATING EXPENSES:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Research and development
|
|
$
|
2,996,942
|
|
|
$
|
2,366,494
|
|
|
$
|
2,111,817
|
|
|
$
|
3,407,069
|
|
General and administrative
|
|
|
849,251
|
|
|
|
742,992
|
|
|
|
713,367
|
|
|
|
674,560
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
LOSS FROM OPERATIONS
|
|
|
(3,846,193
|
)
|
|
|
(3,109,486
|
)
|
|
|
(2,825,184
|
)
|
|
|
(4,081,629
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Interest income and other expense (net)
|
|
|
37,938
|
|
|
|
(49,312
|
)
|
|
|
(108,521
|
)
|
|
|
1,721
|
|
Beneficial conversion features
|
|
|
|
|
|
|
(1,392,601
|
)
|
|
|
(2,725,943
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
NET LOSS
|
|
$
|
(3,808,255
|
)
|
|
$
|
(4,551,399
|
)
|
|
$
|
(5,659,648
|
)
|
|
$
|
(4,079,908
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss per share basic and diluted
|
|
$
|
(3.15
|
)
|
|
$
|
(3.45
|
)
|
|
$
|
(4.05
|
)
|
|
$
|
(2.83
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Shares used in computing basic and diluted net loss per share
|
|
|
1,210,757
|
|
|
|
1,317,862
|
|
|
|
1,398,120
|
|
|
|
1,443,843
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Quarter Ended
|
|
|
|
March 31
|
|
|
June 30
|
|
|
September 30
|
|
|
December 31
|
|
|
2009
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
OPERATING EXPENSES:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Research and development
|
|
$
|
2,914,766
|
|
|
$
|
2,286,415
|
|
|
$
|
2,525,948
|
|
|
$
|
688,285
|
|
General and administrative
|
|
|
846,243
|
|
|
|
999,331
|
|
|
|
884,908
|
|
|
|
695,208
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
LOSS FROM OPERATIONS
|
|
|
(3,761,009
|
)
|
|
|
(3,285,746
|
)
|
|
|
(3,410,856
|
)
|
|
|
(1,383,493
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Interest income and other expense (net)
|
|
|
(24,351
|
)
|
|
|
(50,310
|
)
|
|
|
(193,556
|
)
|
|
|
(94,171
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
NET LOSS
|
|
$
|
(3,785,360
|
)
|
|
$
|
(3,336,056
|
)
|
|
$
|
(3,604,412
|
)
|
|
$
|
(1,477,664
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Net loss per share basic and diluted
|
|
$
|
(2.57
|
)
|
|
$
|
(2.23
|
)
|
|
$
|
(2.37
|
)
|
|
$
|
(0.95
|
)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Shares used in computing basic and diluted net loss per share
|
|
|
1,470,722
|
|
|
|
1,496,011
|
|
|
|
1,520,875
|
|
|
|
1,557,708
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
61
|
|
|
|
|
|
|
|
|
|
|
Quarter Ended
|
|
|
|
March 31
|
|
|
June 30
|
|
|
2010
|
|
|
|
|
|
|
|
|
OPERATING EXPENSES:
|
|
|
|
|
|
|
|
|
Research and development
|
|
$
|
5,241,814
|
|
|
$
|
6,438,149
|
|
General and administrative
|
|
|
1,224,110
|
|
|
|
1,509,869
|
|
|
|
|
|
|
|
|
|
|
LOSS FROM OPERATIONS
|
|
|
(6,465,924
|
)
|
|
|
(7,948,018
|
)
|
|
|
|
|
|
|
|
|
|
Interest income and other expense (net)
|
|
|
(4,637,868
|
)
|
|
|
11,655
|
|
|
|
|
|
|
|
|
|
|
NET LOSS
|
|
$
|
(11,103,792
|
)
|
|
$
|
(7,936,363
|
)
|
|
|
|
|
|
|
|
|
|
Net loss per share basic and diluted
|
|
$
|
(0.83
|
)
|
|
$
|
(0.36
|
)
|
|
|
|
|
|
|
|
|
|
Shares used in computing basic and diluted net loss per share
|
|
|
13,344,231
|
|
|
|
22,223,941
|
|
|
|
|
|
|
|
|
|
|
Contractual
Obligations and Commitments
The following table summarizes our long-term contractual
obligations and commitments as of June 30, 2010:
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Payments Due by Period
|
|
|
|
|
Less Than
|
|
|
|
|
|
After
|
|
|
Total
|
|
1 Year
|
|
1-3 Years
|
|
4-5 Years
|
|
5 Years
|
|
Operating lease
obligations(1)
|
|
$
|
35,952
|
|
|
$
|
29,712
|
|
|
$
|
6,240
|
|
|
$
|
|
|
|
$
|
|
|
|
|
(1) |
Operating lease obligations reflect our obligation to make
payments in connection with a sublease that commenced in October
2008 and will expire on September 30, 2010 (which was
extended to January 31, 2011) for approximately
7,800 square feet of office space and office equipment
leases that commenced in October 2007 and will expire in June
2013.
|
The above amounts exclude potential payments to be made under
our license agreements to our licensors that are based on the
progress of our product candidates in development, as these
payments are not determinable. Under our license agreement with
Eli Lilly and Shionogi & Co., Ltd. to develop and
commercialize certain
sPLA2
inhibitors, we are obligated to make additional milestone
payments upon the achievement of certain development,
regulatory, and commercial objectives. We are also obligated to
pay royalties on future net sales of products that are developed
and approved as defined by this collaboration. Our obligation to
pay royalties with respect to each licensed product in each
country will expire upon the later of (a) 10 years
following the date of the first commercial sale of such licensed
product in such country, and (b) the first date on which
generic version(s) of the applicable licensed product achieve a
total market share, in the aggregate, of 25% or more of the
total unit sales of wholesalers to pharmacies of licensed
product and all generic versions combined in the applicable
country.
Also excluded from the table above are potential milestone
payments on the development of
A-623. Under
our license agreement with Amgen to develop and commercialize
A-623, we
are obligated to make additional milestone payments upon the
achievement of certain development, regulatory, and commercial
objectives. We are also obligated to pay royalties on future net
sales of products that are developed and approved as defined by
this collaboration. Our royalty obligations as to a particular
licensed product will be payable, on a
country-by-country
and licensed
product-by-licensed
product basis, for the longer of (a) the date of expiration
of the last to expire valid claim within the licensed patents
that covers the manufacture, use or sale, offer to sell, or
import of such licensed product by us or a sublicensee in such
country, or (b) 10 years after the first commercial
sale of the applicable licensed product in the applicable
country.
62
Funding
Requirements
We expect to incur substantial expenses and generate significant
operating losses as we continue to advance our product
candidates into preclinical studies and clinical studies and as
we:
|
|
|
|
|
continue clinical development of the Phase 3 VISTA-16 study for
varespladib;
|
|
|
|
continue clinical development of the Phase 2b PEARL-SC study for
A-623;
|
|
|
|
hire additional clinical, scientific and management
personnel; and
|
|
|
|
implement new operational, financial and management information
systems.
|
Our future capital uses and requirements depend on numerous
forward-looking factors. These factors include the following:
|
|
|
|
|
the progress of preclinical development and clinical studies of
our product candidates;
|
|
|
|
the time and costs involved in obtaining regulatory approvals;
|
|
|
|
delays that may be caused by evolving requirements of regulatory
agencies;
|
|
|
|
the costs involved in filing and prosecuting patent applications
and enforcing or defending patent claims;
|
|
|
|
our ability to establish, enforce and maintain selected
strategic alliances; and
|
|
|
|
the acquisition of technologies, product candidates and other
business opportunities that require financial commitments.
|
To date, we have not generated any revenue. We do not expect to
generate revenue unless or until we obtain regulatory approval
of, and commercialize, our product candidates. We expect our
continuing operating losses to result in increases in cash used
in operations over the next several years. Our future capital
requirements will depend on a number of factors including the
progress and results of our clinical studies, the costs, timing
and outcome of regulatory review of our product candidates, our
revenue, if any, from successful development and
commercialization of our product candidates, the costs of
commercialization activities, the scope, progress, results and
costs of preclinical development, laboratory testing and
clinical studies for other product candidates, the emergence of
competing therapies and other market developments, the costs of
preparing, filing and prosecuting patent applications and
maintaining, enforcing and defending intellectual property
rights, the extent to which we acquire or invest in other
product candidates and technologies, and our ability to
establish collaborations and obtain milestone, royalty or other
payments from any collaborators.
We expect our existing resources as of the date of this
prospectus, to be sufficient to fund our planned operations,
including our continued product candidate development, for at
least the next 12 months. However, we may require
significant additional funds earlier than we currently expect to
conduct additional or extended clinical studies and seek
regulatory approval of our product candidates. Because of the
numerous risks and uncertainties associated with the development
and commercialization of our product candidates, we are unable
to estimate the amounts of increased capital outlays and
operating expenditures associated with our current and
anticipated clinical studies.
63
Additional funding may not be available to us on acceptable
terms or at all. In addition, the terms of any financing may
adversely affect the holdings or the rights of our stockholders.
For example, if we raise additional funds by issuing equity
securities or by selling debt securities, if convertible,
further dilution to our existing stockholders may result. To the
extent our capital resources are insufficient to meet our future
capital requirements, we will need to finance our future cash
needs through public or private equity offerings, collaboration
agreements, debt financings or licensing arrangements.
If adequate funds are not available, we may be required to
terminate, significantly modify or delay our development
programs, reduce our planned commercialization efforts, or
obtain funds through collaborators that may require us to
relinquish rights to our technologies or product candidates that
we might otherwise seek to develop or commercialize
independently. We may elect to raise additional funds even
before we need them if the conditions for raising capital are
favorable.
Off-Balance
Sheet Arrangements
We do not currently have, nor have we ever had, any
relationships with unconsolidated entities or financial
partnerships, such as entities often referred to as structured
finance or special purpose entities, established for the purpose
of facilitating off-balance sheet arrangements or other
contractually narrow or limited purposes. In addition, we do not
engage in trading activities involving non-exchange traded
contracts.
Quantitative
and Qualitative Disclosure About Market Risk
Our primary exposure to market risk is interest income
sensitivity, which is affected by changes in the general level
of U.S. interest rates. However, since a majority of our
investments are in short-term certificates of deposit and money
market funds, we do not believe we are subject to any material
market risk exposure. We do not have any foreign currency or any
other material derivative financial instruments.
Recent
Accounting Pronouncements
In June 2009, the FASB issued FASB ASC 105, Generally
Accepted Accounting Principles, that establishes the FASB
Accounting Standards Codification as the sole source of
Generally Accepted Accounting Principles, or GAAP. Pursuant to
the provisions of FASB ASC 105, we have updated references
to GAAP in our financial statements issued for the period ending
December 31, 2009 and thereafter. The adoption of FASB
ASC 105 had no impact on our financial position or results
of operations.
In June 2008, the FASB issued FASB
ASC 815-40,
Derivatives and Hedging. FASB
ASC 815-40
provides guidance on how to determine if certain instruments (or
embedded features) are considered indexed to a companys
own stock, including instruments similar to warrants to purchase
the companys stock. FASB
ASC 815-40
requires companies to use a two-step approach to evaluate an
instruments contingent exercise provisions and settlement
provisions in determining whether the instrument is considered
to be indexed to its own stock and therefore exempt from the
application of FASB ASC 815. Although FASB
ASC 815-40
is effective for fiscal years beginning after December 15,
2008, any outstanding instrument at the date of adoption will
require a retrospective application of the accounting through a
cumulative effect adjustment to retained earnings upon adoption.
We do not expect the adoption of FASB
ASC 815-40
to have a material impact on either our financial position or
results of operations.
64
BUSINESS
Overview
We are a biopharmaceutical company focused on developing and
commercializing products to treat serious diseases associated
with inflammation, including cardiovascular and autoimmune
diseases. We currently have one Phase 3 clinical program,
varespladib, and two Phase 2 clinical programs,
A-623 and
A-001. Two
of our product candidates, varespladib and
A-001, are
designed to inhibit a novel enzyme target known as secretory
phospholipase
A2,
or
sPLA2.
Elevated levels of
sPLA2
have been implicated in a variety of acute inflammatory
conditions, including acute coronary syndrome and acute chest
syndrome associated with sickle cell disease, as well as in
chronic diseases, including stable coronary artery disease, or
CAD. In addition, our Phase 2 product candidate,
A-623,
targets elevated levels of
B-lymphocyte
stimulator, or BLyS, also known as B-Cell Activating Factor, or
BAFF, which has been associated with a variety of B-cell
mediated autoimmune diseases, including systemic lupus
erythematosus, or lupus, lupus nephritis, or LN, rheumatoid
arthritis, multiple sclerosis, Sjögrens Syndrome,
Graves Disease and others.
Product
Development Programs
We have worldwide rights to develop and commercialize our
products in all indications and markets, with the exception of
Japan where Shionogi & Co., Ltd. retains commercial
rights to our
sPLA2
product candidates. Our current development plans are focused on
acute treatment and orphan indications that may provide an
accelerated and cost-efficient path to regulatory approval and
commercialization. We believe that certain of these markets can
be commercialized through a limited specialty sales force. In
addition, we believe that our product candidates can also
address market opportunities in chronic indications and we may
seek development and commercialization partners to address
chronic, non-specialty and international markets.
Inflammation
and Diseases
The inflammatory process is a powerful and essential early line
of defense for protection against injury and to repair body
tissue. As a result, it is tightly regulated by the body to
ensure appropriate activation and prompt resolution. However,
under certain circumstances, the normal process can malfunction,
leading to acute or chronic inflammation or inappropriate
activation directed against the bodys own tissues. All of
these circumstances can cause significant damage to cells and
tissues, leading to a range of inflammatory disorders, such as
cardiovascular and autoimmune diseases.
65
Our
sPLA2
Inhibition Portfolio
Building upon our knowledge of the regulation of inflammatory
pathways and the growing body of evidence that links
inflammation to multiple disease states, we believe that we have
developed a leadership position in the field of
sPLA2
inhibition. Our
sPLA2
inhibitors have been studied in a number of inflammatory
disorders in multiple therapeutic areas. The effect of our
sPLA2
inhibitors on
sPLA2
concentration and activity have been implicated in acute
coronary syndrome and acute chest syndrome associated with
sickle cell disease. We currently have the two most advanced
sPLA2
inhibitors in clinical development.
Our lead product candidate, varespladib (an oral prodrug of
A-001), is a
broad-spectrum inhibitor of
sPLA2
enzymes and is being evaluated in a Phase 3 clinical study for
short-term (16-week) treatment of patients who have experienced
an acute coronary syndrome. The American Heart Association
defines acute coronary syndrome as any group of clinical
symptoms related to acute myocardial ischemia, including
unstable angina, or UA. Varespladib, when combined with Lipitor
(atorvastatin), is one of only a few therapeutics in development
with the potential to offer a unique and synergistic treatment
approach targeting inflammation, elevated lipid levels and
atherosclerosis as part of physician-directed standard of care.
Through its novel mechanism of action, varespladib may have
applications in a broad range of acute and chronic
cardiovascular diseases. Based on the successful results of our
recently completed Phase 2b clinical study, we initiated a Phase
3 clinical study, VISTA-16, in patients with acute coronary
syndrome in June 2010.
Our second product candidate, varespladib sodium,
A-001, is an
intravenously administered inhibitor of
sPLA2,which
is in a Phase 2 clinical study for the prevention of acute chest
syndrome associated with sickle cell disease. Acute chest
syndrome is a form of inflammation-induced lung failure and is
the most common cause of death in patients with sickle cell
disease. Given that there are currently no approved drugs for
the prevention of acute chest syndrome associated with sickle
cell disease, we have received orphan drug designation and fast
track status from the FDA for
A-001.
We also have a broad series of additional
sPLA2
inhibitors designed with distinct chemical scaffolds in
preclinical development. These product candidates are intended
to provide new
sPLA2
inhibitors for our existing target indications as well as new
candidates for other therapeutic areas. Our lead candidate
within the series,
A-003, is
chemically distinct from
A-001 and
varespladib and has shown increased potency against the target
enzymes and higher drug exposure after dosing in preclinical
studies. As a result,
A-003 may
confer beneficial pharmacodynamic effects in patients and can be
formulated for oral or intravenously administered use. We plan
to file an investigational new drug application, or IND, for
A-003 in the
future and we may continue to assess additional new compounds.
We have explored the use of our varespladib and
A-001
sPLA2
inhibitors as both topical and inhalation therapies in animal
models for the treatment of atopic dermatitis and asthma,
respectively. Results from a standard mouse model of edema
demonstrated that topically administered varespladib was
equivalent to the marketed immunosuppressant Elidel in resolving
inflammation. In a sheep model of allergen-induced asthma,
inhaled
A-001
demonstrated an improvement in lung function similar to inhaled
steroids.
sPLA2
Biology
sPLA2
is a family of enzymes directly involved in the acute and
chronic steps of an inflammatory response.
sPLA2
activity is highly elevated during the early stages of
inflammation, and its acute effects serve to substantially
amplify the inflammatory process. The
sPLA2
enzyme catalyzes the first step in the arachidonic acid pathway
of inflammation, one of the main metabolic processes for the
production of inflammatory mediators, which, when amplified, are
responsible for causing damage to cells and tissue.
Specifically,
sPLA2
breaks down phospholipids that result in the formation of fatty
acids such as
66
arachidonic acid. Arachidonic acid is subsequently metabolized
to form several pro-inflammatory and thrombogenic molecules.
