Armata Pharmaceuticals, Inc. - Annual Report: 2005 (Form 10-K)
Table of Contents
UNITED STATES SECURITIES AND EXCHANGE COMMISSION
Washington, D.C. 20549
Form 10-K
þ
|
ANNUAL REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934 | |
For the fiscal year ended December 31, 2005 | ||
OR | ||
o
|
TRANSITION REPORT PURSUANT TO SECTION 13 OR 15(d) OF THE SECURITIES EXCHANGE ACT OF 1934 | |
For the transition period from to |
Commission file number No. 0-23930
TARGETED GENETICS CORPORATION
(Exact name of Registrant as specified in its charter)
Washington | 91-1549568 | |
(State of Incorporation) | (IRS Employer Identification No.) |
1100 Olive Way, Suite 100
Seattle, WA 98101
(Address of principal executive offices, including, zip
code)
(206) 623-7612
(Registrants telephone number, including area code)
Securities registered pursuant to Section 12(b) of the
Act:
None
Securities registered pursuant to Section 12(g) of the
Act:
Common Stock, $0.01 Par Value
Indicate by check mark if the registrant is a well-known
seasoned issuer, as defined in Rule 405 of the Securities
Act. Yes o No þ
Indicate by check mark if the registrant is not required to file
reports pursuant to Section 13 or Section 15(d) of the
Act. Yes o No þ
Indicate by check mark whether the Registrant (1) has filed
all reports required to be filed by Section 13 or 15(d) of
the Securities Exchange Act of 1934 during the preceding
12 months (or for such shorter period that the Registrant
was required to file such reports), and (2) has been
subject to such filing requirements for the past
90 days. Yes þ No o
Indicate by check mark if disclosure of delinquent filers
pursuant to Item 405 of
Regulation S-K is
not contained herein, and will not be contained, to the best of
Registrants knowledge, in definitive proxy or information
statements incorporated by reference in Part III of this
Form 10-K or any
amendment to this
Form 10-K. þ
Indicate by check mark whether the registrant is a large
accelerated filer, an accelerated filer, or a non-accelerated
filer. See definition of accelerated filer and large
accelerated filer in
Rule 12b-2 of the
Exchange Act. (Check one):
Large accelerated
filer o Accelerated
filer þ Non-accelerated
filer o
Indicate by check mark whether the registrant is a shell company
(as defined in
Rule 12b-2 of the
Act). Yes o No þ
The aggregate market value of common stock held by
non-affiliates of the Registrant as of June 30, 2005 was
approximately $48.6 million based on the closing price of
$0.81 per share of the Registrants common stock as
listed on the NASDAQ Capital Market.
Indicate the number of shares outstanding of each of the
Registrants classes of common stock as of March 13,
2006
Title of Class | Number of Shares | |
Common Stock, $0.01 par value
|
98,498,855 |
DOCUMENTS INCORPORATED BY REFERENCE
(1) The information required by Part III of this
report, to the extent not set forth in this report, is
incorporated by reference from the Proxy Statement for the 2006
annual meeting of shareholders to be held on May 8, 2006.
The definitive proxy statement for the 2006 annual meeting of
shareholders will be filed with the Securities and Exchange
Commission within 120 days after December 31, 2005,
the end of the fiscal year to which this report relates.
TARGETED GENETICS CORPORATION
ANNUAL REPORT ON
FORM 10-K
TABLE OF CONTENTS
1
Table of Contents
PART I
Item 1. | Business |
This annual report on
Form 10-K contains
forward-looking statements that involve risks and uncertainties.
Forward-looking statements include statements about our product
development and commercialization goals and expectations,
potential market opportunities, our plans for and anticipated
results of our clinical development activities and the potential
advantage of our product candidates, our cash resources and
future financial condition, our ability to obtain additional
funding or enter into strategic collaborations and other
statements that are not historical facts. Words such as
may, can be, may depend,
will, believes, estimates,
expects, anticipates, plans,
projects, intends, or statements
concerning potential or opportunity and
other words of similar meaning or the negative thereof may
identify forward-looking statements, but the absence of these
words does not mean that the statement is not forward-looking.
In making these statements, we rely on a number of assumptions
and make predictions about the future. Our actual results could
differ materially from those stated in or implied by
forward-looking statements for a number of reasons, including
the risks described in the section entitled Factors
Affecting Our Operating Results, Our Business and Our Stock
Price in Part I, Item 1A of this annual
report.
You should not unduly rely on these forward-looking
statements, which speak only as of the date of this annual
report. We undertake no obligation to publicly revise any
forward-looking statement after the date of this annual report
to reflect circumstances or events occurring after the date of
this annual report or to conform the statement to actual results
or changes in our expectations. You should, however, review the
factors, risks and other information we provide in the reports
we file from time to time with the Securities and Exchange
Commission, or SEC.
BUSINESS OVERVIEW
Targeted Genetics Corporation is a clinical-stage biotechnology
company. We are at the forefront of developing, with the goal of
commercializing, a new class of gene therapeutics. We believe
that a wide range of diseases may potentially be treated or
prevented with gene-based products. In addition to treating
diseases which have not had treatments in the past, we believe
that there is also a significant opportunity to use gene-based
products to more effectively treat diseases that are currently
treated using other therapeutic classes of drugs including
proteins, monoclonal antibodies, or small molecule drugs. We
have multiple product candidates, two of which are currently in
clinical trials. Our clinical-stage candidates are aimed at
inflammatory arthritis and HIV/ AIDS. Our preclinical product
candidates, both in development with collaboration partners, are
aimed at congestive heart failure and Huntingtons disease.
Our gene therapeutics consists of a delivery vehicle, called a
vector, and genetic material. The role of the vector is to carry
the genetic material into a target cell. Once delivered into the
cell, the gene can express or direct production of the specific
proteins encoded by the gene. Gene therapy may be used to treat
disease by replacing the missing or defective gene to facilitate
the normal protein production or gene regulation capabilities of
cells. In addition, gene delivery may be used to enable cells to
perform additional roles in the body. Gene delivery may also be
used to shut down cellular functions.