In cardiovascular diseases such as acute coronary syndrome,
excess
sPLA2
activity has acute and chronic implications on disease
progression and patient outcomes. In published studies and our
own clinical studies, significant elevations in
sPLA2
activity and mass have been seen from 24 hours to two weeks
following an acute coronary syndrome and can persist for up to
an additional 12 weeks thereafter. Shortly after a heart
attack,
sPLA2
is dramatically elevated, amplifying inflammation that is
associated with more frequent and secondary cardiovascular
events. This resulting elevated level of inflammation is
problematic for acute coronary syndrome patients who are already
at higher risk of complications during the weeks following their
initial event. For example, increased inflammation can
destabilize vulnerable vascular lesions or atherosclerotic
plaque, destroy damaged but viable cardiac cells and adversely
modify lipids, any of which may lead to the recurrence of a
major adverse cardiovascular event, or MACE.
Historical and recent clinical results have demonstrated
circulating levels of
sPLA2
are significantly correlated with a well-established
inflammatory marker, C-reactive protein, or CRP. These and other
clinical studies have also demonstrated that
sPLA2
independently predicts coronary events in patients that have
recently experienced an acute coronary syndrome and patients
with stable CAD independent of other standard risk factors. In a
stable cardiovascular patient,
sPLA2
not only sustains chronic vascular inflammation as discussed
earlier, but it also adversely remodels lipoproteins such as
low-density lipoprotein cholesterol, or LDL-C.
sPLA2
interacts with LDL-C in a series of reactions that result in
smaller, more pro-atherogenic and pro-inflammatory LDL-C
particles. Moreover, these modified lipoproteins have a reduced
affinity for LDL-C receptors, which are responsible for removal
of cholesterol from the body. As a result, LDL-C remains in
circulation longer and has a greater tendency to deposit in the
artery wall. This increased LDL-C deposition and sustained
chronic vascular inflammation may contribute to the development
of atherosclerosis.
The family of
sPLA2
enzymes includes at least three forms that play a role in
inflammation and the development of cardiovascular disease or
lung injury. While
sPLA2
enzymes are a member of the phospholipase family that includes a
lipoprotein associated phospholipase
A2,
or
Lp-PLA2,
there are important distinctions. Although both are present in
blood,
Lp-PLA2
is mostly bound to LDL-C and high-density lipoprotein, or HDL,
while
sPLA2
enzymes are not. Based on our clinical studies, we believe that
our
sPLA2
inhibitor, varespladib, can be distinguished from other
PLA2
enzyme inhibitors such as those targeted at inhibiting
Lp-PLA2
because varespladib treatment:
|
|
|
|
|
is synergistic with HMG-CoA reductase inhibitors, or statins,
including Lipitor (atorvastatin), in reducing LDL-C, total
cholesterol and non-HDL cholesterol in patients with CAD;
|
|
|
|
lowers circulating small, dense and pro-atherogenic, or
plaque-building LDL-C particles, while
Lp-PLA2
inhibition has not demonstrated similar effects;
|
|
|
|
has been shown to lower CRP, a well-established marker of
inflammation in a statistically significant manner; and
|
|
|
|
reduces plaque volume and aneurysms in standard rodent models of
atherosclerosis and has demonstrated synergistic reductions of
plaque volume in standard rodent models of atherosclerosis when
used in combination with statins.
|
In diseases such as acute chest syndrome, a very serious form of
lung injury associated with sickle cell disease,
sPLA2
acts acutely on a number of substrates that amplify the
inflammatory disease process. Sickle cell disease is a genetic
disorder which leads to the structural alteration, or
sickling, of
67
otherwise healthy red blood cells. Patients with sickle cell
disease experience periods of intense pain known as
vaso-occlusive crisis, or VOC, as structurally altered red blood
cells bind together and occlude small blood vessels that supply
blood and nutrients to vital tissue and bone.
sPLA2
levels are dramatically elevated in sickle cell patients during
an episode of VOC as well as within 24 to 48 hours of the
onset of acute chest syndrome. During VOC, microscopic fat
emboli, or droplets of fat from the bone marrow, are prevalent
and can break free and become lodged in the lung. These emboli
are substrates for
sPLA2
enzymes and provide fuel for an already established inflammatory
response, increasing lung injury. In addition,
sPLA2
has been demonstrated to degrade human lung surfactant, a
component necessary in maintaining appropriate lung function,
which further complicates lung injury.
We believe that early intervention with a drug designed to
inhibit
sPLA2
activity may offer a unique opportunity to reduce the
complications associated with certain inflammatory diseases such
as acute coronary syndrome in cardiovascular patients and acute
chest syndrome in patients with sickle cell disease.
Our
BAFF Antagonism Portfolio
BAFF has been associated with a wide range of B-cell mediated
autoimmune diseases including lupus, LN, rheumatoid arthritis,
multiple sclerosis, Sjögrens Syndrome, Graves
Disease and others. The role of BAFF in lupus and rheumatoid
arthritis has recently been validated in multiple clinical
studies with other BAFF antagonists. We are advancing the
development of our BAFF inhibitor molecule,
A-623, a
selective peptibody, to exploit its broad potential clinical
utility in autoimmune diseases. A peptibody is a novel fusion
protein that is distinct from an antibody. We have worldwide
rights to
A-623 in all
potential indications. We have initiated PEARL-SC, the Phase 2b
clinical study of
A-623, for
the treatment of Systemic Lupus Erythematosus (lupus). Lupus
patients suffer from a chronic autoimmune disease, which often
leads to severe skin rash, fatigue, joint pain, major organ
complications and cardiovascular disease.
A-623
demonstrates anti-BAFF activity and has shown statistically
significant reductions in B-cells in two Phase 1 clinical
studies in patients with lupus. We believe
A-623 may
offer a number of potential differentiations over other BAFF
antagonists, as well as other novel B-cell directed therapies
including:
|
|
|
|
|
convenient, at-home, patient-administered subcutaneous dosing
with a range of dosing frequencies including monthly and weekly;
|
|
|
|
the ability to bind to both membrane-bound and soluble BAFF;
|
|
|
|
|
|
selective modulation and reduction of relevant B-cell
sub-types in
lupus patients;
|
|
|
|
|
|
a novel molecular structure, which may confer differentiating
pharmacokinetic and pharmacodynamic characteristics, potentially
providing efficacy and dosing benefits, as well as manufacturing
benefits and lower cost of goods based on a bacterial
fermentation manufacturing process; and
|
|
|
|
|
|
multiple binding domains achieve highest reported affinity for
inhibition of BAFF.
|
68
Product
Development Programs
We have focused our product development programs on
anti-inflammatory therapeutics for cardiovascular diseases,
lupus and other serious diseases for which we believe that
current treatments are either inadequate or non-existent. Our
current product development programs are listed in the table
below.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Worldwide
|
|
|
|
|
|
|
|
|
Development
|
|
|
Product
|
|
|
|
|
|
Product Candidate
|
|
|
Phase
|
|
|
Rights
|
|
|
Description
|
|
Next Milestone(s)
|
Lead Development Programs
|
|
|
|
|
|
|
|
|
|
|
|
|
Varespladib with Lipitor (atorvastatin)
|
|
|
Phase 3
|
|
|
Anthera(1)
|
|
|
Orally administered sPLA2 inhibitor
Indicated for the prevention of secondary MACE following an acute coronary syndrome (16-week treatment)
|
|
1000 patient biomarker futility analysis in the first quarter of 2011
Data Safety Monitoring Board, or DSMB, review of clinical data in the first quarter of 2011
|
|
|
|
|
|
|
|
|
|
|
|
|
A-623
|
|
|
Phase 2b
|
|
|
Anthera
|
|
|
Selective peptibody antagonist of BAFF cytokine being developed for the treatment of B-cell mediated autoimmune diseases
Indicated for systemic lupus erythematosus
|
|
Selection of bulk drug contract manufacturer in the fourth quarter of 2010
Completion of technology transfer to contract manufacturer in the first quarter of 2011
Safety and biomarker review in the second quarter of 2011
Initiation of Phase 3 manufacturing campaign from cell bank in the second quarter of 2011
|
|
|
|
|
|
|
|
|
|
|
|
|
Additional Programs
|
|
|
|
|
|
|
|
|
|
|
|
|
A-001-varespladib
sodium
|
|
|
Phase 2
|
|
|
Anthera(1)
|
|
|
Intravenous sPLA2 inhibitor with orphan drug and fast track status
Indicated for prevention of acute chest syndrome in hospitalized patients with sickle cell disease
|
|
Publication of IMPACTS data
Submission of IMPACTS-2 protocol to FDA
|
|
|
|
|
|
|
|
|
|
|
|
|
Varespladib
|
|
|
Phase 2
investigator study
|
|
|
Anthera(1)
|
|
|
Orally administered
sPLA2
inhibitor to reduce inflammatory markers in patients undergoing
interventional cardiovascular procedures
|
|
Enrollment complete. Data publication targeted in
2011
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
(1) |
Shionogi & Co., Ltd. retains product rights in Japan
|
Varespladib
Varespladib is an orally administered pro-drug of
A-001, which
is a broad-spectrum, once-daily inhibitor of the IIa, V and X
iso-forms of the
sPLA2
enzyme that has demonstrated potent anti-inflammatory,
lipid-lowering and lipid-modulating treatment effects in
multiple clinical studies. We have commenced the Phase 3
VISTA-16 study to evaluate varespladib in combination with
atorvastatin therapy, specifically Lipitor, for the short-term
(16-week) treatment of acute coronary syndrome. We have an
agreement with the FDA on an SPA for the VISTA-16 study. An SPA
provides an opportunity for the clinical study sponsor to
receive feedback from the FDA regarding the adequacy of a
clinical study to meet regulatory and scientific requirements if
conducted in accordance with the SPA agreement.
69
An SPA is not a guarantee of an approval of a product candidate
or any permissible claims about the product candidate.
To date, over 1,000 patients and healthy volunteers in at
least 15 clinical studies have been exposed to varespladib.
Varespladib was generally well-tolerated in studies where
patients were exposed to a maximum of 48 weeks of therapy.
Varespladib has been studied in combination with Lipitor
(atorvastatin) in a Phase 2b clinical study in acute coronary
syndrome patients and two earlier Phase 2 clinical studies in
stable CAD patients, the majority of whom were on various statin
therapies.
We currently have all worldwide product rights to varespladib,
except in Japan where Shionogi & Co., Ltd. retains
rights. We originally licensed our
sPLA2
inhibitor portfolio, including varespladib and
A-001, from
Eli Lilly & Company, or Eli Lilly, and
Shionogi & Co., Ltd. in July 2006.
Market
Opportunity Acute Coronary Syndrome
According to the American Heart Association, over
18 million people in the United States have experienced an
acute coronary syndrome and an estimated 1.5 million
Americans will have a new or recurrent heart attack. In
addition, the American Heart Association estimates that
worldwide, cardiovascular disease kills an estimated
17.5 million people each year. According to British Heart
Foundation statistics, CAD, which often leads to acute coronary
syndrome or heart attacks, accounts for 1.9 million deaths
in Europe annually. According to the World Health Organization,
or the WHO, cardiovascular disease is the most common cause of
death in the western world and a major cause of hospital
admissions. In addition, the American Heart Association provides
that for people over the age of 40, 20% of them will die within
one year following an initial heart attack, and over one-third
of them will die within the first five years of an initial heart
attack. These numbers are expected to increase given an aging
population, as well as the rising epidemics of diabetes and
obesity, two conditions known to increase the risk of acute
coronary syndrome.
The American Heart Association defines acute coronary syndrome
as any group of clinical signs and symptoms related to acute
myocardial ischemia. Acute myocardial ischemia can often present
as chest pain due to insufficient blood supply to the heart
muscle that results from CAD. Acute coronary syndrome covers a
spectrum of clinical conditions that include ST-elevated
myocardial infarction, or STEMI, non-ST-elevated myocardial
infarction, or NSTEMI, and UA. Both STEMI and NSTEMI are forms
of a heart attack, where damage to the heart muscle occurs due
to ischemia, which is lack of blood flow to tissues due to a
blockage of a vessel. Typically, UA results in chest pain from
ischemia, but does not cause permanent damage to the heart
muscle.
Furthermore, for any patient who experiences an acute coronary
syndrome, the risk of a secondary MACE is significantly
increased immediately following the initial event. Large
clinical outcome studies such as MIRACL and PROVE-IT have
previously reported, and data from our own FRANCIS Phase 2b
clinical study supports, the 16-week rate of secondary MACE in
acute coronary syndrome patients to be between 6.1% and 14.8%.
Current treatments for CAD other than interventional procedures
include a variety of medications such as aspirin, statins and
anti-platelet and anti-coagulant therapeutics. These medications
are used to offer both acute and chronic benefits to patients.
For patients presenting with acute coronary syndrome,
therapeutics are administered quickly to improve blood flow to
the heart and limit the risk associated with continued ischemia
and thrombosis, which is the formation of a blood clot inside a
vessel, which obstructs blood flow. In addition, interventional
procedures and other medications, such as statins that are
initiated early primarily for lipid benefits, are continued in
an attempt to provide chronic protection against secondary MACE
through improvement in lipid profiles such as lowering LDL-C.
70
Inflammation
in Cardiovascular Disease
In patients experiencing an acute coronary syndrome, the
relationship between higher levels of inflammation, as measured
by CRP,
sPLA2
and interleukin-6, or IL-6, and increased risk for MACE has been
demonstrated extensively. In numerous clinical studies with a
variety of therapeutic interventions, reductions in CRP have
been correlated with reductions in subsequent MACE. We believe,
if our Phase 3 pivotal study is successful, that varespladib
would represent the first anti-inflammatory therapeutic approved
for prevention of MACE.
CRP is the most commonly used marker of inflammation. It has
been independently and strongly correlated with adverse
cardiovascular outcomes in multiple clinical studies. Although a
causative role for CRP has not been established, inflammation is
known to promote acute coronary syndrome and CRP may play a
direct role in both vascular inflammation as well as plaque
rupture.
Statins reduce the level of CRP and other markers of
inflammation in patients with stable CAD. In April 2001, the
Journal of the American Medical Association published results
from the MIRACL study describing the effect of statins in acute
coronary syndrome, where inflammation is greatly elevated. 3,086
were randomized within 96 hours of their index event to
treatment with high-dose Lipitor (atorvastatin) or placebo.
Lipitor (atorvastatin) significantly reduced secondary MACE
after 16 weeks. A second paper from the same study,
published in Circulation in 2003, described the rapid decline of
inflammatory markers in patients on statin treatment that was
associated with reduced MACE. After 16 weeks, Lipitor
(atorvastatin) reduced CRP levels by 34%.
More recently, in 2005, the New England Journal of Medicine
published data from the PROVE-IT study. A total of
3,745 patients were randomized to either intensive statin
therapy with 80 mg Lipitor (atorvastatin) or moderate
statin therapy with 40 mg pravastatin. Patients with low
CRP or LDL-C had fewer MACE than those with higher levels of
either CRP or LDL-C. Patients who had both LDL-C
< 70 mg/dL and CRP < 1 mg/L had the
fewest number of secondary events over all.
LDL-C
in Cardiovascular Disease
The direct relationship between lower LDL-C levels and reduced
risk for major cardiovascular events has been consistently
demonstrated for over a decade in 18 outcome studies involving
over 119,000 patients. Results from large clinical outcome
studies demonstrate achieving incrementally lower LDL-C levels
reduces the risk of future cardiovascular events and provides
continued patient benefit. As a result, the lipid treatment
guidelines have been revised to establish more aggressive LDL-C
treatment goals over time. The most recent guidelines from the
National Cholesterol Education Programs Adult Treatment
Panel III, or NCEP ATP III, updated in 2004 advocate treatment
goals for LDL-C below 100 mg/dL for high-risk patients and
70 mg/dL for very high-risk patients. Given the breadth of
more recent clinical data available, we believe that future
treatment guidelines from the NCEP will likely establish new
LDL-C treatment goals that apply the 70 mg/dL standard or
lower to a broader population of at risk patients. Patients
enrolled in our FRANCIS Phase 2b clinical study and our planned
Phase 3 acute coronary syndrome study represent high-risk
patients as defined by the NCEP.
In order to achieve these more aggressive LDL-C targets, doctors
prescribe other approved lipid-lowering therapies such as
cholesterol absorption inhibitors, nicotinic acid and fish oils
in combination with statins to further reduce LDL-C. Still, many
acute coronary syndrome patients who represent the NCEP ATP III
guideline categories of high-risk and very high-risk do not
achieve these recommended lipid goals despite maximum
lipid-lowering therapies. Moreover, substantial residual risk
remains even among the group of patients that do achieve these
aggressive LDL-C goals suggesting additional biological
mechanisms, including inflammation, may be relevant.
71
This is exemplified in a November 2008 publication in the New
England Journal of Medicine that detailed the results from a
17,000 patient, multinational, primary prevention study
named JUPITER. The study randomized patients with relatively
normal levels of LDL-C, but elevated levels of inflammation
based on CRP to statin or placebo therapy. The JUPITER study was
stopped early because those patients randomized to statin
therapy demonstrated a statistically significant reduction in
CRP, which also translated to a statistically significant
reduction in cardiovascular events versus those on placebo. The
reduction in events was well in excess of that which would be
predicted from historical data evaluating LDL-C reductions
alone. While these results were generated in a primary
prevention setting, we believe that the benefits of reducing
inflammation may prove to be even more meaningful in settings
where patients are in a hyper-inflammatory state, such as
following an acute coronary syndrome. As a result of these
studies, we believe that there is a substantial need for novel
therapies that provide meaningful reductions in inflammation
while also improving LDL-C levels in high-risk cardiovascular
patients beyond the benefits of statin therapy. Therefore, it is
our belief that targeting inflammation and elevated LDL-C with
sPLA2
inhibition during the early phase of an acute coronary syndrome
will further improve patient outcomes.
We believe that varespladib is one of only a few novel drugs in
development with the potential to offer a clinical benefit to
high-risk cardiovascular patients. Varespladibs unique
mechanism provides potent anti-inflammatory activity, as
measured by reductions in
sPLA2,
CRP and IL-6; incremental lipid-lowering, as measured by LDL-C;
and lipid-modulating activity beyond that achievable with statin
therapy alone. Furthermore, because of their complementary
mechanisms, we believe that the combination of statins and
varespladib can provide synergistic anti-inflammatory and
lipid-lowering benefits. We also have preliminary data to
suggest that varespladib may be synergistic with other
cardiovascular therapeutic regimens, such as niacin.
Pivotal
VISTA-16 Study Acute Coronary Syndrome
In February 2008, based on the results from Phase 2 stable CAD
studies, as discussed below, we met with the FDA to discuss the
next steps of clinical development of varespladib during our end
of Phase 2 meeting. As a result of that meeting and the results
from our Phase 2b acute coronary syndrome study, we submitted an
SPA to the FDA for the Phase 3 VISTA-16 study of varespladib for
the short-term
(16-week)
treatment of patients who have recently experienced an acute
coronary syndrome. We reached agreement with the FDA on all
aspects of the VISTA-16 study protocol, including patient
inclusion/exclusion criteria, study size, statistical
considerations, efficacy endpoints, study duration,
randomization and lipid management strategies.
An independent DSMB will continually evaluate the performance of
the VISTA-16 study over time to ensure patient safety. In
addition, after a minimum of 1,000 patients have been
enrolled in the VISTA-16 study, an independent committee not
involved with the VISTA-16 study will complete a biomarker
futility analysis to ensure patient levels of inflammation, as
measured by
sPLA2,
CRP and IL-6, and lipid profiles, as measured by LDL-C, have met
pre-specified reductions from baseline at various time-points.
These markers of inflammation and lipid profiles are
well-established in the clinical community and pharmaceutical
industry as independent predictors of future cardiovascular risk
and, if positive, will provide additional validation of our
previous findings from the FRANCIS Phase 2b clinical study. At
the
72
same time, the independent DSMB will review all clinical data
from the VISTA-16 study to ensure no emergent adverse safety
signal.
In June 2010, we initiated enrollment in the VISTA-16 clinical
study. Pursuant to our SPA agreement with the FDA, our
multinational, randomized, double-blind, placebo-controlled
Phase 3 acute coronary syndrome VISTA-16 study will enroll up to
6,500 patients in up to 15 countries and up to 500 centers.
However, enrollment may be stopped anytime after a minimum of
385 adjudicated endpoint events as described in the protocol
have occurred. This number of events will allow us to detect a
treatment effect on the composite endpoint as low as 18.1% with
a p-value of less than 0.05. We may increase the sample size if
the adjudicated endpoint events occur at a lower rate than we
expect. Patients will be randomized at entry to receive
16 weeks of either 500 mg once-daily of varespladib or
placebo in addition to a 20, 40 or 80 milligram dose of Lipitor
(atorvastatin). The dose of Lipitor (atorvastatin) may be
adjusted after eight weeks if the patients LDL-C level
remains above 100mg/dL. Survival status will be obtained for
patients six months after the completion of dosing. The clinical
study will recruit a similar population of high-risk
cardiovascular patients with acute coronary syndrome to those
enrolled in the FRANCIS study. As in FRANCIS, randomization must
occur within 96 hours of hospitalization for the acute
coronary syndrome event, or if already hospitalized, within
96 hours of event diagnosis. Patient blood chemistry will
be evaluated at baseline, 24 hours and weeks one, two,
four, eight and 16. Randomization is being stratified by the
presence or absence of lipid-lowering therapy prior to the index
event as well as the type of acute coronary syndrome event, such
as UA, NSTEMI or STEMI. The number of subjects who undergo
percutaneous coronary intervention following the index event and
prior to randomization will be limited to no more than 55% of
the total patient population.
The primary endpoint of the VISTA-16 study is to determine
whether 16 weeks of once-daily treatment with varespladib
plus a dose of Lipitor (atorvastatin) is superior to placebo
plus Lipitor (atorvastatin) in
73
the time to the first occurrence of the combined endpoint of
cardiovascular death, non-fatal myocardial infarction, non-fatal
stroke or documented UA with objective evidence of ischemia
requiring hospitalization as defined by recent FDA draft
guidance.
On July 22, 2009 the Center for Drug Education and Research
division of the FDA issued draft recommendations for
standardized definitions for cardiovascular outcomes trials. The
VISTA-16 clinical study endpoint definitions conform to these
guidelines.
Components
of VISTA-16 Primary Endpoint
|
|
|
|
|
Cardiovascular Death
|
|
|
|
Non-Fatal Myocardial Infarction
|
|
|
|
Non-Fatal Stroke
|
|
|
|
Documented UA with Objective Evidence of Ischemia Requiring
Hospitalization
|
A secondary endpoint for the VISTA-16 study is to determine
whether varespladib plus a dose of Lipitor (atorvastatin) is
superior to placebo plus Lipitor (atorvastatin) in the time to
the first occurrence of the combined endpoint of all cause
mortality, non-fatal myocardial infarction, non-fatal stroke or
documented UA with objective evidence of ischemia requiring
hospitalization. A comparison between treatment groups will also
be made for each component of the primary efficacy endpoint.
Additionally, the time to multiple occurrences of any non-fatal
component of the composite primary endpoint will also be
explored. The biomarkers CRP, IL-6, LDL-C and
sPLA2,
will also be evaluated at each time point of the clinical study.
Historical
Clinical Studies
Phase 2b Acute Coronary Syndrome Study FRANCIS
(Fewer Recurrent Acute coronary events with Near-term
Cardiovascular Inflammation Suppression)
In July 2008, we initiated a randomized, double-blind,
placebo-controlled Phase 2b clinical study that enrolled 625
acute coronary syndrome patients across 35 centers in three
countries. Given the drugs combined anti-inflammatory,
lipid-lowering and lipid-modulating effects, we evaluated the
effects of varespladib in acute coronary syndrome patients with
high levels of inflammation and dislipidemia. The clinical study
was designed to evaluate the safety and efficacy of varespladib
when co-administered with the highest dose (80 mg) of
Lipitor (atorvastatin). The clinical study randomized all
patients to a minimum of 24 weeks of treatment with either
500 mg once-daily of varespladib or placebo in combination
with 80 mg Lipitor (atorvastatin) and physician-directed
standard of care.
Patients were eligible for enrollment if they had a diagnosis of
UA, NSTEMI or STEMI. In addition, they must have had one of the
following risk factors: diabetes, body mass index (BMI)
³
25 kg/m2,
³
CRP
³
2 mg/L (NSTEMI/STEMI) or CRP
³
3 mg/L (UA) and presence of three (pre-defined)
characteristics of metabolic syndrome. Subjects must have been
randomized within
£
96 hours of hospital admission for the index event, or, if
already hospitalized, within
£
96 hours of index event diagnosis. Any percutaneous
revascularization was required to occur prior to randomization.
In addition, because we wanted to assess the effects of
varespladib with the highest available dose of Lipitor
(atorvastatin), patients were not allowed to use any other
lipid-lowering therapies during the clinical study.
Follow-up
visits for evaluation occurred post-randomization at weeks two,
four, eight, 12, 16, 20, 24 and then monthly thereafter until
clinical study completion. All enrolled subjects remained on
treatment until all subjects had been treated for a minimum of
24 weeks or until the occurrence of MACE. Patients
randomized into the FRANCIS study had baseline characteristics
such as LDL-C
74
indexed-event risk factors and demographics similar to other
studies of this type. All patients who completed the clinical
study received a final evaluation.
The primary efficacy endpoint evaluated the change in LDL-C
after 500 patients completed eight weeks of treatment.
LDL-C is the most widely recognized surrogate for predicting
cardiovascular risk where percentage reductions in LDL-C have
been highly correlated with reductions in future cardiovascular
risk. Secondary endpoints included:
|
|
|
|
|
changes in established markers of inflammation such as
sPLA2,
CRP and IL-6; and
|
|
|
|
the occurrence of secondary MACE (for purposes of this clinical
study, all-cause mortality, non-fatal myocardial infarction,
documented UA requiring urgent hospitalization,
revascularization occurring
³
60 days post the index event or non-fatal stroke).
|
Results of the primary endpoint demonstrated a statistically
significant incremental LDL-C reduction of 5.7% (p = 0.0023) in
varespladib treated patients versus those treated with
80 mg Lipitor (atorvastatin) alone after eight weeks of
therapy. A p-value is a probability with a value ranging from 0
to 1, which indicates the likelihood that a clinical study is
different between treatment and control groups. P-values below
0.05 are typically referred to as statistically significant. A
statistically significant difference was observed in LDL-C
reduction from baseline as early as two weeks after treatment.
The treatment effect was maintained throughout the observation
period.
Figure 1: Mean Percentage Change in LDL-C from Baseline
Secondary endpoints measured effects of varespladib on
sPLA2,
CRP and IL-6 levels, which are well-established markers of
inflammation. While the FRANCIS study was not designed to
demonstrate statistically significant changes in CRP and IL-6,
the results were consistent with previous studies, which
demonstrated improvement across these biomarkers and achieved
statistical significance at some time points.
sPLA2
concentration was statistically significantly reduced from the
earliest time point of two weeks through the 16-week time point
(p < 0.0001) as compared to high-dose statin (80 mg
Lipitor (atorvastatin)) therapy alone. While our first
sPLA2
measurement in this clinical study occurred at two
75
weeks, data from previous clinical studies utilizing varespladib
or A-001
demonstrated reductions in
sPLA2
as early as two days following treatment.
Figure 2: Median Percentage Change in
sPLA2
Concentration from Baseline
In addition, treatment-related reductions in CRP and IL-6 levels
were also greater in varespladib treated patients compared to
those treated with placebo at all time points in the clinical
study. The percent decrease in CRP at week two was nearly
two-fold greater among varespladib and 80 mg Lipitor
(atorvastatin) treated patients than those treated with placebo
and 80 mg Lipitor (atorvastatin) alone (-39% versus -20%, p
= 0.183), and at week 16, the difference between treatment
groups was statistically significant (-82% versus -73%, p =
0.0067). At weeks two, four, eight and 16, varespladib treated
patients had numerically reduced levels of CRP versus patients
treated with placebo.
Figure 3: Median Percentage Change in CRP Concentration from
Baseline
The percent decrease in IL-6 in patients on varespladib at week
two was more than three times the reduction in IL-6 in patients
on placebo (-18% versus -5.1%, p = 0.18).
76
Figure 4: Median Percentage Change in IL-6 Concentration from
Baseline
Treatment with varespladib resulted in more subjects with LDL-C
levels lower than 70 mg/dL and lower than 50 mg/dL
than those on placebo (80 mg Lipitor (atorvastatin) and
physician-directed standard of care) alone at eight, 16 and
24 weeks of treatment. As discussed above, the NCEP ATP III
guidelines have established an LDL-C of 70 mg/dL as an
optional target for very high-risk patients. As indicated in the
table below, the data suggests varespladib treatment helps
patients achieve their LDL-C target levels more quickly and
maintain them longer than with high-dose statin (80 mg
Lipitor (atorvastatin)) therapy alone.
Finally, given the importance of reducing inflammation as well
as LDL-C following an acute coronary syndrome event, we examined
the proportion of patients in the clinical study that were able
to achieve both LDL-C levels less than 70 mg/dL and CRP
levels below 1 mg/L. As indicated in the figure below,
significantly more patients at week four and week 16 (p = 0.02
and p = 0.01) reached this combined target when treated with
varespladib and 80 mg Lipitor (atorvastatin) than with
placebo and 80 mg Lipitor (atorvastatin) alone. (The actual
proportion of subjects in the varespladib group was 25% and 16%
in the placebo group). Additionally, in the PROVE-IT study a
comparable proportion (16%) of patients treated with 80 mg
Lipitor (atorvastatin) achieved these goals.
77
Figure 5: Percentage of Patients with LDL-C <
70 mg/dL, < 50mg/dL and Achieving Combined Targets of
CRP < 1.0 mg/L and LDL-C < 70 mg/dL
We also conducted an exploratory analysis of MACE in the
clinical study. At 16 weeks, there were 14 (4.2%) MACE in
the varespladib treated group as compared to 19 (6.1%) in the
placebo group. At the completion of the clinical study, all
patients had received at least six months of therapy and there
were 23 (7.4%) MACE in the varespladib treated group as compared
to 24 (7.7%) MACE in the placebo group. While the MACE analysis
was not designed to demonstrate any statistical differences
between the two treatment groups, we believe that the results
are encouraging and will help us to design our VISTA-16 study.
Overall, varespladib was generally well-tolerated in this
clinical study and no imbalance was seen in dropouts due to drug
effects. After completing patient treatment, overall exposure to
varespladib was a mean of 30 weeks and median of
34 weeks. In total, 485 total patients completed six months
of treatment, with 167 subjects completing 40 weeks and 70
completing 44 weeks. There was no imbalance of overall
adverse events between the treatment arms. During the clinical
study, at week four and week eight, occasional mild and
transient elevations in liver enzymes, defined as elevations
three times the upper limit of normal, were seen among more
patients taking varespladib, but the frequency and magnitude of
the elevations were not meaningfully different between the
active and control groups at the end of the clinical study. The
frequency of the elevations was also similar to that reported
for Lipitor (atorvastatin) and other currently approved
lipid-lowering agents. Furthermore, there were no effects on
blood pressure or the QT interval, an electro-cardiographic
safety endpoint.
Summary data from FRANCIS was presented at the American College
of Cardiology meeting in 2010 and we anticipate publishing
detailed results from the study in 2010 in a scientific journal.
78
Phase 2 Stable Coronary Artery Disease Study
PLASMA (Phospholipase Levels and Serological Markers of
Atherosclerosis): Varespladib Twice-Daily Versus Placebo
Our Phase 2 PLASMA study was designed to confirm the safety and
effect of varespladib on
sPLA2
concentration, other inflammatory biomarkers and lipids in
patients with stable CAD. In October 2007, we completed a
randomized, double-blind, placebo-controlled study evaluating
four doses of varespladib administered twice-daily versus
placebo among 396 patients with stable CAD from 38 centers
in two countries. The clinical study enrolled patients more than
12 weeks after a myocardial infarction or six weeks after
an episode of UA. The varespladib doses tested were 50 mg,
100 mg, 250 mg and 500 mg administered twice per
day. Following randomization, patients were treated for eight
weeks and safety and efficacy evaluations were conducted at
weeks two, four and eight. Physician-directed standard of care
therapies were permitted during the clinical study, including
259 patients who were on background statin therapy.
The primary endpoint of the clinical study was the change in
sPLA2
concentration from baseline to week eight in varespladib, across
all doses, versus placebo patients. Secondary endpoints in the
clinical study included the change in lipids, including LDL-C,
lipoprotein subclasses and certain inflammatory biomarkers, from
baseline to each of weeks two, four and eight.
Our Phase 2 PLASMA results were selected for a late-breaking
presentation at the American Cardiology Conference and published
in the Lancet journal in February 2009. Results from the
clinical study demonstrated that treatment with varespladib led
to statistically significant reductions in
sPLA2,
LDL-C and various plaque-building and pro-inflammatory forms of
LDL-C. In patients receiving varespladib, there were incremental
reductions in CRP versus placebo (-55.6% versus -24.8%, p =
0.47) from baseline to eight weeks.
Among all patients treated with varespladib, median
sPLA2
concentration decreased by 86.7% from baseline to week eight, as
compared to 4.8% in the placebo group (p < 0.0001). Median
sPLA2
concentration decreased among the varespladib groups in a
dose-dependent manner.
At week eight, across all dosage groups, LDL-C was reduced by
9.7% versus placebo (p = 0.0035). In a subgroup of patients
taking statins with LDL-C > 70 mg/dL, LDL-C was
reduced by 12.0% (p = 0.0065) versus placebo at the
eight week time point. Notably, the reductions in LDL-C appear
to be driven primarily by a shift in the distribution of LDL-C
particles with fewer pro-atherogenic, pro-inflammatory small
LDL-C particles present in the circulation. In addition,
statistically significant reductions from baseline to week eight
were seen in total cholesterol and non-HDL cholesterol in the
overall clinical study population treated with varespladib.
Varespladib was generally well-tolerated among all patients
treated. In general, adverse effects were mild or moderate with
no imbalance of adverse events in the varespladib groups as
compared to placebo. The most common adverse effects seen in the
varespladib groups were headache (6.4%) and nausea (5.4%). There
were mild and transient elevations of liver function tests,
defined as elevations three times the upper limit of normal, in
patients taking varespladib.
Phase 2 Stable Coronary Artery Disease Study
PLASMA-2 (Phospholipase Levels and Serological Markers of
Atherosclerosis -2): Once-Daily of Varespladib Versus
Placebo
Based on data from our first PLASMA study, we initiated a second
Phase 2 clinical study (PLASMA-2) to evaluate the effect of
once-daily varespladib treatment on inflammatory and lipid
biomarkers. In December 2007, we completed a randomized,
double-blind, placebo-controlled Phase 2 clinical study
evaluating two doses of varespladib versus placebo amongst
138 patients with stable CAD. The clinical study, conducted
in the United States, involved 13 clinical sites. Following
randomization to one of two
79
doses of varespladib or placebo, patients were treated for eight
weeks with safety and efficacy evaluations at weeks two, four
and eight. Physician-directed standard of care therapies were
permitted during the clinical study, including 123 patients
(89.1%) who were on background statin therapy.