We are a leader in the preclinical and clinical development of
adeno-associated viral vector, or AAV-based gene products and in
the manufacture of AAV-vectors. We have treated over 250
subjects using AAV-based products, which we believe is more than
any other entity. Through our research and development
activities, we have acquired expertise and intellectual property
related to a variety of gene delivery technologies. We believe
that our activities to date have resulted in important
characteristics of our business, including:
Diverse product development pipeline. Each of our clinical and preclinical product candidates addresses a different market: in each case where we believe there is significant medical need for new or improved therapies. |
2
Table of Contents
We are focused on the following product development programs:
Development Status | ||||||||||||||||||||
Research & | ||||||||||||||||||||
Description | Indication | Preclinical | Phase I | Phase II | Phase III | |||||||||||||||
AAV delivery of TNF-alpha antagonist (tgAAC94)
|
Inflammatory Arthritis | xxxxxxxx | xxxxxxxx | |||||||||||||||||
AAV delivery of HIV antigen (tgAAC09)
|
HIV/AIDS | xxxxxxxx | xxxxxxxx | xxxxxxxx | ||||||||||||||||
AAV delivery of SERCA2a and phospholamban gene variants
|
Congestive Heart Failure | xxxxxxxx | ||||||||||||||||||
AAV expression of htt siRNA
|
Huntingtons disease | xxxxxxxx |
Significant development and manufacturing expertise. We believe we are leaders in the development and application of processes to manufacture our potential products at a scale amenable to late-stage clinical development and expandable to large-scale commercial production. We have established broad capabilities in applying our AAV-based gene delivery technologies to several indications, and we have developed an infrastructure that allows us to expand these capabilities into multiple product opportunities. We have built development and regulatory expertise specific to gene therapy products. We believe that our collective product development expertise, including our experience with research and development, manufacturing, quality, and clinical and regulatory matters will serve as a critical asset in attracting product collaboration partners and provides a necessary foundation to move gene therapy from product discovery to commercialization. | |
Intellectual property assets. We have developed proprietary intellectual property, including methods of transferring genetic material into cells, processes to manufacture and purify gene delivery product candidates and other proprietary technologies and processes. Because patent and license rights are important assets of our business, our strategy is to file or license patent applications to protect technology, inventions and improvements to inventions that we consider important to developing our business. We have filed or licensed numerous patents or patent applications with the United States Patent and Trademark Office, or USPTO, and foreign jurisdictions. We also rely on unpatented proprietary technology such as trade secrets, know-how and continuing technological innovations. |
Business Strategy |
Our current strategic focus entails:
| Continuing aggressive development of our pipeline. We plan to continue evaluating options in our clinical development efforts that will accelerate development and lead to nearer term clinical trial results and, ultimately commercialization. Our current priorities are the advancement of tgAAC94 as a therapy to treat inflammatory arthritis and the advancement of our partnered programs, including the development of an HIV/ AIDS vaccine, and product candidates for the treatment of congestive heart failure and Huntingtons disease. | |
| Maximizing the value of our manufacturing and development expertise. It is our strategy to utilize our product development, regulatory and manufacturing capabilities in new product collaboration opportunities, similar to the collaborations we have entered into with Celladon Corporation, or Celladon, and Sirna Therapeutics, Inc., or Sirna. In addition, we may evaluate opportunities to maximize the value we have built in manufacturing viral vectors, including AAV vectors. Our manufacturing expertise may be applied to the production of other biologics, and we may pursue contract manufacturing relationships during periods of excess capacity in our manufacturing facility. | |
| Pursuing additional product opportunities. Although we have made substantial progress toward the commercialization of AAV gene therapy product candidates, gene therapy as a field has not yet reached commercialization in the U.S., Europe, or Japan. We are committed to gene therapy but realize it may be prudent to pursue product development opportunities within the broad classes of |
3
Table of Contents
diseases on which we currently focus. We are evaluating a range of opportunities that include mergers and acquisitions and product diversification with a particular focus on therapeutic indications of inflammation and infectious disease. We believe that a combination of novel candidates developed from our expertise in gene delivery along with complementary candidates may provide a beneficial balance of value and risk diversification for our shareholders. If we identify an appropriate opportunity to add additional value to our current business we intend to pursue the opportunity. |
On January 24, 2006, we restructured our operations to
reduce expenses and realign resources to focus on advancing our
lead product candidate, tgAAC94, through clinical testing.
Over the longer term, our business strategy is to leverage our
technology assets, manufacturing capabilities and gene therapy
product development expertise into multiple development programs
and collaborations to maximize our opportunities to
commercialize a product candidate. We believe that, if
successful, our product candidates will have significant market
potential. We intend to pursue product development programs that
enable us to demonstrate the potential of our technology and
eventually to commercialize gene-based therapeutics that address
currently unmet medical needs.
2005 Achievements |
In 2005, we made progress in our development collaborations and
our product development programs, expanded our patent portfolio,
strengthened our Board of Directors and realigned our cost
structure to focus our resources on our inflammatory arthritis
program. More specifically:
| We commenced work under our December 2004 collaboration agreement with Celladon to develop AAV-based therapies for congestive heart failure. | |
| We expanded our AAV-based gene delivery platform into the area of RNA interference through our collaboration with Sirna Therapeutics to develop a treatment for Huntingtons disease. | |
| In February 2005, we and the International Aids Vaccine Initiative, or IAVI, reported preliminary results of a Phase I HIV/ AIDS vaccine clinical trial that met safety standards and indicated and that the vaccine was well tolerated. | |
| In July 2005, we reported results of an initial Phase I clinical trial of our inflammatory arthritis candidate, demonstrating safety of an intra-articular injection of tgAAC94 into inflamed joints and suggestive of improvements in arthritic signs and symptoms. | |
| In October 2005, we initiated an extended Phase I clinical trial to evaluate tgAAC94 at higher dose levels in patients with inflammatory arthritis and who may be receiving concurrent treatments of anti-TNF-alpha therapy, but who continue to experience inflammation in one or more joints. | |
| In November 2005, we initiated a Phase II HIV/ AIDS vaccine clinical trial in South Africa. | |
| We were named as a subcontractor to receive up to $18 million of a five year, $22 million NIAID contract with Columbus Childrens Research Institute and The Childrens Hospital of Philadelphia to develop AAV-based HIV/ AIDS vaccine for use in the developed world. | |
| We were issued five additional patents from the USPTO covering our AAV vector patent portfolio. | |
| We strengthened our financial position through our agreement with Biogen Idec, or Biogen, to restructure the repayment of approximately $10.7 million of debt. | |
| In January 2006, we restructured operations to reduce expenses and concentrate resources on key product development programs and business development activities. |
4
Table of Contents
PROGRAMS IN CLINICAL TRIALS
Inflammatory Arthritis |
According to the College of Rheumatology and the Centers for
Disease Control and Prevention, two to three million people in
the U.S. are living with inflammatory arthritis, including
rheumatoid arthritis, psoriatic arthritis and ankylosing
spondylitis. Researchers have found that the cytokine called
tumor necrosis factor-alpha, or TNF-alpha, plays a pivotal role
in this disease process and have shown anti-TNF-alpha therapies
to be a valuable strategy to treat inflammatory arthritis.
TNF-alpha inhibition is a validated therapeutic strategy for
treating a variety of inflammatory diseases. Three TNF-alpha
inhibitors are now sold world-wide. These products, which are
delivered systemically by intravenous infusion or sub-cutaneous
injection, can improve the signs and symptoms of the disease,
inhibit the structural damage in the joints, and impact
functional outcomes in patients with these inflammatory
arthritis conditions. However, some patients do not have a
complete response to systemically delivered anti-TNF-alpha
agents and still have significant room for improvement in
inflammation and tender and swollen joint counts. These patients
are potentially ideal candidates for a localized, more
concentrated delivery of anti-TNF-alpha therapy administered
directly to the joint.
We are developing a product candidate, which we call tgAAC94,
for the treatment of inflammatory arthritis. tgAAC94 is an
AAV-based product candidate designed to deliver a DNA sequence
encoding a potent inhibitor of TNF-alpha. We believe that there
may be market receptivity to local delivery of a
TNF-alpha antagonist
for several types of inflammatory arthritis. In addition, we
believe that patients who are partial responders to
anti-TNF-alpha therapy or those who are contraindicated for
systemic therapy may also be strong candidates for a localized
TNF-alpha inhibitor approach. We are designing tgAAC94 for
administration by direct injection into affected joints and
developing it for use in patients who have one or more joints
that have not responded to other therapies, or for patients who
may have only a few inflamed joints and may benefit from
localized rather than systemic treatment of their disease.
In March 2004, we initiated a Phase I clinical trial to
evaluate the safety of escalating doses of tgAAC94 in subjects
with rheumatoid arthritis, psoriatic arthritis or ankylosing
spondylitis. This double-blind, randomized trial evaluated
safety of a single dose of tgAAC94 injected locally into an
arthritic joint of subjects suffering from inflammatory
arthritis. Enrollment was limited to those who were not
currently on concomitant TNF-alpha antagonist therapies. Eleven
of fifteen subjects who enrolled in the trial received one of
two escalating doses of tgAAC94, while the other four received a
placebo. The placebo arm was included at each dose level to
assess safety and to determine whether any adverse events that
might occur were attributable to the intra-articular injection
procedure itself, as opposed to an intra-articular injection of
tgAAC94. The study was not powered to show efficacy. Data from
the trial demonstrated that:
| Intra-articular injections of tgAAC94 were safe and well-tolerated in subjects who were also taking conventional disease-modifying anti-rheumatic drugs. | |
| No tgAAC94 related serious adverse events were reported in the eleven tgAAC94 treated subjects. | |
| In those treated with a single dose of tgAAC94, continued measurable improvements in swelling and tenderness were observed. Six of the eleven tgAAC94 treated subjects received a higher dose and the reduction in mean tenderness and swelling scores appears to be greater than in the five of the eleven tgAAC94 treated subjects who received a lower dose. This suggests there may be a dose response effect. There was some improvement in mean tenderness and swelling scores in subjects receiving placebo, but these subjects were included primarily for safety analysis. | |
| In the non-injected joints of the tgAAC94 treated subjects, there also appears to be a trend in the decrease in mean tenderness and swelling scores over time. This trend was not observed in the subjects receiving placebo. |
We are following these subjects for a period of twenty-four
weeks after treatment, and we expect to present complete data
from this trial in the second quarter of 2006.
In October 2005, we initiated a follow-on Phase I clinical
trial of tgAAC94 administered directly to affected joints of
subjects with inflammatory arthritis. This double-blinded,
placebo-controlled study is
5
Table of Contents
designed to evaluate different doses of tgAAC94 in patients with
rheumatoid arthritis, psoriatic arthritis or ankylosing
spondylitis, who may be receiving concomitant treatments of
anti-TNF-alpha therapy, but are partial responders who continue
to experience signs and symptoms of inflammatory arthritis in
some joints. In the first segment of the double-blinded, placebo
controlled study, subjects will receive a single intra-articular
injection of tgAAC94 or placebo in the affected joint and be
monitored until swelling in the target joint reaches
pre-determined criteria for re-injection. At that time, both
tgAAC94 injected subjects and those initially injected with
placebo will receive a second injection of tgAAC94 in the
affected joint as part of the open-label segment of the study.
The primary endpoint of the study is to establish the safety of
higher doses and of repeat administration of tgAAC94 into the
joints of subjects with and without concomitant TNF-alpha
inhibitor therapy. Secondary endpoints include evaluation of
pain, swelling, duration of response, and overall disease
activity following intra-articular administration of tgAAC94 to
affected joints. Additionally, changes in joint inflammation and
joint damage will be assessed in a subset of subjects using
magnetic resonance imaging.
HIV/ AIDS Vaccine |
There is an urgent need to stop the spread of human
immunodeficiency virus (HIV) infection worldwide. More than
60 million people have been infected and more than
25 million have died due to HIV/ AIDS since the worldwide
epidemic began in 1981. In 2005, over 5 million new
infections occurred. Of these, more than 85% were in
Sub-Saharan Africa and
South East Asia. Further rises of HIV incidence can only be
slowed by a massive expansion of prevention efforts.
Historically, vaccines have been the most powerful public health
tool able to provide a safe, cost-effective and efficient means
of preventing illnesses, disability, and death from infectious
diseases. A safe and effective preventive HIV vaccine, as part
of a comprehensive prevention plan will significantly reduce the
spread of HIV.
We are developing an AAV-based prophylactic vaccine candidate,
which we call tgAAC09, for high-risk populations in developing
nations to protect against HIV/ AIDS. This product candidate is
in Phase I and Phase II clinical trials and is being
developed in a public-private collaboration with IAVI, a
non-profit organization, and the Columbus Childrens
Research Institute at Childrens Hospital in Columbus,
Ohio, or CCRI. The principle investigator for the collaboration
is Dr. Philip R. Johnson of Childrens Hospital of
Philadelphia.