The primary endpoint of the clinical study was a comparison
between once-daily doses of varespladib and placebo in changes
in
sPLA2
concentration at week eight. Secondary endpoints in the clinical
study included measurements of lipids including LDL-C and
certain other inflammatory biomarkers from baseline to each of
weeks two, four and eight.
Results of the primary endpoint,
sPLA2,
were statistically significant and consistent with those
generated from the first PLASMA study described above. Patients
on varespladib demonstrated a 77.8% reduction in
sPLA2
concentration as compared to an increase of 8.3% in placebo
treated patients (p < 0.0001). Pharmacokinetic data
indicated that once-daily dosing with varespladib would be
sufficient to achieve over 90% inhibition of
sPLA2
mass and activity over a
24-hour
period.
The anti-inflammatory, lipid-lowering and lipid-modulating
effects of varespladib treatment were consistent with those seen
in the first PLASMA study: LDL-C was decreased by 8.3% compared
to 0.7% in placebo (p = 0.014). Due to the small size of this
clinical study, and the low baseline inflammation present in
these patients, no meaningful changes with CRP could be detected
between the active and control groups. As was observed in the
first clinical study, there were statistically significant
reductions from baseline to week eight in total cholesterol and
non-HDL cholesterol in the overall clinical study population
treated with varespladib.
The adverse effect profile for varespladib was consistent with
earlier studies and there was no imbalance of adverse events
among the varespladib groups and placebo. Varespladib was
generally well-tolerated. The most common effects seen in the
varespladib groups were diarrhea (6.7%), nausea (5.6%), any
increase in alanine aminotransferase (5.6%), which is an enzyme
that indicates liver cell injury, and any increase in aspartate
aminotransferase (5.6%), which is another enzyme that indicates
liver cell injury. However, mild and transient elevations of
these liver enzymes, defined as elevations three times the upper
limit of normal, were infrequent in patients taking varespladib.
Table 6: Placebo-corrected Percent Decrease from Baseline to
Week Eight in Biomarkers
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
LDL
|
|
|
Total
|
|
|
Non-HDL
|
|
|
Oxidized
|
|
|
|
sPLA2
|
|
|
Cholesterol
|
|
|
Cholesterol
|
|
|
Cholesterol
|
|
|
LDL-C
|
PLASMA
(All doses varespladib)
|
|
|
81.9%
(p < 0.0001)
|
|
|
9.7%
(p = 0.0035)
|
|
|
4.9%
(p = 0.0069)
|
|
|
7.2%
(p = 0.0009)
|
|
|
5.4%
(p = 0.0065)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
PLASMA-2
(500 mg varespladib)*
|
|
|
86.1%
(p < 0.0001)
|
|
|
13.9%
(p = 0.0007)
|
|
|
9.2%
(p = 0.0006)
|
|
|
14.2%
(p = 0.0001)
|
|
|
7.3%
(pNS)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
* |
Dose selected for Phase 3
|
|
|
|
Probability not significant
|
Investigator-Sponsored Phase 2 Percutaneous Intervention
Study SPIDER-PCI
(sPLA2)
Inhibition to Decrease Enzyme Release After PCI): Varespladib
Once-Daily Versus Placebo for up to
10 Days.
In May 2007, Dr. Vladimir Dzavik at University Health
Network Hospital in Toronto, Ontario, Canada initiated an
investigator sponsored study with varespladib in patients
undergoing a percutaneous intervention, or PCI. The primary
endpoint of this study was to determine if inhibition of
sPLA2
with varespladib will result in a decrease in peri-PCI
myocardial necrosis, or heart muscle damage, as measured by
elevations of myocardial enzyme markers creatine kinase-MB, or
CK-MB, or troponin I. The study was to enroll a maximum of
164 patients who are scheduled to undergo PCI. Elevated
levels of troponin I following PCI are associated with an
increase in in-hospital complications and, in one
80
study, were an independent predictor of major cardiac events.
After PCI, circulating levels of
sPLA2
increase and patients with higher levels have an increased risk
of events after a two-year
follow-up.
This study explores the notion that
sPLA2
inhibition may reduce myocardial damage after PCI and improve
patient outcomes.
As of August 2009, enrollment and dosing in the SPIDER-PCI
investigator study were completed with 144 patients
evaluated for purposes of assessing the primary endpoint. On
December 11, 2009, we received a statistical analysis of
the patient evaluations, which showed that the primary endpoint
of the study, a reduction in the elevation of CK-MB or troponin
I above the upper limit of normal at six to eight hours or 18 to
24 hours, was not met (varespladib patients 57% versus
placebo patients 51%, p = 0.55). However, the results
showed statistically significant reductions of
sPLA2
as early as 18 hours post-PCI procedure, which persisted
throughout the five days of dosing (-93.0%, p < 0.001).
Consistent with results from other clinical studies with
varespladib, there were numerical reductions in CRP from
baseline versus placebo at three to five days (-82.1%, p = 0.23).
Previous
Experience at Eli Lilly and Shionogi & Co.,
Ltd.
Eli Lilly and Shionogi & Co., Ltd. previously
conducted a series of clinical studies evaluating varespladib
and A-001 in
various inflammatory conditions. In total, at least 17 Phase 1
and Phase 2 clinical studies evaluated varespladib and
A-001 as a
treatment in sepsis, rheumatoid arthritis, asthma and ulcerative
colitis, an inflammatory bowel disease. Results from these
studies provide a large body of safety data for varespladib and
A-001 with
more than 1,000 healthy volunteers and subjects receiving
treatment.
Throughout these studies, varespladib was generally
well-tolerated.
Non-Clinical
Studies with Varespladib and
A-001
Approximately 150 preclinical pharmacology and toxicology
studies have been completed with varespladib and
A-001,
including two-year rat and mouse carcinogenicity studies,
one-year primate study and three-month rat study in combination
with Lipitor (atorvastatin).
A-623
A-623 is a
peptibody antagonist of the BAFF cytokine that is initially
being developed as a treatment for lupus. BLyS, also known as
B-cell activating factor, or BAFF, is a tumor necrosis family
member and is critical to the development, maintenance and
survival of B-cells. It is primarily expressed by macrophages,
monocytes and dendritic cells and interacts with three different
receptors on B-cells including BAFF receptor, or BAFF-R, B-cell
maturation, or BCMA, and transmembrane activator and cyclophilin
ligand interactor, or TACI. The BAFF-R receptor is expressed
primarily on peripheral B-cells.
Two randomized, dose-ranging, placebo-controlled Phase 1
clinical studies
A-623 in 104
lupus patients have already been completed. Results from these
studies demonstrated
A-623
generated anti-BAFF activity and showed statistically
significant reductions in B-cells of
50-70% (p
< 0.001) in lupus patients across multiple subcutaneous and
intravenous formulations.
After successfully reactivating our Investigational New Drug
Application, or IND, we initiated a Phase 2b clinical study with
A623 for the treatment of lupus in July 2010 called PEARL-SC. We
may also study
A-623 in
other B-cell mediated autoimmune diseases such as
Sjögrens Syndrome or orphan indications such as
myasthenia gravis and pemphigus. We are actively pursuing a
partnership with major pharmaceutical companies to develop and
commercialize
A-623.
81
We intend to advance the development of our BAFF targeting
molecule,
A-623, a
selective peptibody, to exploit its broad clinical utility in
autoimmune diseases.
A-623, as a
peptibody directed against BAFF, was developed as an alternative
to antibodies and is produced in escherichia coli versus
antibodies that are produced in mammalian cells. In addition,
A-623 offers
a number of potential differentiations over other anti-BAFF
compounds, as well as other novel B-cell directed therapies,
including:
|
|
|
|
|
convenient, at-home, patient-administered subcutaneous dosing
with a range of dosing frequencies including monthly and weekly;
|
|
|
|
|
|
ability to bind to both membrane-bound and soluble BAFF, which
may confer differentiating pharmacodynamic characteristics;
|
|
|
|
|
|
non-glycosylated protein that is produced in a bacterial
fermentation manufacturing process, which may reduce the
potential to be immunogenic, and may provide manufacturing
benefits and lower cost of goods; and
|
|
|
|
|
|
multiple binding domains achieve highest reported affinity for
inhibition of BAFF.
|
Market
Opportunity
Lupus is an autoimmune disorder that involves inflammation that
causes swelling, pain and tissue damage throughout the body.
Lupus can affect any part of the body, but especially the skin,
heart, brain, lungs, joints and the kidneys. The course of the
disease is unpredictable, with periods of illness, called flares
alternating with remission. The Lupus Foundation estimates that
approximately 1.5 million people in the United States and
five million worldwide suffer from lupus. Although lupus may
affect people of either sex, women are 10 times more likely to
suffer from the disease than men, according to the Lupus
Foundation.
Patients with active lupus may have a broad range of symptoms
related to the inflammation. Inflammation of the brain may cause
seizures and other neurologic abnormalities. Inflammation of the
heart may cause heart failure or sudden death. Lung inflammation
causes shortness of breath. Lupus may also cause swollen joints
and severe rash. In addition, LN may lead to kidney dialysis or
transplantation.
Although the cause of lupus is still not completely understood,
B-cell activation and autoantibody production are known to be
central to the process. Evidence has emerged that
over-expression of BAFF plays an important role in this disease
process. In preclinical studies, transgenic mice created to
over-express BAFF begin to exhibit symptoms similar to lupus. In
addition, treatment of these same mice with BAFF antagonists
appears to ameliorate the disease.
PEARL-SC
Phase 2b Clinical Study in Patients with Lupus
Based on positive results among 104 patients in our Phase
1a and 1b clinical studies, we initiated a Phase 2b clinical
study in lupus patients called PEARL-SC. PEARL-SC is a
randomized, placebo-controlled, phase 2b clinical study which
may enroll up to 600 patients in approximately
60 centers worldwide. Subjects will be randomized into
three active subcutaneous treatment arms and one placebo
treatment arm for a minimum of 24 weeks. The primary
endpoint of the PEARL-SC study will be clinical improvement at
24 weeks in responder rates of a composite systemic lupus
erythematosus responder index, or SRI, in the pooled treatment
arms versus placebo. The primary SRI endpoint is a composite
score based upon changes in SELENA and SLEDAI disease activity
scale, Physicians Global Assessment scores and British
Isles Lupus Assessment Group scores, which are clinical
standards for the measurement of disease severity in lupus
patients. Secondary endpoints will include safety, improvement
in other clinical assessment scores, clinical response in
patients with various baseline disease severities, resolution of
fatigue, steroid utilization and time to flare. An interim
biomarker analysis to establish the appropriate drug effect on
total B-Cells is included early in the study. Initially we
intend to randomize
82
only 480 patients. This total number of patients will allow
us to detect a treatment effect of 14% with a p-value of 0.05
between the pooled active arms and the placebo arm.
The following table represents our current PEARL-SC study as
well as a the option to further extend the study for collection
of long-term safety collection.
Future
Development of
A-623
A-623
Manufacturing Strategy
In May 2010, we successfully completed a manufacturing campaign
for a high concentration
A-623
injection formulation for subcutaneous administration.
Manufacturing was conducted per current good manufacturing
practices, or cGMP, and the product was released to clinical
sites in July 2010. Our active and placebo inventory is
sufficient to complete dosing of 120 patients for up to
16 weeks of therapy. In August 2010, we manufactured a
second batch of vials of the high-concentration
A-623
injection formulation from 34 liters of Amgen manufactured bulk
drug substance and, upon appropriate quality inspection and
testing, plan to release this batch for clinical use by November
2010. Upon successful release of this batch, we believe we will
have sufficient clinical material, both placebo and
A-623, to
dose up to 480 patients for a minimum of six months in the
PEARL-SC study.
We are currently evaluating proposals from a number of contract
manufacturers, or CMOs, with large-scale GMP manufacturing
capabilities for the production of
A-623. In
the fourth quarter of 2010, we expect to announce the selection
of a CMO and to begin the technical transfer of the full
manufacturing process to a large scale contract manufacturer. As
a result, in early 2011, we plan to initiate manufacturing of
GMP bulk product for eventual pivotal clinical studies and an
optional expansion
and/or
extension of the PEARL-SC study.
83
The following chart outlines the basic manufacturing steps
required for the production of
A-623.
A-623
Regulatory Strategy
In July 2010, we received clearance from the FDA to begin
recruitment of lupus patients into the PEARL-SC clinical study.
The approved protocol allows for enrollment of up to
600 patients treated for a maximum of 12 months.
Patients enrolled in this study will be randomized into three
active treatment arms and one placebo arm. Subsequent to this
approval the FDA requested additional information regarding
characterization and qualification of the manufactured vials of
A-623. In
addition the FDA requested minor changes to aspects of PEARL-SC
study including collection of ECG testing at the end of dosing
and a recommendation for a corticosteroid tapering strategy.
Neither of these changes are considered material to the conduct
of the PEARL-SC study. The FDA also recommended we submit to the
IND analytical and comparability data from our recently
completed manufacturing lot of
A-623 vials
and a comparability proposal for purposes of soliciting their
input prior to implementation. We submitted a response to the
FDA in October 2010.
It is our intent to continuously submit results of comparability
testing from all of our manufacturing campaigns to the FDA. We
also intend to discuss with the FDA a comparability proposal
that would ensure future batches of material manufactured by our
eventual CMO would meet the FDAs standards for
equivalence. If these batches meet specification and the FDA
agrees A-623
product manufactured by our CMO meets comparability
requirements, we believe these batches could be used to further
extend and expand the PEARL-SC study
and/or
initiate identical Phase 3 clinical studies for purposes of
registration.
Historical
Clinical Studies
Prior to our in-licensing of
A-623, Amgen
completed two Phase 1 clinical studies of
A-623 in
lupus patients to evaluate the safety and pharmacokinetics of
single and multiple doses of drug using intravenous and
subcutaneous formulations. Prior to conducting Phase 1 clinical
studies in lupus patients, Amgen conducted a pre-Phase 1
clinical study in lupus patients. In Amgens pre-Phase 1
clinical study, individual B-cell subsets, such as mature
naïve B-cells, activated B-cells and memory B-cells, all
therapeutic targets for
A-623, were
quantified in order to characterize the specific B-cell subset
abnormalities associated with lupus.
The randomized, placebo-controlled, dose-escalation Phase 1a
clinical study evaluated
A-623 as a
single intravenous or subcutaneous therapy among 56 lupus
patients. Intravenous doses included 1, 3 and 6 mg/kg, and
subcutaneous doses included 0.1, 0.3, 1 and 3 mg/kg. The
primary endpoint was to assess the safety and tolerability of
single dose administrations of
A-623.
Secondary endpoints were designed to assess the plasma
pharmacokinetic profile and immunogenicity of
A-623.
Results from this clinical study indicated the safety and
tolerability of
A-623
administered as single dose of intravenous or subcutaneous was
comparable to placebo. Single doses of
A-623
exhibited linear pharmacokinetics after both intravenous and
subcutaneous administration. There were comparable adverse
events between the
A-623 and
placebo groups with no deaths reported. In addition, no
neutralization antibodies were seen across all doses. The most
common adverse events were nausea (15%), headache (10%), upper
respiratory tract infection (10%) and diarrhea (8%).
A-623 was
evaluated in a randomized, placebo-controlled, multi-dose Phase
1b clinical study as an intravenous or subcutaneous therapy
among 63 lupus patients. The intravenous dose was 6 mg/kg,
and
84
subcutaneous doses included 0.3, 1 and 3 mg/kg. Patients
received their doses of
A-623 or
placebo once-weekly for four weeks. The primary endpoint was to
assess the safety and tolerability of multiple dose
administrations of
A-623.
Secondary endpoints were designed to assess the plasma
pharmacokinetic profile and immunogenicity of
A-623 after
multiple doses. Results showed that multiple doses of
A-623
exhibited dose-proportional pharmacokinetics after both
intravenous and subcutaneous administration. Further, results
demonstrated a dose-dependent decrease in total B-cells as early
as 15 days of treatment, and total B-cell reduction (up to
approximately
60-70% of
baseline) reached its nadir after about 160 days of
therapy. By six months after treatment, the B-cell populations
had returned to baseline levels.
Figure 7: Total B-cell Depletion
An experimental analysis was also conducted to assess B-cell
subsets in patients following multiple doses. Results
demonstrated that
A-623
selectively modulate certain B-cell subsets and induced trends
toward normalizing the B-cell abnormalities that were observed
in lupus patients in the pre-Phase 1 clinical study.
Results indicated that the tolerability of
A-623
administered as multiple doses of intravenous or subcutaneous
administration was generally comparable to placebo. There were
no deaths reported between the
A-623 and
placebo. Few neutralization antibodies were seen, and all
resolved in subsequent visits. Based on these results and
published data from competitor studies, we initiated a Phase 2b
clinical study evaluating
A-623 in
lupus patients during the second half of 2010.
85
A-001
A-001 is an
intravenously administered, potent, broad-spectrum inhibitor of
sPLA2,
including forms IIa, V and X.
A-001 is
currently being evaluated in a Phase 2 clinical study for the
prevention of acute chest syndrome associated with sickle cell
disease in at-risk patients. Substantial scientific evidence
implicates
sPLA2
activity in the development of acute chest syndrome associated
with sickle cell disease, as well as other forms of acute lung
injury. The FDA granted orphan drug and fast-track designation
for A-001
for the prevention of acute chest syndrome in at-risk patients.
We currently retain all worldwide product rights, except in
Japan where Shionogi & Co., Ltd. retains rights. We
also licensed
A-001 from
Eli Lilly and Shionogi & Co., Ltd. in July 2006.
sPLA2
levels increase in advance of acute chest syndrome episodes and
can be used alongside the presence of fever to strongly predict
an impending episode. There is a strong correlation between
levels of CRP and
sPLA2
in this patient population. Patients with acute chest syndrome
associated with sickle cell disease can exhibit levels of
sPLA2
that can be 100 times greater than normal. We believe that early
intervention with
A-001 to
inhibit
sPLA2
activity may offer a novel preventative therapy to improve
outcome among sickle cell disease patients presenting with a
high risk of acute chest syndrome.
Market
Opportunity
Sickle cell disease is a lifelong genetic, blood disorder
typically diagnosed during early childhood. According to the
Sickle Cell Information Center, in the United States, over
70,000 people currently suffer from the disease and
approximately 1,000 children are born with the disease annually.
According to Medtech Insight, in Europe, there are over
200,000 people suffering from the disease, and the numbers
increase dramatically in Africa, where, according to the WHO,
200,000 children alone are born with sickle cell disease each
year. Life expectancy for these patients is significantly
shortened, with most expected to live only until their mid-40s.
The disease is characterized by structurally altered red blood
cells that assume an abnormal shape, similar to a sickle, and
produce an altered form of hemoglobin. These altered red blood
cells have a shortened life-cycle, become stiff and have
difficulty passing through the bodys small blood vessels.
At times, these abnormal cells may obstruct or block blood flow
through small blood vessels, leading to significant damage in
tissue and bone. This damage is more commonly labeled as VOC.
During VOC, blockage occurs within the circulation of the long
bones, causing microscopic bone damage. Fragments of bone or
bone fat may break free and embolize to the lungs, causing lung
injury.
VOC is a common trigger for the more serious complication of
acute chest syndrome associated with sickle cell disease. Acute
chest syndrome exhibits symptoms and characteristics similar to
acute lung injury. There are an estimated 10,000 episodes of
acute chest syndrome associated with sickle cell disease per
year in the United States. It represents the most common cause
of death in sickle cell patients and the second most common
cause of hospitalization among such patients. A majority of
sickle cell patients will experience at least one episode of
acute chest syndrome and repeated episodes can result in
progressive lung disease. The disorder is most common in the
two- to four- year age group and gradually declines in incidence
with age.
There are no marketed therapies targeting acute chest syndrome
associated with sickle cell disease. The most common treatment
regimen includes heavy doses of corticosteroids, opiates,
transfusion and antibiotics while the patient suffers through
the attack. In addition, hydroxyurea, a chemotherapy, was found
to reduce the frequency of VOC and the need for blood
transfusions in adult patients with sickle
86
cell disease. However, all of these therapeutics are associated
with significant adverse effects while only offering limited
patient benefit.
Our planned multinational, randomized, double-blind,
placebo-controlled Phase 3 clinical study will enroll up to
200 patients with sickle cell disease who are at an
elevated risk of developing acute chest syndrome as a result of
fever, vaso-occlusive crisis, and CRP 5.0 mg/l
³
at the time of hospitalization. Patients will be randomized to
receive a continuous infusion of
A-001 or
placebo for 48 hours after randomization. The primary
endpoint of this study will be freedom from acute chest syndrome
as determined by physician assessment and independent review of
chest X-rays. This study represents a unique treatment approach
for a small, orphan designated indication. As a result the
appropriateness of the design and endpoints of this study for
purposes of registration will only be known at the conclusion of
the study and upon submission to the FDA.
Historical
Clinical Studies
Phase 2 Acute Chest Syndrome in Hospitalized Patients with
Sickle Cell Disease Study Investigation of the
Modulation of Phospholipase in Acute Chest Syndrome, or
IMPACTS.
In January 2007, we initiated a randomized, double-blind,
placebo-controlled Phase 2 clinical study to assess the safety
and tolerability of escalating doses of
A-001
therapy when administered as a
48-hour
continuous infusion. The clinical study was designed to enroll
up to 75 patients across approximately 30 sites in the
United States. This clinical study enrolls hospitalized sickle
cell disease patients, at risk for acute chest syndrome on the
basis of VOC, fever and serum
sPLA2
concentration level greater than 50 mg/mL. The primary
endpoint for the clinical study was designed to assess safety
and tolerability. Secondary endpoints included the absence of
acute chest syndrome, suppression of
sPLA2,
reduced need for blood transfusions and assessment of
pharmacokinetics.
The first group of patients was randomized 2:1 to receive low
dose A-001
or placebo as a
48-hour
continuous infusion. A pre-specified interim analysis was
conducted in February 2009 after the 30th patient completed
treatment to examine safety and adjust dosing schedules. The
interim data was balanced between two dosing arms of 30
mug/kg/hr (n = 11) and 55 mug/kg/hr (n = 6). Interim
results indicated serum levels of
A-001 when
dosed at 55 mug/kg/hr reduced
sPLA2
activity levels by more than
87
80% from baseline within 48 hours. Furthermore, the
prevention of acute chest syndrome associated with sickle cell
disease appeared to be related to the level of
sPLA2
activity. The DSMB recommended the clinical study continue based
on safety and tolerability. In addition, given the safety
profile, the DSMB approved the addition of a higher dose group
of 110 mug/kg/hr via continuous infusion during the second half
of the clinical study. We believe that the data suggest
A-001 can
suppress
sPLA2
at levels that may prevent the complication of acute chest
syndrome associated with sickle cell disease.
Table 8: Reductions of
sPLA2
activity from baseline and incidence of acute chest syndrome
(including placebo patients and patients receiving
A-001).
Exploratory analysis to determine correlation between degree of
sPLA2
suppression and incidence of acute chest syndrome.
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
48-Hour
sPLA2Activity
as
|
|
|
|
|
|
|
|
|
|
|
|
|
a Percentage of Baseline
|
|
|
0.0% < 25.0%
|
|
|
³25%
< 50%
|
|
|
³50%
< 75%
|
|
|
³
75%
|
Number of Subjects
|
|
|
|
7
|
|
|
|
|
7
|
|
|
|
|
3
|
|
|
|
12
|
Number of Subjects Developing Acute Chest Syndrome(%)
|
|
|
|
0(0
|
)
|
|
|
|
2(28
|
)
|
|
|
|
1(33
|
)
|
|
|
4(25)
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Our
Strategy
Our objective is to develop and commercialize our product
candidates to treat serious diseases associated with
inflammation, including cardiovascular and autoimmune diseases.
To achieve these objectives, we intend to initially focus on:
Advancing
Varespladib Through Phase 3.
Inflammatory processes and lipid abnormalities are central to
the onset of acute coronary syndrome and the development of CAD.
varespladib operates through a novel mechanism of action to
offer both targeted anti-inflammatory activity and incremental
lipid reductions, including LDL-C, when used in combination with
statins. Despite the benefits of statin therapy, many acute
coronary syndrome patients still remain at substantial risk of a
coronary event, suggesting additional biological mechanisms may
be relevant, including inflammation. We believe that combination
therapy with varespladib and statins will provide acute coronary
syndrome patients with a unique, short-term therapeutic option
unavailable with existing agents today. In addition, we believe
that an opportunity exists in the future to evaluate varespladib
in chronic indications such as CAD.
Advancing
Clinical Development of
A-623.
We are advancing the development of
A-623 to
exploit the broad potential clinical utility of BAFF antagonism.
We have initiated the Phase 2b clinical study known as PEARL-SC
in lupus patients. We may opportunistically enter into
collaborations with third parties for development of this
compound in lupus or in other B-cell mediated diseases, such as
multiple sclerosis, rheumatoid arthritis or Sjögrens
Syndrome, that may benefit from BAFF antagonism, including
securing corporate partners whose capabilities complement ours.
We are also actively pursuing a partnership with major
pharmaceutical companies to develop and commercialize
A-623. We
believe that a partnership could enable us to obtain funding for
development of
A-623 and to
accelerate its clinical, manufacturing and commercial
development with collaborators whose capabilities complement
ours.
We are seeking to structure a partnership that allows us to
retain significant control over the development and
commercialization of
A-623 in the
United States, and to retain economic interests in regions
outside of the United States. Given the recent positive results
of a BAFF-specific antagonist in multiple large, late-stage
clinical studies, we believe that
A-623 could
be an attractive product candidate for pharmaceutical companies
interested in exploiting opportunities in autoimmune diseases
directed at lupus, as well as to other B-cell related autoimmune
diseases.
88
In the future, if additional funds are available, we may develop
A-001, an
intravenous
sPLA2
inhibitor for prevention of acute chest syndrome associated with
sickle cell disease, because we identified that elevations in
sPLA2
activity are known to precede and predict disease progression.
Given that there are currently no approved drugs for the
prevention of acute chest syndrome associated with sickle cell
disease, we have received orphan drug designation and fast track
status from the FDA for
A-001.
Leveraging
Our
sPLA2
Expertise to Develop Products for Additional Disease
Indications.
We believe that we have developed a leadership position in the
field of
sPLA2
inhibition. Beyond our acute coronary syndrome and acute chest
syndrome program, we believe that
sPLA2
inhibition may have applications in other acute disease settings
where early intervention may have an impact and reduce
anti-inflammatory activity, such as acute lung injury.
Additionally, we believe that we can apply our
sPLA2
expertise to develop novel therapeutics for a number of chronic
diseases. For example,
sPLA2
has been shown to be involved in the development of such chronic
inflammatory diseases as atherosclerosis and dermatitis. We plan
to pursue these indications opportunistically and potentially in
collaboration with third parties.
We are also developing new and unique
sPLA2
inhibitor compounds for additional therapeutic areas.
A-003 is our
second generation lead candidate. We plan to continue
preclinical development of
A-003 for an
IND filing and we will continue to assess additional new
compounds.
Developing
Commercial Strategies Designed to Maximize Our Product
Candidates Market Potential.
Our primary product candidates are focused on either the acute
care setting in the hospital or highly-specialized physician
segments, such as rheumatologists. We believe that we can build
a small, focused sales force capable of marketing our products
effectively in acute care and orphan indications such as acute
coronary syndrome and acute chest syndrome associated with
sickle cell disease. In other chronic indications such as CAD,
we intend to seek commercial collaborations with companies that
have a large, dedicated sales force focused on general
practitioners and cardiologists and we plan to seek
commercialization partners for products in non-specialty and
international markets.
Competition
Our industry is highly competitive and subject to rapid and
significant technological change. Our potential competitors
include large pharmaceutical and biotechnology companies,
specialty pharmaceutical and generic drug companies, academic
institutions, government agencies and research institutions. We
believe that key competitive factors that will affect the
development and commercial success of our product candidates are
efficacy, safety and tolerability profile, reliability,
convenience of dosing, price and reimbursement.
Many of our potential competitors, including many of the
organizations named below, have substantially greater financial,
technical and human resources than we do and significantly
greater experience in the discovery and development of product
candidates, obtaining FDA and other regulatory approvals of
products and the commercialization of those products.
Accordingly, our competitors may be more successful than we may
be in obtaining FDA approval for drugs and achieving widespread
market acceptance. Our competitors drugs may be more
effective, or more effectively marketed and sold, than any drug
we may commercialize and may render our product candidates
obsolete or non-competitive before we can recover the expenses
of developing and commercializing any of our product candidates.
We anticipate that we will face intense and increasing
competition as new drugs enter the market and advanced
technologies become available. Finally, the development of new
treatment methods for the diseases we are targeting could render
our drugs non-competitive or obsolete.
89
The
sPLA2
product candidates we are currently developing, if approved,
will face intense competition, either as monotherapies or in
combination therapies. Although there are no
sPLA2
inhibitors currently approved by the FDA, we are aware of other
pharmaceutical companies, as described below, that are
developing product candidates in this area for separate
indications.
sPLA2
in Acute Coronary Syndrome
Our lead product candidate, varespladib, for the short-term
(16-week) treatment of acute coronary syndrome has a dual
mechanism of action that we believe confers anti-inflammatory
and lipid-lowering and lipid-modulating benefits. The market for
cardiovascular therapeutics and acute coronary syndrome,
specifically, is especially large and competitive. A wide range
of medications are typically administered to patients suffering
an acute coronary syndrome event in order to reduce ischemia and
thrombosis and improve blood flow. We expect that varespladib
for the treatment of acute coronary syndrome patients, if
approved, may compete with the following anti-inflammatory
therapeutics in development.
|
|
|
|
|
|
|
|
|
|
|
|
|
Compound
|
|
|
Stage
|
|
|
Company
|
|
|
Indications
|
|
|
Notes
|
Darapladib
|
|
|
Phase 3
|
|
|
GlaxoSmithKline plc
|
|
|
Acute coronary
syndrome
|
|
|
Lp-PLA2
Inhibitor
Collaboration with Human
Genome Sciences, Inc.
Various back-up compounds
|
|
|
|
|
|
|
|
|
|
|
|
|
|
VIA-2291
|
|
|
Phase 2
|
|
|
Via Pharmaceuticals, Inc.
|
|
|
Acute coronary
syndrome or
atherosclerosis
|
|
|
5-lipoxygenase inhibitor
Discussions on-going with
FDA
|
|
|
|
|
|
|
|
|
|
|
|
|
|
E-5555
|
|
|
Phase 2
|
|
|
Eisai Inc.
|
|
|
Acute coronary
syndrome or
atherosclerosis
|
|
|
600 patient study completed
October 2009
Thrombin receptor
antagonist
Evaluating biomarkers and
events
|
|
|
|
|
|
|
|
|
|
|
|
|
|
Other
Agents Under Development
Additionally, we are aware of other products in development that
are being tested for anti-inflammatory benefits in patients with
acute coronary syndrome such as Via Pharmaceuticals, Inc. and
its
5-lipoxygenase,
or 5-LO, inhibitor, which has been evaluated in Phase 2 clinical
studies, GlaxoSmithKline plc and its product candidate,
darapladib, which is an
Lp-PLA2
inhibitor currently being evaluated in Phase 3 clinical studies.
If approved, these products or others in development may compete
directly with varespladib.
Approved
Categories of Drugs
Statins Treatment with varespladib is
designed to offer anti-inflammatory benefits for acute coronary
syndrome patients that are additive to treatment with statins.
However, statin therapy is thought to confer some element of
anti-inflammatory benefit as monotherapy. In certain
circumstances, it is possible the anti-inflammatory benefits of
statin monotherapy with products such as Lipitor (atorvastatin),
which is marketed by Pfizer Inc., Crestor (rosuvastatin), which
is marketed by AstraZeneca UK Limited and Zocor (simvastatin),
which is marketed by Merck & Co., Inc. may be viewed
as competitive to that offered by varespladib.
90
Other Lipid-Lowering Therapies
Increasingly, additional lipid-lowering agents are being
administered either in combination with statins or as
monotherapy to help acute coronary syndrome patients reduce
levels of LDL-C. varespladib has demonstrated LDL-C lowering
benefits when tested as monotherapy and in combination with
statin therapy. To the extent acute coronary syndrome patients
need additional LDL-C lowering, varespladib may compete for use
with other approved agents such as Vytorin, which is a fixed
dose combination therapy combining ezetimibe and Zocor, Tricor
(fenofibrate tablets) and Niaspan (niacin), both of which are
marketed by Abbott Laboratories, Zetia (ezetimibe) and fish oils
(omega-3).
Lupus
No new therapies have been approved for lupus in the last
50 years. Current therapies such as non-steroidal
anti-inflammatory drugs, or NSAIDs, corticosteroids and
immunosuppressants generally act to hold back broadly the
proliferation of many types of cells, including white blood
cells. However, use of these agents is associated with
significant adverse events and broad immune suppression.
Recently, several new biological agents under development have
targeted BAFF (or BLyS) and other B-cell related pathways for
the treatment of lupus. These product candidates include
Benlysta (belimumab) from Human Genome Sciences, Inc., LY2127399
from Eli Lilly and Company, atacicept, or TACI-Ig, from
ZymoGenetics Inc. and what we believe to be more non-specific
B-cell depleting agents such as Rituxan from Genentech, Inc. and
epratuzumab from Immunomedics, Inc. We believe that
A-623 may
offer potential differentiation from these agents, including:
demonstrated dosing flexibility with both subcutaneous and
intravenous delivery; selective modulation and reduction of
relevant B-cell types in lupus patients; the ability to bind to
both membrane-bound and soluble BAFF; its smaller size as
compared to a full antibody, which may confer differentiating
pharmacokinetic and pharmacodynamic characteristics; and
distinct patent protection based on a novel and proprietary
technology developed and commercialized by Amgen, which may also
confer potential manufacturing advantages with lower cost of
goods based on a bacterial fermentation manufacturing process.