In December 2003, IAVI initiated a Phase I initial dose
escalation safety trial in humans for tgAAC09 in Europe. This
dose-escalation safety trial is designed to enroll up to 50
volunteers who are uninfected with HIV and in good health. Each
participant in this trial received a single injection of the
vaccine candidate or placebo and was monitored for safety and
immune response. Preliminary results from this study were
announced in February 2005 and the data suggests that tgAAC09
was safe and well-tolerated. The safety results also showed that
at the dose levels evaluated in this initial trial, a single
administration of tgAAC09, did not elicit a significant immune
response. These results support further development of tgAAC09
and clinical evaluation at higher dose levels. Additionally, in
a non-human primate study, it was demonstrated that antibody and
T-cell responses to the expressed HIV antigens increased after a
second dose, or boost, of tgAAC09 vaccine. Based on this
preclinical data and upon receiving the necessary regulatory
approvals, we expanded the European Phase I trial to
evaluate the safety and immunogenicity of this vaccine after a
second dose. After volunteers who receive a second dose of
tgAAC09 have been monitored according to protocol, we intend to
report the data from the study in the second half of 2006. The
current Phase I clinical trial of tgAAC09 is the initial
step in a comprehensive development strategy of this vaccine
program. IAVI expanded the single-dose Phase I trial to
include a site in India. The purpose of this study is to further
evaluate the safety of the vaccine in the population that would
participate in subsequent efficacy trials, assuming continued
development of the vaccine candidates.
In 2005, IAVI initiated a Phase II clinical trial of the
tgAAC09 vaccine candidate in South Africa. The trial should take
about 18 months to complete and will enroll 78 volunteers
who are in good health. tgAAC09 is designed to elicit two
different types of immune responses, an antibody response and a
cell mediated response. While these clinical trials are
underway, we continue to pursue the development of additional
vaccine candidates, including vaccines based on different
serotypes, or strains, of AAV believed to be more
6
Table of Contents
efficient delivery systems for gene-based vaccines to muscle.
Preclinical studies of these vaccine approaches have
demonstrated an ability to elicit an immune response at lower
dose levels compared to tgAAC09.
The tgAAC09 vaccine is aimed against HIV Clade C for the
developing world and the program is completely funded by IAVI.
The program is mainly focused in its initial stages upon
comparing the safety and the immune responses induced after
administration of the candidate tgAAC09 which contains the gene
for three HIV Clade C proteins (called gag, pol and rt) in an
AAV serotype 2 capsid and an analogous second candidate vaccine
that contains the same HIV gene in an AAV serotype 1 capsid.
This second candidate is currently undergoing preclinical tests
before advancing into clinical trials in healthy volunteers.
Initial preclinical studies of this AAV serotype vaccine have
demonstrated an ability to elicit an immune response at lower
dose levels.
Under the terms of this public-private collaboration, IAVI funds
us, Childrens Hospital of Philadelphia and CCRI for work
that is focused on development and preclinical studies of a
vaccine candidate. IAVI funds our development activities based
upon an agreed upon annual work plan and budget. IAVI also
coordinates and directly funds the cost of clinical trials
conducted under the collaboration. This collaboration was
initiated in 2000 and in 2004 was extended through the end of
2006. Under the terms of the agreement any of the parties can
terminate this collaboration, without cause, with ninety days
advance notice. If we terminate the agreement for certain
financial conditions including insolvency or nonviability of
commercial manufacture, IAVI retains only those limited rights
to the intellectual property controlled by us that was utilized
during the program and is necessary for IAVI to develop and
commercialize a HIV/ AIDS vaccine in the developing world. Under
certain circumstances, IAVI may owe us an additional royalty for
the use of those rights. If IAVI terminates the agreement for
failure to fund the development activities associated with the
program, we retain all our rights to the intellectual property
that we control.
We have the right to commercialize in developed countries any
vaccine that may result from this development collaboration in
industrialized countries, and we have a qualified right, subject
to IAVIs determination that our prices are reasonable, to
manufacture the vaccine for non-industrialized nations and sell
it to IAVI at full cost of manufacturing plus a reasonable
public sector profit. IAVI has retained rights to ensure that
any safe and efficacious HIV/ AIDS vaccines developed as part of
this collaboration will be distributed in developing countries
at a reasonable price to be determined by IAVI. If we are not
able or decline to produce the vaccine for developing countries
in reasonable quantities and at a reasonable price, IAVI has
rights that will allow IAVI to contract with other manufacturers
to make the vaccines available at a reasonable price in those
countries.
In November 2005, we extended the scope of our HIV/ AIDS vaccine
program via a contract awarded by the National Institute of
Allergy and Infectious Diseases, or NIAID, to CCRI in
collaboration with Childrens Hospital of Philadelphia and
us. NIAID is a component of the National Institutes of Health,
Bethesda, Maryland. Under this program, we may receive up to
$18.2 million over five years for the manufacture and
preclinical testing of the vaccine candidates. Investigators at
Childrens Hospital of Philadelphia and CCRI will design
the vaccine candidates and we will manufacture the vectors for
the clinical testing that will be conducted in the U.S. The
direct costs of any clinical trials will be borne directly by
NIAID and are not part of the contract. This NIAID vaccine
program will complement work performed under the IAVI vaccine
program but will be focused on candidate HIV/ AIDS vaccines for
the developed world. Consequently, the vaccine candidates that
we will be evaluating in this program will contain certain HIV
genes of types that are common in the western world. We will
evaluate administering an initial, or prime, dose of the vaccine
followed some weeks later by a second, or boost, dose. These
prime and boost doses may be comprised of different AAV capsids.
We began work on our portion of the collaboration in the first
quarter of 2006.
PRECLINICAL PROGRAMS
Congestive Heart Failure |
In December 2004, we formed a collaboration with Celladon to
evaluate delivery to the heart, of genes that may have a
therapeutic benefit in the treatment of congestive heart
failure, or CHF. This collaboration
7
Table of Contents
combines our expertise in the development, manufacture and
clinical evaluation of AAV-based therapies with Celladons
portfolio of genes and cardiovascular expertise.
Congestive heart failure is a leading cause of morbidity and
mortality which affects about 5 million Americans. CHF
involves a loss of contractility of the heart muscle which in
turn decreases the ability of the heart to pump blood. The
contraction and expansion of heart muscle is dependent upon
movement of calcium within the heart muscle. Two genes that code
for proteins, called SERCA2a and phospholamban, that are central
to the process of calcium movement in the heart muscle. The
SERCA2a protein is a pump that moves calcium and the
phospholamban protein regulates the activity of SERCA2a. The
relative amount of SERCA2a and phospholamban, and the activity
of SERCA2a, is decreased in failing hearts but it has not been
possible to develop conventional pharmaceutical drugs to address
this problem. However, this problem may be addressed by delivery
of SERCA2a or phospholamban genes. Thus, delivery of the SERCA2a
gene directly to the heart muscle should to lead to more
production of SERCA2a protein and may improve the ability of the
heart to contract and thus improve its ability to pump blood.
Celladon plans to develop a combination product composed of a
therapeutic agent, AAV2/1SERCA2a, and a delivery device which
enables delivery of the therapeutic agent to the heart.
As part of this collaboration, we are contributing our
proprietary AAV technology for use in the field of CHF to
deliver these and other genes of interest. We are also
developing and manufacturing an AAV vector to deliver the
SERCA2a gene, which is currently being studied in preclinical
tests prior to regulatory filings for initiation of clinical
trials in heart failure patients. We will manufacture the
vectors for clinical trials of the CHF product candidate and
assist Celladon in certain regulatory filings. Celladon
anticipates making these regulatory filings in late 2006 to
initiate a Phase I clinical trial in early 2007. In this
program, Celladon is providing its proprietary intellectual
property including the SERCA2a gene or phospholamban variant
genes that are believed to be capable of mediating the
contractility of the heart muscle.
In connection with the formation of this collaboration, we
received $6.0 million cash from the sale of our common
stock to investors of Celladon. During 2005, we incurred
$2.8 million of program related costs to support
development activities under the Celladon collaboration, which
consisted primarily of internal development efforts. We agreed
to contribute up to $2.0 million to support these
development activities and we are reimbursed for efforts over
that amount. During the current year, we were reimbursed
$0.8 million for those labor hours and those outside costs
associated with the Celladon collaboration. In the future, we
are also entitled to development milestones, royalties on sales
and manufacturing profits on potential future products that
result from the collaboration.
Under the terms of the agreement either of the parties can
terminate this collaboration, without cause, with ninety days
advance notice. If we terminate the agreement for certain
financial conditions including insolvency or nonviability of
commercial manufacture, Celladon retains limited rights to the
intellectual property controlled by us that was utilized during
the program and is necessary for Celladon to develop and
commercialize a gene therapy product for the treatment of CHF
associated with the SERCA2a pathway. Under certain
circumstances, they may owe us an additional royalty for the use
of those rights. If Celladon terminates the agreement for
failure to fund the development activities associated with the
program, we retain all our rights to the intellectual property
that we control.
Huntingtons Disease |
In January 2005, we formed a collaboration with Sirna to develop
therapies for the treatment of Huntingtons disease, or HD.
This collaboration also includes two academic groups at the
University of Iowa, or UI, and the University of California,
San Francisco.
HD generally shows onset in mid-life and there are currently
about 30,000 patients in the U.S. and an additional 120,000
to 250,000 at risk of onset. HD is an incurable
neurodegenerative disorder that results from a mutation in the
HD gene which codes for the huntingtin protein. Genes express
their information by being copied into a messenger RNA, or mRNA,
that instructs the cell machinery to make a specific protein.
This mutant HD gene produces a defective huntingtin protein that
interferes with normal function of nerve cells in
8
Table of Contents
the brain leading to eventual development of the progressive
disease. There are no effective drugs to treat or prevent this
disease.
HD is a dominant genetic disease; which means that a single copy
of the mutant gene can cause the disease. It also means that
delivery of a correct copy of the gene will not be effective.
Rather, the function of the mutant HD gene must be blocked. A
potentially effective way to do this is to use recently
discovered entities called small interfering RNA, or siRNA.
siRNAs can bind to mRNAs and cause their degradation before
being used for production of protein. In this way, siRNA can be
used to prevent or reduce the production of proteins.
Our Huntingtons disease program is focused on developing
therapeutic siRNA to target the gene that encodes the HD
protein. This siRNA must be administered directly to the brain
which is the site of the disease. Therefore, infrequent dosing
is highly desirable. Consequently this program uses the AAV
delivery system to deliver a gene for a siRNA that targets the
HD gene and can be expressed for a prolonged period. The program
is based upon initial proof of concept of correction of HD using
this approach in a mouse model of HD that was recently reported
by our collaborators at UI.
The program takes advantage of our expertise and intellectual
property in production and use of the AAV delivery system and
Sirnas expertise in design of siRNA molecules. The program
currently is focused upon generating AAV vectors that express
siRNAs that target the HD gene and testing these in animal
models to select a lead product for formal preclinical testing
and progression to clinical studies. We and Sirna have agreed to
co-develop product candidates under the collaboration and to
share the costs of development and any future revenues that
result from the collaboration. We expect that a substantial
portion of our development costs will consist primarily of
internal development and manufacturing efforts.
Under the terms of the agreement either of the parties can
terminate this collaboration, without cause, with ninety days
advance notice. If we terminate the agreement for certain
financial conditions including insolvency or nonviability of
commercial manufacture, Sirna retains limited rights to the
intellectual property controlled by us that that was utilized
during the program and is necessary for Sirna to develop and
commercialize a siRNA for the treatment of Huntingtons
disease. Under certain circumstances, they may owe us an
additional royalty for the use of those rights. If Sirna
terminates the agreement for failure to fund the development
activities associated with the program, we retain all our rights
to the intellectual property that we control.
Hyperlipidemia |
We are exploring gene therapies for cardiovascular disease by
applying our AAV vector technology to treating hyperlipidemia,
the elevation of lipids, or fats, such as cholesterol in the
bloodstream. As part of our acquisition of Genovo, Inc. in 2000,
we acquired a product development program aimed at assessing the
delivery of genetic material to treat dyslipidemia, a condition
of increased levels of LDL-type cholesterol. We have an ongoing
collaboration with an academic laboratory to assess the
potential clinical utility of AAV vector product candidates for
treating hyperlipidemia. We have exclusive rights to certain
intellectual property related to the use of AAV-based gene
therapy for treating hypercholesterolemia.
FORMER COLLABORATIONS
Cystic Fibrosis Foundation |
Until 2005, we were developing tgAAVCF, a product candidate for
treating cystic fibrosis. In 2003, we established a
collaboration with the Cystic Fibrosis Foundation related to
Phase II clinical trials for tgAAVCF. In March 2005, we
discontinued the development of tgAAVCF and concluded the
collaboration with the CF Foundation following the analysis of
Phase II clinical trial data in which tgAAVCF failed to
achieve the efficacy endpoints of the trial.