91
|
|
|
|
|
|
|
|
|
|
|
|
|
Compound
|
|
|
Stage
|
|
|
Company
|
|
|
Indications
|
|
|
Notes
|
Benlysta (intravenous and subcutaneous)
|
|
|
BLA submission
|
|
|
Human Genome Sciences, Inc./GlaxoSmithKline plc
|
|
|
Lupus
|
|
|
Monoclonal antibody against BAFF, an agent that demonstrated partial reduction in B-cells
Inhibits soluble BAFF only
Positive results reported in two Phase 3 clinical studies
|
Atacicept (intravenous)
|
|
|
Phase 3
|
|
|
ZymoGenetics Inc./Merck Serono S.A.
|
|
|
Lupus, LN
|
|
|
Fusion protein against BAFF and APRIL; Phase 3 clinical study in LN stopped due to safety issues
Phase 3 clinical study in lupus on-going
|
Epratuzumab (intravenous)
|
|
|
Phase 2b
|
|
|
Immunomedics, Inc./UCB S.A.
|
|
|
Lupus, Non-Hodgkins Lymphoma
|
|
|
Humanized antibody against CD-22, an agent that specifically targets B-cells and leads to partial depletion of peripheral B-cells
Positive Phase 2b clinical study results reported
|
LY2127399 (subcutaneous)
|
|
|
Phase 2
|
|
|
Eli Lilly and Company
|
|
|
Rheumatoid Arthritis Multiple Myelomas
|
|
|
Monoclonal antibody against BAFF inhibits soluble and membrane-bound BAFF
Recent positive results in RA study
|
Ocrelizumab (intravenous)
|
|
|
Phase 3
|
|
|
F. Hoffman - La Roche Ltd./Biogen Idec Inc.
|
|
|
LN
|
|
|
Monoclonal antibody against CD-20 that leads to rapid and profound depletion of circulating B-cells
Phase 3 clinical study in lupus halted
|
Lupuzor (subcutaneous)
|
|
|
Phase 2b
|
|
|
Cephalon, Inc./ImmuPharma PLC
|
|
|
Lupus
|
|
|
Modulates CD4 T cells
Positive Phase 2b clinical study results reported
|
|
|
|
|
|
|
|
|
|
|
|
|
|
sPLA2
for Acute Chest Syndrome Associated with Sickle Cell
Disease
There are no currently approved agents for treatment or
prophylaxis of acute chest syndrome associated with sickle cell
disease. Droxia (hydroxyurea) is approved for prevention of VOC
in sickle cell disease and thus could reduce the pool of
patients with VOC at risk for acute chest syndrome. In addition,
there is evidence in the literature that blood transfusions may
prevent the occurrence of acute chest syndrome associated with
sickle cell disease, and a randomized clinical study is underway
by the National Heart, Lung and Blood Institute to explore this
possibility.
Intellectual
Property
Our policy is to pursue, maintain and defend patent rights,
developed internally and licensed from third parties, to protect
the technology, inventions and improvements that are
commercially important to the development of our business. We
also rely on trade secrets that may be important to the
development of our business.
Our success will depend significantly on our ability to:
|
|
|
|
|
obtain and maintain patent and other proprietary protection for
the technology, inventions and improvements we consider
important to our business;
|
|
|
|
defend our patents;
|
|
|
|
preserve the confidentiality of our trade secrets; and
|
92
|
|
|
|
|
operate our business without infringing the patents and
proprietary rights of third parties.
|
Varespladib
and
A-001
As of the date of this prospectus, our licensed varespladib and
A-001 patent
portfolio includes:
|
|
|
|
|
13 U.S. patents;
|
|
|
|
One pending U.S. non-provisional patent application;
|
|
|
|
Five European, or EP, patents, each validated in one or more of
Austria, Belgium, Denmark, France, Germany, Greece, Ireland,
Italy, Liechtenstein, Luxembourg, the Netherlands, Portugal,
Spain, Sweden, Switzerland and the United Kingdom;
|
|
|
|
One pending EP patent application;
|
|
|
|
18 non-EP foreign patents in Argentina, Australia, Brazil,
Canada, China, Finland, India, Malaysia, Mexico, the
Philippines, South Korea, Taiwan and Turkey; and
|
|
|
|
Four pending non-EP foreign patent applications in Brazil,
Canada, China and Thailand.
|
We hold exclusive worldwide licenses from Eli Lilly and
Shionogi & Co., Ltd. to all of these patents and
patent applications with the exception of licensing rights in
Japan, which Shionogi & Co., Ltd. retains. These
licenses are described below under
Licenses. The patents and applications
described above contain claims directed to varespladib and
A-001
compositions of matter and to various methods of making and
using varespladib and
A-001,
including methods of treating various inflammatory conditions.
The issued U.S. patents are currently scheduled to expire
between 2014 and 2021.
As of the date of this prospectus, our internally developed
varespladib and
A-001 patent
portfolio includes:
|
|
|
|
|
Three pending U.S. non-provisional patent applications;
|
|
|
|
Three pending U.S. provisional patent applications;
|
|
|
|
Two pending Patent Cooperation Treaty, or PCT, patent
applications; and
|
|
|
|
National phase applications in the European Patent Office, the
Eurasian Patent Organization and 17 other countries (Australia,
Brazil, Canada, China, Hong Kong, India, Indonesia, Israel,
Japan, Malaysia, Mexico, New Zealand, The Philippines,
Singapore, South Africa, South Korea and Vietnam).
|
We own, and therefore hold all worldwide rights in and to, these
patent applications, which contain claims directed to
varespladib and
A-001
compositions of matter and methods of treating various
cardiovascular indications.
A-003
As of the date of this prospectus, our licensed
A-003 patent
portfolio includes:
|
|
|
|
|
Two licensed U.S. patents;
|
93
|
|
|
|
|
One licensed pending U.S. non-provisional patent
application (also listed above as covering varespladib and
A-001);
|
|
|
|
Five licensed EP patents (two also listed above as covering
varespladib and
A-001), each
validated in one or more of Albania, Austria, Belgium, Denmark,
Finland, France, Germany, Greece, Ireland, Italy, Latvia,
Liechtenstein, Lithuania, Luxembourg, the Netherlands, Portugal,
Romania, Slovenia, Spain, Sweden, Switzerland and the United
Kingdom;
|
|
|
|
One licensed pending EP patent application (also listed above as
covering varespladib and
A-001);
|
|
|
|
Thirteen licensed non-EP foreign patents (four also listed above
as covering varespladib and
A-001) in
Argentina, Australia, Canada, China, India, Mexico, South Korea
and Taiwan; and
|
|
|
|
Three licensed pending non-EP foreign patent applications (all
also listed above as covering varespladib and
A-001) in
Brazil, Canada and China.
|
We hold exclusive worldwide licenses from Eli Lilly and
Shionogi & Co., Ltd. to these patents and patent
applications with the exception of licensing rights in Japan,
which Shionogi & Co., Ltd. retains. These licenses are
described below under Licenses. The
patents and applications listed above contain claims directed to
A-003
compositions of matter and to various methods of making and
using A-003,
including methods of treating various inflammatory indications.
The issued U.S. patents are currently scheduled to expire
between 2017 and 2018.
As of the date of this prospectus, our internally developed
A-003 patent
portfolio includes:
|
|
|
|
|
Three U.S. non-provisional patent applications (all also
listed above as covering varespladib and
A-001);
|
|
|
|
Three pending U.S. provisional patent applications (both
also listed above as covering varespladib and
A-001);
|
|
|
|
Two pending PCT patent applications (both also listed above as
covering varespladib and
A-001); and
|
|
|
|
National phase applications in the European Patent Office, the
Eurasian Patent Organization and 17 other countries (Australia,
Brazil, Canada, China, Hong Kong, India, Indonesia, Israel,
Japan, Malaysia, Mexico, New Zealand, The Philippines,
Singapore, South Africa, South Korea and Vietnam).
|
We own, and therefore hold all worldwide rights in and to, these
patent applications, which contain claims directed to
A-003
compositions of matter and methods of treating various
cardiovascular indications.
New
sPLA2
Compounds
As of the date of this prospectus, our new
sPLA2
compound patent portfolio includes over 30 licensed
U.S. patents, three EP patents, and one pending EP
application not listed above as covering
A-001,
varespladib or
A-003. We
hold exclusive worldwide licenses from Eli Lilly and
Shionogi & Co., Ltd. to these patents and patent
applications with the exception of licensing rights in Japan,
which Shionogi & Co., Ltd. retains. These licenses are
described below under Licenses. The
patents and applications
94
listed above contain claims directed to various
sPLA2
second generation compounds, as well as methods of making and
using these new
sPLA2
compounds. The issued U.S. patents are currently scheduled
to expire between 2013 and 2024.
A-623
As of the date of this prospectus, our
A-623 patent
portfolio includes:
|
|
|
|
|
Two U.S. patents;
|
|
|
|
One pending U.S. non-provisional patent application;
|
|
|
|
One EP patent validated in Albania, Austria, Belgium, Cyprus,
Denmark, Finland, France, Germany, Greece, Ireland, Italy,
Latvia, Liechtenstein, Lithuania, Luxembourg, Monaco, the
Netherlands, Portugal, Romania, Slovenia, Spain, Sweden,
Switzerland, Turkey and the United Kingdom;
|
|
|
|
|
|
Two pending EP patent applications;
|
|
|
|
|
|
Ten non-EP foreign patents in Australia, China, Estonia, Eurasia
(validated in all nine Eurasian countries), Japan, New Zealand,
Singapore, South Korea and South Africa; and
|
|
|
|
14 pending non-EP foreign patent applications in Brazil,
Bulgaria, Canada, China, the Czech Republic, Hong Kong, Hungary,
Israel, Mexico, Norway, the Philippines, Poland,
Serbia/Yugoslavia and Slovakia.
|
We hold exclusive worldwide licenses from Amgen to all of these
patents and patent applications. In addition, we hold a
non-exclusive worldwide license to one pending US application,
one EP patent validated in 15 countries, one pending EP patent
application, ten non-EP foreign patents and 31 non-EP foreign
patent applications relating to peptibody compositions and
formulations.
The U.S. patent system permits the filing of provisional
and non-provisional patent applications. A non-provisional
patent application is examined by the U.S. Patent Office,
or USPTO, and can mature into a patent once the USPTO determines
that the claimed invention meets the standards for
patentability. A provisional patent application is not examined,
and automatically expires 12 months after its filing date.
As a result, a provisional patent application cannot mature into
a patent. The requirements for filing a provisional patent
application are not as strict as those for filing a
non-provisional patent application. Provisional applications are
often used, among other things, to establish an early filing
date for a subsequent non-provisional patent application.
The filing date of a non-provisional patent application is used
by the USPTO to determine what information is prior art when it
considers the patentability of a claimed invention. If certain
requirements are satisfied, a non-provisional patent application
can claim the benefit of the filing date of an earlier filed
provisional patent application. As a result, the filing date
accorded by the provisional patent application may remove
information that otherwise could preclude the patentability of
an invention.
Depending upon the timing, duration and specifics of FDA
approval of varespladib,
A-623,
A-001,
A-003 or one
or more new
sPLA2
compounds, one or more of the U.S. patents listed above may
be eligible for limited patent term restoration under the Drug
Price Competition and Patent Term Restoration Act of 1984,
commonly referred to as the Hatch-Waxman Act. See
Regulatory Matters Patent Term
Restoration and Marketing Exclusivity.
95
Licenses
Eli Lilly
and Shionogi & Co., Ltd.
In July 2006, we entered into a license agreement with Eli Lilly
and Shionogi & Co., Ltd., pursuant to which we
obtained an exclusive license in all countries except for Japan
to certain technology and compounds relating to
sPLA2
inhibitors. The licensed technology was largely developed under
a research and development agreement between Eli Lilly and
Shionogi & Co., Ltd., which was entered into between
the two parties in August 1992 and terminated in December 2004.
Under the agreement, we obtained exclusive rights to
(i) use licensed patent rights and know-how to identify and
develop
sPLA2
inhibitors, (ii) develop, make, have made, use, import,
offer for sale and sell licensed compounds and pharmaceutical
formulations thereof, including varespladib,
A-001,
A-003 and
other
sPLA2
inhibitors and (iii) grant sublicenses. The licensed patent
rights include a specific set of previously filed U.S. and
foreign patents and applications, as well as any applications
filed after the execution date by Eli Lilly or
Shionogi & Co., Ltd. that relate to licensed know-how.
Certain patents and applications within the licensed patent
rights are defined as core patents. Although the
agreement does not allow us to sell or offer for sale licensed
products in Japan, it does allow us to conduct preclinical and
clinical studies in Japan in support of applications for
marketing authorization outside of Japan, and to make and have
made licensed products in Japan for use or sale outside of
Japan. Eli Lilly and Shionogi & Co., Ltd. retain the
right to use licensed products for research purposes only. Eli
Lilly also retains the right to conduct studies of specific
compounds in animals for research purposes, but only with our
prior written approval. In addition, Shionogi & Co.,
Ltd. retains the non-exclusive right to make and have made
licensed products for supply to us, as well as its rights to
continue research, development and marketing of licensed
technology in Japan.
Upon entering into the license agreement, we took over all
prosecution and maintenance of core patents prosecuted and
maintained by Eli Lilly prior to the agreement. All core patents
prosecuted and maintained by Shionogi & Co., Ltd.
prior to the agreement remained under the control of
Shionogi & Co., Ltd. Licensed patent rights that were
not classified as core remained under the control of Eli Lilly
and Shionogi & Co., Ltd. However, control of certain
of these patents and applications has since been transferred to
us following the decision by Eli Lilly or Shionogi &
Co., Ltd. to discontinue prosecution and maintenance.
Upon entering into the license agreement, we made one-time
payments of cash in the amount of $250,000 and issued shares of
convertible preferred stock with a total aggregate value of
$2.3 million to Eli Lilly and Shionogi & Co.,
Ltd. In addition, we are required to make various milestone
payments, including payment upon initiation of the first Phase 3
clinical study for a particular product. We amended the
milestone payment terms with each of Eli Lilly and
Shionogi & Co., Ltd. to no later than 12 months
from the enrollment of the first patient in a Phase 3 clinical
study for varespladib. In consideration for the extension, the
milestone payments increased to $1.75 million to each
party. The $1.75 million milestone payment to Eli Lilly was
paid in the form of 265,957 shares of our common stock
issued at the price per share at which shares were sold to the
public in our initial public offering, minus any per-share
underwriting discounts, commissions or fees. The
$1.75 million milestone payment to Shionogi &
Co., Ltd. was paid in the form of 265,957 shares of our
common stock issued at the price per share at which shares were
sold to the public in our initial public offering, minus any
per-share underwriting discounts, commissions or fees. We are
also required to pay tiered royalty payments on net sales, which
increase as a percentage as net sales increase. Both the
milestone and royalty payment schedules vary depending on the
specific formulation (e.g., oral versus intravenously
administered). For varespladib, we are required to pay up to
$3.5 million (as discussed above) upon achievement of
certain clinical development milestones and up to
$32.0 million upon achievement of certain approval and
post-approval sales milestones. For
A-001, we
are required to pay up to $3.0 million upon achievement of
certain clinical development milestones and up to
$25.0 million upon
96
achievement of certain approval and post-approval sales
milestones. For other product formulations that we are not
currently developing, we would be required to pay up to
$2.0 million upon achievement of certain clinical
development milestones and up to $35.5 million upon
achievement of certain approval and post-approval sales
milestones. Our royalty payments vary based upon type of
formulation and annual net sales, but generally range from the
mid-single digits to the low double digits. Our royalty payment
obligations for a particular licensed product in a particular
country begin on the date of the first commercial sale of the
licensed product in that country, and end upon the later of
10 years from the date of first commercial sale in that
country or the first date on which a generic version of the
licensed product reaches a 25% total market share in that
country.
The license agreement will remain in effect for the length of
our royalty obligation on a
product-by-product
and
country-by-country
basis, unless we elect to terminate earlier or until termination
by mutual agreement. Upon expiration of the agreement, our
license will remain in effect and will convert to an
irrevocable, perpetual royalty-free license. If we fail to meet
our obligations under the agreement, Eli Lilly or
Shionogi & Co., Ltd. can terminate the agreement,
resulting in a loss of our exclusive rights to the licensed
technology.
Amgen
In December 2007, we entered into a license agreement with
Amgen, pursuant to which we obtained an exclusive worldwide
license to certain technology and compounds relating to
A-623, as
well as a non-exclusive worldwide license to technology relating
to peptibody compositions and formulations.
Under the agreement, we obtained exclusive rights under the
licensed patents and know-how to research, develop, make, have
made, use, sell, offer for sale and import pharmaceutical
products containing
A-623, as
well as the right to grant sublicenses. The licensed patents
included a specific set of previously filed U.S. and
foreign patents and applications, as well as any applications
filed after the execution date by Amgen and covering licensed
know-how. During the period of the agreement, we are responsible
for the filing, prosecution, defense and maintenance of all
exclusively licensed
A-623
patents and applications. Amgen retains the right to review all
documents relating to said filing, prosecution, defense and
maintenance, and we are required to incorporate all reasonable
comments or suggestions that Amgen makes with regard to these.
During the seven-year period after execution of the agreement,
Amgen is prohibited from clinically developing or
commercializing any BAFF peptibody. Similarly, we are prohibited
during the term of the agreement from clinically developing or
commercializing any molecule other than
A-623 that
modulates BAFF as the primary intended therapeutic mechanism of
action.
The license agreement provided for a first installment fee of
$3.0 million and a second installment fee of
$3.0 million upon the earlier of our termination of the
agreement or February 1, 2009. We have paid all of these
up-front fees. In addition, we are required to make various
milestone payments upon the achievement of certain development,
regulatory and commercial objectives, including payment upon
initiation of the first Phase 3 clinical study for any
A-623
formulation. We are also required to pay up to
$10.0 million upon achievement of certain pre-approval
clinical development milestones and up to $23.0 million
upon achievement of certain post- approval milestones.
Furthermore, we are required to make tiered quarterly royalty
payments on net sales, which increase as a percentage from the
high single digits to the low double digits as net sales
increase. Our royalty payment obligations for a particular
product in a particular country begin on the date of the first
commercial sale of the licensed product in that country, and end
upon the later of 10 years from the date of first
commercial sale in that country or the expiration date of the
last valid claim of a licensed patent that covers the
manufacture, use or sale, offer to sell or import of the product.