9
Table of Contents
Biogen Idec |
In connection with our acquisition of Genovo in 2000, we
established a three-year, multiple-product development and
commercialization collaboration with Biogen. This collaboration
ended in September 2003 upon the completion of the development
period.
Under this collaboration, Biogen paid us $8.0 million in
research funding and upfront payments and $1.0 million per
year in research and development funding over the initial
three-year development period. In connection with the
collaboration, in 2002, we raised $4.0 million through the
sale of 5,804,673 shares of our common stock to Biogen at a
price of $0.69 per share, and in 2003, we raised
$4.8 million through the sale of 2,515,843 shares of
our common stock to Biogen at a price of $1.91 per share.
The equity purchase commitment with Biogen has expired. As a
result of these share purchases and shares Biogen received when
we purchased Genovo, as of December 31, 2005 Biogen held
11.7 million shares of our common stock, approximately
13.7% of our common shares outstanding. In addition, we borrowed
$10.0 million from Biogen under the loan commitment
outlined in the collaboration agreement. In September 2005, we
repaid $2.5 million of the $10.0 million loan
principal and modified the loan payment schedule so that
payments of $2.5 million of principal amount plus accrued
interest are due on each of August 1, 2007, 2008 and 2009.
As part of the debt modification, we extended the maturity date
of our $650,000 promissory note to Biogen until August 1,
2007.
During 2003, we earned $5.1 million of revenue from our
collaboration with Biogen including $2.6 million in revenue
we recognized upon the completion of this development
collaboration in September 2003. From the inception of the
collaboration in 2000 through December 31, 2003, we earned
$11.0 million in revenue from Biogen under this
collaboration and received $18.8 million in proceeds from
the issuance of debt and sales of equity securities.
As part of our collaboration with Biogen, we provided Biogen
with limited manufacturing process development support for its
product development program directed at treating glioma using an
adenoviral vector to deliver the gene for interferon beta.
Interferon beta is a potent stimulator of the immune system, and
sustained expression of this protein at the site of brain tumors
may help the body rid itself of cancer cells. Prior to the
merger of Biogen and IDEC Pharmaceuticals in November 2003,
Biogen had licensed its rights to this program to IDEC as part
of a co-development agreement covering multiple oncology product
development programs. Based on the initial results of clinical
trials conducted by Biogen, the further development of this
glioma product candidate was discontinued by Biogen to pursue
development activities towards targeted indications in malignant
pleural effusions and liver metastases of colorectal cancer. We
are entitled to receive royalties on future product sales by
Biogen of product commercialized based on adenoviral delivery of
interferon beta.
Wyeth |
In 2000, we entered into a collaboration with Wyeth to develop
AAV vector-based gene therapy products for treating hemophilia A
and, potentially, hemophilia B. In November 2002, Wyeth elected
to terminate this hemophilia collaboration and related
agreements. Under the terms of our agreements with Wyeth, all
rights that we granted or otherwise extended to Wyeth related to
the hemophilia technology returned to us. In connection with the
termination of our collaboration with Wyeth, we entered into a
settlement agreement with Wyeth in 2003 and received
$3.2 million in settlement of outstanding expenses that we
incurred under the collaboration and as an early termination
payment.
Emerald Gene Systems, Ltd. |
In 1999, we formed Emerald Gene Systems, Ltd., or Emerald, our
joint venture with Elan International Services, Ltd., a
wholly-owned subsidiary of Elan Corporation plc, or Elan.
Emerald was formed to develop enhanced gene delivery systems.
Emeralds three-year development period ended during 2002
and Emerald has had no operating activities since. From
inception in 1999 through March 2004, we accounted for our
investment in Emerald under the equity method of accounting. In
March 2004, we became the 100% owner of
10
Table of Contents
Emerald and consolidated the results into our financial
statements until Emerald was dissolved in January 2005.
On March 31, 2004, we entered into a termination agreement
with Elan. In accordance with the termination agreement, the
Series B preferred stock was converted into
4.3 million shares of our common stock. As of
December 31, 2005, Elan held 11.6 million shares of
our common stock, approximately 13.6% of our common shares
outstanding. Under the termination agreement Elan is permitted
to trade these shares of our common stock in quantities equal to
175% of the volume limitation set forth in Rule 144(e)(1)
promulgated under the Securities Act of 1933, as amended,
subject to certain exceptions.
LICENSING ARRANGEMENTS
Alkermes, Inc. |
In 1999, we entered into a license agreement with Alkermes,
Inc., or Alkermes, in which we received exclusive rights to
issued patents and other pending patent applications related to
AAV vector technologies including manufacturing. The license
broadly covers a manufacturing method that we believe is
critical to making AAV-based products in a commercially viable,
cost-effective manner. The license to this technology, developed
by Childrens Hospital in Columbus, Ohio, covers the use of
cell lines for manufacturing AAV vectors in multiple disease
areas. Under the terms of the license agreement, we issued to
Alkermes 500,000 shares of our common stock and warrants to
purchase 2,000,000 shares of our common stock, which
warrants expire in June 2007 and June 2009. Alkermes will also
receive milestone payments and royalties on the sale of any
products manufactured using the licensed technology and is
entitled to a portion of any sub-licensing payments that we may
receive.
Amgen, Inc. |
Targeted Genetics was formed in 1989 as a subsidiary of Immunex
Corporation, or Immunex, a biopharmaceutical company developing
recombinant proteins as therapeutics. In connection with our
formation and the entering into of Gene Transfer Technology
License Agreement, we issued Immunex shares of our preferred
stock that were subsequently converted into
1,920,000 shares of our common stock. In exchange, we
received rights from Immunex under a Gene Transfer Technology
License Agreement, including an exclusive worldwide license to
certain Immunex proprietary technology specifically applicable
to gene therapy applications. The licensed technology relates to
gene identification and cloning, panels of retroviral vectors,
packaging cell technology, recombinant cytokines, DNA
constructs, cell lines, promoter/enhancer elements and
immunological assays. In July 2002, Immunex was acquired by
Amgen, Inc, or Amgen. Our license to the Immunex technology was
not affected by the acquisition and we retain all rights granted
under the original license.
Prior to Amgens acquisition of Immunex, we exchanged
correspondence and engaged in discussions with Immunex regarding
the terms, scope and possible amendment of the Gene Transfer
Technology License Agreement. Some of these communications have
included, among other things, differing views about our rights
to the gene construct coding for TNFR:Fc used in the development
of our inflammatory arthritis product candidate tgAAC94. These
communications did not lead to either a final resolution or an
active dispute regarding our differences with Immunex. Following
Amgens acquisition of Immunex, we communicated to Amgen
our desire to resume discussions seeking clarification of our
relationship with Amgen. Our subsequent communications with
Amgen have not yet resulted in a resolution of our differences.
In February 2004, in response to our January 2004 announcement
that we had received regulatory approval for a Phase I
clinical study for tgAAC94, Amgen sent a letter to us taking the
position that we were not licensed, either exclusively or
non-exclusively, under Immunex intellectual property covering
TNFR:Fc or therapeutic uses for TNFR:Fc. We have responded with
a letter confirming our confidence that the Gene Transfer
Technology License Agreement gives us an exclusive worldwide
license to use the gene construct coding for TNFR:Fc for gene
therapy applications. We have had, and expect to have further,
communications with Amgen regarding our differences.
Notwithstanding our confidence, it is possible that a resolution
of those differences, through
11
Table of Contents
litigation or otherwise, could cause delay or discontinuation of
our development of tgAAC94 or our inability to commercialize any
resulting product.
PATENTS AND PROPRIETARY RIGHTS
Patents and licenses are important to our business. Our strategy
is to file or license patent applications to protect technology,
inventions and improvements to inventions that we consider
important to developing our business. We have filed or licensed
numerous patent or patent applications with the USPTO and
foreign jurisdictions. This proprietary intellectual property
includes methods of transferring genetic material into cells,
processes to manufacture and purify gene delivery product
candidates and other proprietary technologies and processes. We
also rely on unpatented proprietary technology such as trade
secrets, know-how and continuing technological innovations to
develop and maintain our competitive position.
The patent positions of pharmaceutical and biotechnology firms,
including our patent positions, are uncertain and involve
complex legal and factual questions for which important legal
principles are largely unresolved, particularly with regard to
gene therapy uses. Patent applications may not result in the
issuance of patents, and the coverage claimed in a patent
application may be significantly reduced before a patent is
issued.
We have licensed technology underlying several issued and
pending patents. Among these are two key patents that relate to
the use of AAV vectors for gene delivery, one of which we have
exclusively licensed from the U.S. National Institutes of
Health, or NIH, and the second from the University of Florida
Research Foundation. Licensing of intellectual property critical
to our business involves complex legal, business and scientific
issues. If disputes over intellectual property that we have
licensed prevent or impair our ability to maintain our current
licensing arrangements on acceptable terms, we may be unable to
successfully develop or commercialize the affected product
candidates.
In addition to patent protection, we rely on trade secret
protection for our confidential and proprietary information and
technology. To protect our trade secrets, we generally require
our employees, consultants, scientific advisors and parties to
collaborative agreements to execute confidentiality agreements.
In the case of employees and consultants, the agreements also
provide that all inventions resulting from work performed by
them while employed by us will be our exclusive property.
Despite these agreements, and other precautions we take to
protect our trade secrets and other proprietary unpatented
intellectual property, we may be unable to meaningfully protect
our trade secrets and other intellectual property from
unauthorized use or misappropriation by a third party. These
agreements may not provide adequate remedies in the event of
unauthorized use or disclosure of our confidential information.
In addition, our competitors could obtain rights to our
nonexclusively licensed proprietary technology or may
independently develop substantially equivalent proprietary
information and technology. If our competitors develop and
market competing products using our unpatented or nonexclusively
licensed intellectual property or substantially similar
technology or processes, our products could suffer a reduction
in sales or be forced out of the market.
A number of pharmaceutical and biotechnology companies and
research and academic institutions have developed technologies,
filed patent applications or received patents for technologies
that may be related to our business. Some of these technologies,
applications or patents may conflict with our technologies or
patent applications. This conflict could limit the scope of any
patents that we may obtain for our technologies or result in
denial of our patent applications. In addition, if patents or
patent applications that cover our activities are or have been
issued to other companies, we may be required to either obtain a
license from the owner or develop or obtain alternative
technology. A license may not be available on acceptable terms,
if at all, and we may be unable to develop or obtain alternative
technology.
As the biotechnology industry expands and more patents are
issued, the risk increases that our processes and potential
products may give rise to claims that they infringe on the
patents of others. These other parties could bring legal actions
against us claiming damages and seeking to stop clinical
testing, manufacturing and marketing of the affected product or
use of the affected process. If we are found by a court to have
infringed on the proprietary rights of others, we could also
face potential liability for significant damages and be required
to obtain a license to the proprietary technology at issue if we
continue to commercialize. A required license may
12
Table of Contents
not be available on acceptable terms, if at all, which could
impair our ability to commercialize our product candidates.
Similarly, administrative proceedings, litigation or both may be
necessary to enforce patents issued to us, to protect trade
secrets or know-how owned by us or to determine the
enforceability, scope and validity of the proprietary rights of
others. This type of litigation, regardless of its merit, could
result in substantial expense to us and significantly divert the
efforts of our technical and management personnel. An adverse
outcome could adversely affect our business.
COMPETITION
Competition among biotechnology and pharmaceutical companies
that research, develop, manufacture and commercialize
therapeutic products is significant. Even in the field of gene
therapy, numerous companies and institutions are developing or
considering the development of gene therapy treatments,
including other gene delivery companies, biotechnology
companies, pharmaceutical companies, universities, research
institutions, governmental agencies and other healthcare
providers.
In addition to competition from sources developing competitive
gene therapy technologies, our potential products will compete
with non-gene therapy products in development and on the market
for the therapeutic indications we are targeting. These
competitors could include small molecules, proteins, monoclonal
antibodies and other pharmaceutical products, medical devices
and surgery. Products in development could make our products
obsolete before they ever get to the market. Products on the
market could negatively impact the commercial opportunity for
our products. Intense competition could heighten disputes
pursued in an effort to slow our development including lawsuits,
demands, threats or patent challenges. We also compete with
others to acquire products or technology from research
institutions, universities and other companies. And we compete
with others to maintain and attract the necessary scientific and
business personnel to advance our programs.
Many of our competitors have substantially more financial and
other resources, larger research and development staffs and more
experience and capabilities in researching, developing and
testing products in clinical trials, obtaining U.S. Food
and Drug Administration, or FDA, and other regulatory approvals
and manufacturing, marketing and distributing products. In
addition, the competitive positions of other companies may be
strengthened through collaborative relationships, such as those
with large pharmaceutical companies or academic institutions. As
a result, our competitors may develop, obtain patent protection
for, receive FDA and other regulatory approvals for or
commercialize products more rapidly than we do or may
manufacture and market their products more successfully than we
do.