97
The license agreement will remain in effect until we elect to
terminate, or until termination for material breach by either
party or insolvency on our part. Under these terms, Amgen can
terminate the agreement if we fail to meet our obligations,
resulting in a loss of our exclusive rights to the licensed
technology.
On October 16, 2009, we executed an amendment to the
license agreement to amend certain terms and conditions,
including the terms and conditions on which technology transfer
activities, support and assistance would be provided to us and
forgiveness of accrued interest on an unpaid license fee, which
has since been paid in full.
Manufacturing
and Supply
We currently rely on contract manufacturers to produce drug
substances and drug products required for our clinical studies
under cGMP with oversight by our internal managers. We plan to
continue to rely upon contract manufacturers and, potentially,
collaboration partners to manufacture commercial quantities of
our product candidates if and when approved for marketing by the
FDA. We currently rely on a single manufacturer for the
preclinical and clinical supplies of each of our product
candidates and do not currently have agreements in place for
redundant supply or a second source for any of our product
candidates. We believe that there are other manufacturers and
alternate sources of supply that can satisfy our clinical study
requirements without significant delay or material additional
costs should our current manufacturer fail to meet our needs.
However, should a supplier or a manufacturer on which we have
relied to produce a product candidate provide us with a faulty
product or such product is later recalled, we would likely
experience significant delays and material additional costs.
Sales and
Marketing
Given our stage of development, we have not developed a
commercial organization or distribution capabilities. We expect
that we would develop these capabilities once we receive Phase 3
data in contemplation of FDA approval and the commercial launch
of our product candidates. In order to commercialize any of our
product candidates, we must develop these capabilities
internally or through collaboration with third parties. In
selected therapeutic areas where we feel that any approved
products can be commercialized by a specialty sales force that
calls on a limited and focused group of physicians, acute care
and orphan indications such as acute coronary syndrome and acute
chest syndrome associated with sickle cell disease, we may seek
to commercialize these product candidates alone. In therapeutic
areas that require a large sales force selling to a large and
diverse prescribing population, such as chronic indications such
as CAD, we currently plan to partner with third parties to
commercialize our product candidates while retaining rights to
co-promote our products to a select audience of high prescribing
physicians in the United States only, thereby supplementing or
enhancing the efforts of a commercial partner. We also plan to
seek commercialization partners for products in non-specialty
and international markets.
In North America and Western Europe, patients in the target
markets for our product candidates are largely managed by
medical specialists in the areas of cardiology and internal
medicine. Historically, companies have experienced substantial
commercial success through the deployment of specialized sales
forces that can address a majority of key prescribers,
particularly within the cardiovascular disease marketplace.
Therefore, we expect to utilize a specialized sales force in
North America for the sales and marketing of product candidates
that we may successfully develop. Based upon sales models, we
estimate that we could effectively promote (supplementing a
commercial partners sales efforts) the treatment of acute
coronary syndrome to 3,000 cardiologists with approximately 300
sales representatives in North America and Western Europe. If we
obtain additional label indications for varespladib or
A-001, we
may choose to increase our sales force size to promote these new
uses. Due to their concentrated and focused nature, specialty
target audiences may be reached with more focused and
cost-effective marketing campaigns. Outside of North America,
and in situations or markets where a more favorable return may
be realized through licensing commercial rights to a third
party, we may license a portion or all of our
98
commercial rights in a territory to a third party in exchange
for one or more of the following: up-front payments, research
funding, development funding, milestone payments and royalties
on drug sales.
We intend to build the commercial infrastructure necessary to
bring varespladib,
A-623 and
A-001 to
market alone or in collaboration with a co-development or
co-promotion partner. In addition to a specialty sales force,
sales management, internal sales support and an internal
marketing group, we will need to establish capabilities to
manage key accounts, such as managed care organizations,
group-purchasing organizations, specialty pharmacies and
government accounts. We may also choose to employ medical sales
liaisons personnel to support the product.
Regulatory
Matters
Government
Regulation and Product Approval
Government authorities in the United States at the federal,
state and local level, and other countries, extensively
regulate, among other things, the research, development,
testing, manufacture, quality control, approval, labeling,
packaging, storage, record-keeping, promotion, advertising,
distribution, marketing, export and import of products such as
those we are developing. Our product candidates must be approved
by the FDA through the new drug application, or NDA, process,
and our biological product candidate,
A-623, must
be approved by the FDA through the biologics license
application, or BLA, process before they may legally be marketed
in the United States.
United
States Drug Development Process
In the United States, the FDA regulates drugs under the Federal
Food, Drug, and Cosmetic Act, or FDCA, and implementing
regulations and biological products under both the FDCA and the
Public Health Service Act, or the PHSA, and implementing
regulations. The process of obtaining regulatory approvals and
compliance with appropriate federal, state, local and foreign
statutes and regulations require the expenditure of substantial
time and financial resources. Failure to comply with the
applicable U.S. requirements at any time during the product
development process, approval process, or after approval, may
subject an applicant to administrative or judicial sanctions.
These sanctions could include the FDAs refusal to approve
pending applications, withdrawal of an approval, a clinical
hold, warning letters, product recalls, product seizures, total
or partial suspension of production or distribution,
injunctions, fines, refusals of government contracts,
restitution, disgorgement or civil or criminal penalties. The
process required by the FDA before a drug or biological product
may be marketed in the United States generally involves the
following:
|
|
|
|
|
completion of preclinical laboratory tests, animal studies and
formulation studies according to Good Laboratory Practices
regulations;
|
|
|
|
submission to the FDA of an IND, which must become effective
before human clinical studies may begin;
|
|
|
|
performance of adequate and well-controlled human clinical
studies according to Good Clinical Practices, or GCP, to
establish the safety and efficacy of the proposed drug or
biological product for its intended use;
|
|
|
|
submission to the FDA of an NDA for a new drug or BLA for a
biological product;
|
|
|
|
satisfactory completion of an FDA inspection of the
manufacturing facility or facilities at which the drug or
biological product is produced to assess compliance with
cGMP; and
|
|
|
|
FDA review and approval of the NDA or BLA.
|
99
The testing and approval process requires substantial time,
effort and financial resources and we cannot be certain that any
approvals for our product candidates will be granted on a timely
basis, if at all.
Once a pharmaceutical or biological product candidate is
identified for development, it enters the preclinical testing
stage. Preclinical tests include laboratory evaluations of
product chemistry, toxicity, formulation and stability, as well
as animal studies to assess its potential safety and efficacy.
An IND sponsor must submit the results of the preclinical tests,
together with manufacturing information, analytical data and any
available clinical data or literature, to the FDA as part of the
IND. The sponsor will also include a protocol detailing, among
other things, the objectives of the initial clinical study, the
parameters to be used in monitoring safety and the effectiveness
criteria to be evaluated if the initial clinical study lends
itself to an efficacy evaluation. Some preclinical testing may
continue even after the IND is submitted. The IND automatically
becomes effective 30 days after receipt by the FDA, unless
the FDA places the clinical study on a clinical hold within that
30-day time
period. In such a case, the IND sponsor and the FDA must resolve
any outstanding concerns before the clinical study can begin.
Clinical holds also may be imposed by the FDA at any time before
or during studies due to safety concerns or non-compliance.
All clinical studies must be conducted under the supervision of
one or more qualified investigators in accordance with GCP
regulations. These regulations include the requirement that all
research subjects provide informed consent. Further, an
institutional review board, or IRB, must review and approve the
plan for any clinical study before it commences at any
institution. An IRB considers, among other things, whether the
risks to individuals participating in the studies are minimized
and are reasonable in relation to anticipated benefits. The IRB
also approves the information regarding the clinical study and
the consent form that must be provided to each clinical study
subject or his or her legal representative and must monitor the
clinical study until completed.
Each new clinical protocol and any amendments to the protocol
must be submitted to the IND for FDA review, and to the IRBs for
approval. Protocols detail, among other things, the objectives
of the clinical study, dosing procedures, subject selection and
exclusion criteria, and the parameters to be used to monitor
subject safety.
Human clinical studies are typically conducted in three
sequential phases that may overlap or be combined:
|
|
|
|
|
Phase 1. The product is initially
introduced into healthy human subjects and tested for safety,
dosage tolerance, absorption, metabolism, distribution and
excretion. In the case of some products for severe or
life-threatening diseases, especially when the product may be
too inherently toxic to ethically administer to healthy
volunteers, the initial human testing is often conducted in
patients.
|
|
|
|
Phase 2. Involves studies in a limited
patient population to identify possible adverse effects and
safety risks, to preliminarily evaluate the efficacy of the
product for specific targeted diseases and to determine dosage
tolerance and optimal dosage and schedule.
|
|
|
|
Phase 3. Clinical studies are
undertaken to further evaluate dosage, clinical efficacy and
safety in an expanded patient population at geographically
dispersed clinical study sites. These studies are intended to
establish the overall risk/benefit ratio of the product and
provide an adequate basis for product labeling.
|
Progress reports detailing the results of the clinical studies
must be submitted at least annually to the FDA and safety
reports must be submitted to the FDA and the investigators for
serious and unexpected adverse events. Phase 1, Phase 2 and
Phase 3 testing may not be completed successfully within any
100
specified period, if at all. The FDA or the sponsor may suspend
or terminate a clinical study at any time on various grounds,
including a finding that the research subjects or patients are
being exposed to an unacceptable health risk. Similarly, an IRB
can suspend or terminate approval of a clinical study at its
institution if the clinical study is not being conducted in
accordance with the IRBs requirements or if the drug or
biological product has been associated with unexpected serious
harm to patients.
Concurrent with clinical studies, companies usually complete
additional animal studies and must also develop additional
information about the chemistry and physical characteristics of
the product and finalize a process for manufacturing the product
in commercial quantities in accordance with cGMP requirements.
The manufacturing process must be capable of consistently
producing quality batches of the product candidate and, among
other things, the manufacturer must develop methods for testing
the identity, strength, quality and purity of the final product.
Additionally, appropriate packaging must be selected and tested
and stability studies must be conducted to demonstrate that the
product candidate does not undergo unacceptable deterioration
over its shelf life.
U.S.
Review and Approval Processes
The results of product development, preclinical studies and
clinical studies, along with descriptions of the manufacturing
process, analytical tests conducted on the drug or biological
product, proposed labeling and other relevant information, are
submitted to the FDA as part of an NDA for a new drug or BLA for
a biological product, requesting approval to market the product.
The submission of an NDA or BLA is subject to the payment of a
substantial user fee; a waiver of such fee may be obtained under
certain limited circumstances.
In addition, under the Pediatric Research Equity Act of 2003, or
PREA, which was reauthorized under the Food and Drug
Administration Amendments Act of 2007, an NDA or BLA or
supplement to an NDA or BLA must contain data to assess the
safety and effectiveness of the drug or biological product for
the claimed indications in all relevant pediatric subpopulations
and to support dosing and administration for each pediatric
subpopulation for which the product is safe and effective. The
FDA may grant deferrals for submission of data or full or
partial waivers. Unless otherwise required by regulation, PREA
does not apply to any drug or biological product for an
indication for which orphan designation has been granted.
The FDA reviews all NDAs and BLAs submitted to ensure that they
are sufficiently complete for substantive review before it
accepts them for filing. The FDA may request additional
information rather than accept a NDA or BLA for filing. In this
event, the NDA or BLA must be re-submitted with the additional
information. The re-submitted application also is subject to
review before the FDA accepts it for filing. Once the submission
is accepted for filing, the FDA begins an in-depth substantive
review. The FDA reviews an NDA to determine, among other things,
whether a product is safe and effective for its intended use and
whether its manufacturing is cGMP-compliant to assure and
preserve the products identity, strength, quality and
purity. The FDA reviews a BLA to determine, among other things,
whether the product is safe, has an acceptable purity profile
and is adequately potent, and whether its manufacturing meets
standards designed to assure the products continued
identity, sterility, safety, purity and potency. Before
approving an NDA or BLA, the FDA will inspect the facility or
facilities where the product is manufactured. The FDA will not
approve an application unless it determines that the
manufacturing processes and facilities are in compliance with
cGMP requirements and adequate to assure consistent production
of the product within required specifications. The FDA may refer
the NDA or BLA to an advisory committee for review, evaluation
and recommendation as to whether the application should be
approved and under what conditions. An advisory committee is a
panel of experts who provide advice and recommendations when
requested by the FDA on matters of importance that come before
the agency. The FDA is not bound by the recommendation of an
advisory committee but it generally follows such recommendations.
101
The approval process is lengthy and difficult and the FDA may
refuse to approve an NDA or BLA if the applicable regulatory
criteria are not satisfied or may require additional clinical
data or other data and information. Even if such data and
information is submitted, the FDA may ultimately decide that the
NDA or BLA does not satisfy the criteria for approval. Data
obtained from clinical studies are not always conclusive and the
FDA may interpret data differently than we interpret the same
data. The FDA will issue a complete response letter if the
agency decides not to approve the NDA or BLA in its present
form. The complete response letter usually describes all of the
specific deficiencies in the NDA or BLA identified by the FDA.
The deficiencies identified may be minor, for example, requiring
labeling changes, or major, for example, requiring additional
clinical studies. Additionally, the complete response letter may
include recommended actions that the applicant might take to
place the application in a condition for approval. If a complete
response letter is issued, the applicant may either resubmit the
NDA or BLA, addressing all of the deficiencies identified in the
letter, or withdraw the application
If a product receives regulatory approval, the approval may be
significantly limited to specific diseases and dosages or the
indications for use may otherwise be limited, which could
restrict the commercial value of the product. Further, the FDA
may require that certain contraindications, warnings or
precautions be included in the product labeling. In addition,
the FDA may require Phase 4 testing which involves clinical
studies designed to further assess a drug or biological
products safety and effectiveness after NDA or BLA
approval and may require testing and surveillance programs to
monitor the safety of approved products that have been
commercialized.
Patent
Term Restoration and Marketing Exclusivity
Depending upon the timing, duration and specifics of FDA
approval of the use of our product candidates, some of our
U.S. patents may be eligible for limited patent term
extension under the Drug Price Competition and Patent Term
Restoration Act of 1984, commonly referred to as the
Hatch-Waxman Amendments. The Hatch-Waxman Amendments permit a
patent restoration term of up to five years as compensation for
patent term lost during product development and the FDA
regulatory review process. However, patent term restoration
cannot extend the remaining term of a patent beyond a total of
14 years from the products approval date. The patent
term restoration period is generally one-half the time between
the effective date of an IND and the submission date of an NDA
plus the time between the submission date of an NDA and the
approval of that application. Only one patent applicable to an
approved drug is eligible for the extension and the application
for the extension must be submitted prior to the expiration of
the patent. The USPTO, in consultation with the FDA, reviews and
approves the application for any patent term extension or
restoration. In the future, we intend to apply for restorations
of patent term for some of our currently owned or licensed
patents to add patent life beyond their current expiration
dates, depending on the expected length of the clinical studies
and other factors involved in the filing of the relevant NDA.
Market exclusivity provisions under the FDCA can also delay the
submission or the approval of certain applications. The FDCA
provides a five-year period of non-patent marketing exclusivity
within the United States to the first applicant to gain approval
of an NDA for a new chemical entity. A drug is a new chemical
entity if the FDA has not previously approved any other new drug
containing the same active moiety, which is the molecule or ion
responsible for the action of the drug substance. During the
exclusivity period, the FDA may not accept for review an
abbreviated new drug application, or ANDA, or a 505(b)(2) NDA
submitted by another company for another version of such drug
where the applicant does not own or have a legal right of
reference to all the data required for approval. However, an
application may be submitted after four years if it contains a
certification of patent invalidity or non-infringement. The FDCA
also provides three years of marketing exclusivity for an NDA,
505(b)(2) NDA or supplement to an existing NDA if new clinical
investigations, other than bioavailability studies, that were
conducted or sponsored by the applicant are deemed by the FDA to
be essential to the approval of the application, for example new
indications, dosages or strengths of an existing drug. This
102
three-year exclusivity covers only the conditions associated
with the new clinical investigations and does not prohibit the
FDA from approving ANDAs for drugs containing the original
active agent. Five-year and three-year exclusivity will not
delay the submission or approval of a full NDA. However, an
applicant submitting a full NDA would be required to conduct or
obtain a right of reference to all of the preclinical studies
and adequate and well-controlled clinical studies necessary to
demonstrate safety and effectiveness. HR 3590 provides
12 years of data exclusivity for innovator biologics.
During this exclusivity period, competitors are barred from
relying on the innovators safety and efficacy data to gain
FDA approval. Therefore, a competitor seeking to obtain
marketing approval during this exclusivity period would be
required to conduct their own preclinical and clinical studies.
Pediatric exclusivity is another type of exclusivity in the
United States. Pediatric exclusivity, if granted, provides an
additional six months to an existing exclusivity or statutory
delay in approval resulting from a patent certification. This
six-month exclusivity, which runs from the end of other
exclusivity protection or patent delay, may be granted based on
the voluntary completion of a pediatric study in accordance with
an FDA-issued Written Request for such a study. The
current pediatric exclusivity provision was reauthorized in
September 2007.
Orphan
Drug Designation
Under the Orphan Drug Act, the FDA may grant orphan designation
to a drug or biological product intended to treat a rare disease
or condition, which is generally a disease or condition that
affects fewer than 200,000 individuals in the United States, or
more than 200,000 individuals in the United States and for which
there is no reasonable expectation that the cost of developing
and making a drug or biological product available in the United
States for this type of disease or condition will be recovered
from sales of the product. Orphan product designation must be
requested before submitting an NDA or BLA. After the FDA grants
orphan product designation, the identity of the therapeutic
agent and its potential orphan use are disclosed publicly by the
FDA. Orphan product designation does not convey any advantage in
or shorten the duration of the regulatory review and approval
process.
If a product that has orphan designation subsequently receives
the first FDA approval for the disease or condition for which it
has such designation, the product is entitled to orphan product
exclusivity, which means that the FDA may not approve any other
applications to market the same drug or biological product for
the same indication, except in very limited circumstances, for
seven years. Competitors, however, may receive approval of
different products for the indication for which the orphan
product has exclusivity or obtain approval for the same product
but for a different indication for which the orphan product has
exclusivity. Orphan product exclusivity also could block the
approval of one of our products for seven years if a competitor
obtains approval of the same drug or biological product as
defined by the FDA or if our product candidate is determined to
be contained within the competitors product for the same
indication or disease. If a drug or biological product
designated as an orphan product receives marketing approval for
an indication broader than what is designated, it may not be
entitled to orphan product exclusivity.
The FDA also administers a clinical research grants program,
whereby researchers may compete for funding to conduct clinical
studies to support the approval of drugs, biologics, medical
devices and medical foods for rare diseases and conditions. A
product does not have to be designated as an orphan product to
be eligible for the grant program. An application for an orphan
grant should propose one discrete clinical study to facilitate
FDA approval of the product for a rare disease or condition. The
clinical study may address an unapproved new product or an
unapproved new use for a product already on the market.
103
Expedited
Development and Review Programs
The FDA has a fast track program that is intended to expedite or
facilitate the process for reviewing new drugs and biological
products that meet certain criteria. Specifically, new drugs and
biological products are eligible for fast track designation if
they are intended to treat a serious or life-threatening
condition and demonstrate the potential to address unmet medical
needs for the condition. Fast track designation applies to the
combination of the product and the specific indication for which
it is being studied. For a fast track product, the FDA may
consider for review on a rolling basis sections of the NDA or
BLA before the complete application is submitted, if the sponsor
provides a schedule for the submission of the sections of the
NDA or BLA, the FDA agrees to accept sections of the NDA or BLA
and determines that the schedule is acceptable, and the sponsor
pays any required user fees upon submission of the first section
of the NDA or BLA.
A fast track product may also be eligible for other types of FDA
programs intended to expedite development and review, such as
priority review and accelerated approval. A fast track product
is eligible for priority review if it has the potential to
provide safe and effective therapy where no satisfactory
alternative therapy exists or a significant improvement in the
treatment, diagnosis or prevention of a disease compared to
marketed products. The FDA will attempt to direct additional
resources to the evaluation of an application for a new drug or
biological product designated for priority review in an effort
to facilitate the review. Additionally, a fast track product may
be eligible for accelerated approval. Drug or biological
products studied for their safety and effectiveness in treating
serious or life-threatening illnesses and that provide
meaningful therapeutic benefit over existing treatments may
receive accelerated approval, which means that they may be
approved on the basis of adequate and well-controlled clinical
studies establishing that the product has an effect on a
surrogate endpoint that is reasonably likely to predict a
clinical benefit, or on the basis of an effect on a clinical
endpoint other than survival or irreversible morbidity. As a
condition of approval, the FDA may require that a sponsor of a
drug or biological product receiving accelerated approval
perform adequate and well-controlled post-marketing clinical
studies. Fast track designation, priority review and accelerated
approval do not change the standards for approval but may
expedite the development or approval process.
We have been granted fast track designation for our product
candidate,
A-001, for
the prevention of acute chest syndrome associated with sickle
cell disease in at-risk patients. Even though we have received
fast track designation for
A-001, the
FDA may later decide that
A-001 no
longer meets the conditions for qualification. In addition,
obtaining fast track designation may not provide us with a
material commercial advantage.
Post-Approval
Requirements
Any drug or biological products for which we receive FDA
approvals are subject to continuing regulation by the FDA,
including, among other things, record-keeping requirements,
reporting of adverse experiences with the product, providing the
FDA with updated safety and efficacy information, product
sampling and distribution requirements, complying with certain
electronic records and signature requirements and complying with
FDA promotion and advertising requirements. The FDA strictly
regulates labeling, advertising, promotion and other types of
information on products that are placed on the market. Drugs and
biological products may be promoted only for the approved
indications and in accordance with the provisions of the
approved label. Further, manufacturers of drugs and biological
products must continue to comply with cGMP requirements, which
are extensive and require considerable time, resources and
ongoing investment to ensure compliance. In addition, changes to
the manufacturing process generally require prior FDA approval
before being implemented and other types of changes to the
approved product, such as adding new indications and additional
labeling claims, are also subject to further FDA review and
approval.
104
Drug and biological product manufacturers and other entities
involved in the manufacturing and distribution of approved drugs
or biological products are required to register their
establishments with the FDA and certain state agencies, and are
subject to periodic unannounced inspections by the FDA and
certain state agencies for compliance with cGMP and other laws.
The cGMP requirements apply to all stages of the manufacturing
process, including the production, processing, sterilization,
packaging, labeling, storage and shipment of the drug or
biological product. Manufacturers must establish validated
systems to ensure that products meet specifications and
regulatory standards, and test each product batch or lot prior
to its release.
Manufacturers of biological products must also report to the FDA
any deviations from cGMP that may affect the safety, purity or
potency of a distributed product; or any unexpected or
unforeseeable event that may affect the safety, purity or
potency of a distributed product. The regulations also require
investigation and correction of any deviations from cGMP and
impose documentation requirements.
We rely, and expect to continue to rely, on third parties for
the production of clinical and commercial quantities of our
products. Future FDA and state inspections may identify
compliance issues at the facilities of our contract
manufacturers that may disrupt production or distribution or may
require substantial resources to correct.
The FDA may withdraw a product approval if compliance with
regulatory standards is not maintained or if problems occur
after the product reaches the market. Later discovery of
previously unknown problems with a product may result in
restrictions on the product or even complete withdrawal of the
product from the market. Further, the failure to maintain
compliance with regulatory requirements may result in
administrative or judicial actions, such as fines, warning
letters, holds on clinical studies, product recalls or seizures,
product detention or refusal to permit the import or export of
products, refusal to approve pending applications or
supplements, restrictions on marketing or manufacturing,
injunctions or civil or criminal penalties.
In addition, from time to time, legislation is drafted,
introduced and passed in Congress that could significantly
change the statutory provisions governing the approval,
manufacturing and marketing of products regulated by the FDA.
For example, in September 2007, the Food and Drug Administration
Amendments Act of 2007 was enacted, giving the FDA enhanced
post-market authority, including the authority to require
post-market studies and clinical studies, labeling changes based
on new safety information and compliance with a risk evaluation
and mitigation strategy, or REMS, approved by the FDA. In
determining whether a REMS is necessary, the FDA must consider
the size of the population likely to use the drug or biological
product, the seriousness of the disease or condition to be
treated, the expected benefit of the product, the duration of
treatment, the seriousness of known or potential adverse events
for varespladib and whether the product is a new molecular
entity. We have submitted a REMS as an appendix to the SPA. If
the FDA determines our REMS is necessary, we must submit a REMS
plan as part of an NDA or BLA. The FDA may require that a REMS
include various elements, such as a medication guide, patient
package insert, a communication plan to educate health care
providers, limitations on who may prescribe or dispense the
product, or other measures.
Failure to comply with any requirements under the new law may
result in significant penalties. The new law also authorizes
significant civil money penalties for the dissemination of false
or misleading
direct-to-consumer
advertisements and allows the FDA to require companies to submit
direct-to-consumer
television drug advertisements for FDA review prior to public
dissemination. Additionally, the new law expands the clinical
study registry so that sponsors of all clinical studies, except
for Phase 1 clinical studies, are required to submit certain
clinical study information for inclusion in the clinical study
registry data bank. In addition to new legislation, the FDA
regulations and policies are often revised or reinterpreted by
the agency in ways that may significantly affect our business
and our
105
products. It is impossible to predict whether further
legislative or FDA regulation or policy changes will be enacted
or implemented and what the impact of such changes, if any, may
be.
Foreign
Regulation
In addition to regulations in the United States, we will be
subject to a variety of foreign regulations governing clinical
studies and commercial sales and distribution of our products to
the extent we choose to sell any products outside of the United
States. Whether or not we obtain FDA approval for a product, we
must obtain approval of a product by the comparable regulatory
authorities of foreign countries before we can commence clinical
studies or marketing of the product in those countries. The
approval process varies from country to country and the time may
be longer or shorter than that required for FDA approval. The
requirements governing the conduct of clinical studies, product
licensing, pricing and reimbursement vary greatly from country
to country.
In the European Union, our products are subject to extensive
regulatory requirements, which provide, among other things, that
no medicinal product may be placed on the market of a European
Union member state unless a marketing authorization has been
issued by the European Medicines Agency or a national competent
authority. European Union member states require both regulatory
clearance by the national competent authority and a favorable
ethics committee opinion prior to the commencement of a clinical
study.
Under the European Union regulatory systems, we may submit
marketing authorization applications either under a centralized
or decentralized procedure. The centralized procedure provides
for the grant of a single marketing authorization that is valid
for all European Union member states. The centralized procedure
is compulsory for medicines produced by certain biotechnological
processes, products with a new active substance indicated for
the treatment of certain diseases such as neurodegenerative
disorder or diabetes and products designated as orphan medicinal
products, and optional for those products which are highly
innovative or for which a centralized process is in the interest
of patients. The decentralized procedure of approval provides
for approval by one or more other, or concerned, member states
of an assessment of an application performed by one member
state, known as the reference member state. Under the
decentralized approval procedure, an applicant submits an
application, or dossier, and related materials (draft summary of
product characteristics, draft labeling and package leaflet) to
the reference member state and concerned member states. The
reference member state prepares a draft assessment and drafts of
the related materials within 120 days after receipt of a
valid application. Within 90 days of receiving the
reference member states assessment report, each concerned
member state must decide whether to approve the assessment
report and related materials. If a member state cannot approve
the assessment report and related materials on the grounds of
potential serious risk to public health, the disputed points may
eventually be referred to the European Commission, whose
decision is binding on all member states.
Reimbursement
Sales of pharmaceutical products depend significantly on the
availability of third-party reimbursement. Third-party payors
include government health administrative authorities, including
at the federal and state level, managed care providers, private
health insurers and other organizations. We anticipate
third-party payors will provide reimbursement for our products.
However, these third-party payors are increasingly challenging
the price and examining the cost-effectiveness of medical
products and services. In addition, significant uncertainty
exists as to the reimbursement status of newly approved health
care products. We may need to conduct expensive pharmacoeconomic
studies in order to demonstrate the cost-effectiveness of our
products. Our product candidates may not be considered
cost-effective. It is time consuming and expensive for us to
seek reimbursement from third-party payors. Reimbursement may
not be available or sufficient to allow us to sell our products
on a competitive and profitable basis.
106
In addition, the U.S. Congress and state legislatures from
time to time propose and adopt initiatives aimed at cost
containment, which could impact our ability to sell our products
profitably. For example, in March 2010, President Obama signed
into law the Patient Protection and Affordable Care Act, and the
associated reconciliation bill, which we refer to collectively
as the Health Care Reform Law, a sweeping law intended to
broaden access to health insurance, reduce or constrain the
growth of healthcare spending, enhance remedies against fraud
and abuse, add new transparency requirements for healthcare and
health insurance industries, impose new taxes and fees on the
health industry and impose additional health policy reforms.
Effective October 1, 2010, the Health Care Reform Law
revises the definition of average manufacturer price
for reporting purposes, which could increase the amount of
Medicaid drug rebates to states once the provision is effective.
Further, beginning in 2011, the new law imposes a significant
annual fee on companies that manufacture or import certain
branded prescription drug products and biologic agents.
Substantial new provisions affecting compliance also have been
enacted, which may require us to modify our business practices
with healthcare practitioners. We will not know the full effects
of the Health Care Reform Law until applicable federal and state
agencies issue regulations or guidance under the new law.
Although it is too early to determine the effect of the Health
Care Reform Law, the new law appears likely to continue the
pressure on pharmaceutical pricing, especially under the
Medicare program, and also may increase our regulatory burdens
and operating costs.
The Medicare Prescription Drug, Improvement, and Modernization
Act of 2003, or the MMA, imposed new requirements for the
distribution and pricing of prescription drugs for Medicare
beneficiaries, and included a major expansion of the
prescription drug benefit under a new Medicare Part D.
Medicare Part D went into effect on January 1, 2006.
Under Part D, Medicare beneficiaries may enroll in
prescription drug plans offered by private entities which will
provide coverage of outpatient prescription drugs. Part D
plans include both stand-alone prescription drug benefit plans
and prescription drug coverage as a supplement to Medicare
Advantage plans. Unlike Medicare Part A and B, Part D
coverage is not standardized. Part D prescription drug plan
sponsors are not required to pay for all covered Part D
drugs, and each drug plan can develop its own drug formulary
that identifies which drugs it will cover and at what tier or
level. However, Part D prescription drug formularies must
include drugs within each therapeutic category and class of
covered Part D drugs, though not necessarily all the drugs
in each category or class. Any formulary used by a Part D
prescription drug plan must be developed and reviewed by a
pharmacy and therapeutic committee.
It is not clear what effect the MMA will have on the prices paid
for currently approved drugs and the pricing options for new
drugs approved after January 1, 2006. Government payment
for some of the costs of prescription drugs may increase demand
for products for which we receive marketing approval. However,
any negotiated prices for our products covered by a Part D
prescription drug plan will likely be lower than the prices we
might otherwise obtain. Moreover, while the MMA applies only to
drug benefits for Medicare beneficiaries, private payors often
follow Medicare coverage policy and payment limitations in
setting their own payment rates. Any reduction in payment that
results from the MMA may result in a similar reduction in
payments from non-governmental payors.
There are also laws that govern a companys eligibility to
participate in Medicare and Medicaid reimbursements. For
example, a company may be debarred from participation if it is
found to have violated federal anti-kickback laws, which could
have a significant effect on a companys ability to operate
its business.
The cost of pharmaceuticals continues to generate substantial
governmental and third-party payor interest. We expect that the
pharmaceutical industry will experience pricing pressures due to
the trend toward managed healthcare, the increasing influence of
managed care organizations, and additional legislative
proposals. Indeed, we expect that there will continue to be a
number of federal and state proposals to implement governmental
pricing controls and limit the growth of healthcare costs,
including the cost of prescription drugs. At the present time,
Medicare is prohibited from negotiating directly with
pharmaceutical
107
companies for drugs. However, Congress is considering passing
legislation that would lift the ban on federal negotiations.
While we cannot predict whether such legislative or regulatory
proposals will be adopted, the adoption of such proposals could
harm our business, financial condition and results of operations.
Some third-party payors also require pre-approval of coverage
for new or innovative drug therapies before they will reimburse
healthcare providers that use such therapies. While we cannot
predict whether any proposed cost-containment measures will be
adopted or otherwise implemented in the future, the announcement
or adoption of these proposals could have a material adverse
effect on our ability to obtain adequate prices for our product
candidates and operate profitably.
In addition, in some foreign countries, the proposed pricing for
a drug must be approved before it may be lawfully marketed. The
requirements governing drug pricing vary widely from country to
country. For example, the European Union provides options for
its member states to restrict the range of medicinal products
for which their national health insurance systems provide
reimbursement and to control the prices of medicinal products
for human use. A member state may approve a specific price for
the medicinal product or it may instead adopt a system of direct
or indirect controls on the profitability of the company placing
the medicinal product on the market. In some countries, pricing
negotiations with governmental authorities can take six to
12 months or longer after the receipt of marketing
approval. To obtain reimbursement or pricing approval in some
countries, we may be required to conduct a clinical trial that
compares the cost-effectiveness of our product candidate to
other available therapies. There can be no assurance that any
country that has price controls or reimbursement limitations for
pharmaceutical products will allow favorable reimbursement and
pricing arrangements for any of our products.
Employees
As of September 30, 2010, we had 25 employees, nine of
whom hold an M.D., Ph.D. or Pharm. D. All of our employees
are engaged in administration, finance, clinical, regulatory and
business development functions. None of our employees are
represented by a labor union, and we believe that our relations
with our employees are good.
Property
and Facilities
We are currently subleasing approximately 7,800 square feet
of office space in Hayward, California, which we occupy under a
sublease that commenced on October 1, 2008 and will expire
on January 31, 2011. We believe our existing facilities are
adequate for our current needs and that any additional space we
need will be available in the future on commercially reasonable
terms.
Legal
Proceedings
We are not currently subject to any material legal proceedings.
108
MANAGEMENT
Executive
Officers and Directors
The following table sets forth information regarding our
executive officers and directors, including their ages as of
September 30, 2010.
|
|
|
|
|
|
|
Name
|
|
Age
|
|
Position
|
|
Paul F. Truex
|
|
|
42
|
|
|
Chief Executive Officer, President and Director
|
Christopher P. Lowe
|
|
|
43
|
|
|
Chief Financial Officer and Vice President of Administration
|
Colin Hislop, M.D.
|
|
|
53
|
|
|
Chief Medical Officer and Senior Vice President
|
Debra Odink, Ph.D.
|
|
|
46
|
|
|
Senior Vice President, Pharmaceutical Research and Development
|
Georgina Kilfoil
|
|
|
42
|
|
|
Senior Vice President, Product Development and Clinical
Operations
|
Christopher S.
Henney, Ph.D. |