Our competitors technologies and products may be more
effective or economically feasible than our potential products.
If we are successful in commercializing our products, we will be
required to compete with respect to commercial manufacturing
efficiency and marketing capabilities, areas in which we have no
experience. These developments could limit the prices we are
able to charge for any products we are able to commercialize or
render our products less competitive or obsolete.
Our lead product candidate, tgAAC94, is aimed at the target
market of inflammatory arthritis. A number of products are
currently successfully marketed to treat people with
inflammatory arthritis, including products which work by the
same mechanism of action, TNF-alpha inhibition. TNF-alpha
inhibitor products currently on the market include products from
Amgen, Johnson and Johnson, Abbott and Centocor. Although
tgAAC94 is targeted to people not completely responding to these
systemic TNF inhibitor drugs, other companies are also
developing products to complement the systemic TNF inhibitors.
Products in development or on the market for inflammatory
arthritis could negatively impact the development path and
market opportunity for tgAAC94.
GOVERNMENTAL REGULATION
All of our potential products must receive regulatory approval
before they can be marketed. Human therapeutic products are
subject to rigorous preclinical and clinical testing and other
pre-market approval procedures administered by the FDA and
similar authorities in foreign countries. In accordance with the
Federal Food, Drug and Cosmetics Act, the FDA exercises
regulatory authority over the development, testing,
13
Table of Contents
formulation, manufacture, labeling, storage, record keeping,
reporting, quality control, advertising, promotion, export and
sale of our potential products. Similar requirements are imposed
by foreign regulatory agencies. In some cases, state regulations
may also apply.
Gene therapy is a relatively new technology that has not been
extensively tested or shown to be effective in humans. The FDA
reviews all product candidates for safety at each stage of
clinical testing. Safety standards must be met before the FDA
permits clinical testing to proceed to the next stage. Also,
efficacy must be demonstrated before the FDA grants product
approval. Obtaining approval from the FDA and other regulatory
authorities for a new therapeutic product candidate, if approval
is ever obtained, is likely to take several years. We may
encounter difficulties or unanticipated costs in our efforts to
secure necessary governmental approvals, which could delay or
prevent the marketing of our product candidates. In addition,
the regulatory requirements governing gene therapy product
candidates and commercialized products are subject to change.
The approval process, and ongoing compliance with applicable
regulations after approval, involves substantial expenditures of
financial and other resources.
Preclinical studies generally require studies in the laboratory
or in animals to assess the potential products safety and
effectiveness. Preclinical studies include laboratory evaluation
of toxicity, pharmacokinetics, how the body processes and reacts
to the drug, and pharmacodynamics, whether the drug is actually
having the expected effect on the body. Preclinical studies must
be conducted in accordance with the FDAs Good Laboratory
Practice regulations and, before any proposed clinical testing
in humans can begin, the FDA must review the results of these
preclinical studies as part of an Investigational New Drug
application.
If preclinical studies of a product candidate, including animal
studies, demonstrate safety, and laboratory test results are
acceptable, then the potential product will undergo clinical
trials to test the therapeutic agent in humans. Human clinical
trials are subject to numerous governmental regulations that
provide detailed procedural and administrative requirements
designed to protect the trial participants. Each institution
that conducts human clinical trials has an Institutional Review
Board or Ethics Committee charged with evaluating each trial and
any trial amendments to ensure that the trial is ethical,
subjects are protected and the trial meets the institutional
requirements. These evaluations include reviews of how the
institution will communicate the risks inherent in the clinical
trial to potential participants, so that the subjects may give
their informed consent. Clinical trials must be conducted in
accordance with the FDAs Good Clinical Practices
regulations and the protocols the company establishes to govern
the trial objectives, the parameters to be used for monitoring
safety, the criteria for evaluating the efficacy of the
potential product and the rights of each trial participant with
respect to safety. FDA regulations require us to submit these
protocols as part of the application. A FDA review or approval
of the protocols, however, does not necessarily mean that the
trial will successfully demonstrate safety and/or efficacy of
the potential product.
Institutions that receive NIH funding for gene therapy clinical
trials must also comply with the NIH Recombinant DNA Guidelines,
and the clinical trials are subject to a review by the
NIHs Office of Biotechnology Activities Recombinant DNA
Advisory Committee, or RAC. The outcome of this review can be
either an approval to initiate the trial without a public review
or a requirement that the proposed trial be reviewed at a
quarterly committee meeting. A clinical trial will be publicly
reviewed when at least three of the committee members or the
Director of the Office of Biotechnology Activities recommends a
public review. Should the RAC require a public hearing, the
start of the trial must be delayed until after the hearing date.
Although the NIH guidelines do not have regulatory status, the
RAC review process can impede the initiation of the trial, even
if the FDA has reviewed the trial and approved its initiation.
Additionally, before any clinical trial can be initiated at an
NIH-funded site, the Institutional Biosafety Committee of that
site must perform a review of the proposed clinical trial and
ensure there are no safety issues associated with the trial.
Clinical trials are typically conducted in three phases often
involving multiple clinical trials in each phase. In
Phase I, clinical trials generally involve a small number
of subjects, who may or may not be afflicted with the target
disease, to determine the preliminary safety profile. In
Phase II, clinical trials are conducted with larger groups
of subjects afflicted with the target disease in order to
establish preliminary effectiveness and optimal dosages and to
obtain additional evidence of safety. In Phase III,
large-scale, multi-center, comparative clinical trials are
conducted with subjects afflicted with the target disease in
order to provide
14
Table of Contents
enough data for the statistical proof of efficacy and safety
required by the FDA and other regulatory agencies for market
approval. We report our progress in each phase of clinical
testing to the FDA, which may require modification, suspension
or termination of the clinical trial if it deems patient risk
too high. The length of the clinical trial period, the number of
trials conducted and the number of enrolled subjects per trial
vary, depending on our results and FDA requirements for the
particular clinical trial. Although we and other companies in
our industry have made progress in the field of gene therapy, we
cannot predict what the FDA will require in any of these areas
to establish to its satisfaction the safety and effectiveness of
the product candidate.
If we successfully complete clinical trials for a product
candidate, we must obtain FDA approval or similar approval
required by foreign regulatory agencies, as well as the approval
of several other governmental and nongovernmental agencies,
before we can market the product in the U.S. or in foreign
countries. Current FDA regulations relating to biologic
therapeutics require us to submit an acceptable Biologics
License Application, or BLA, to the FDA and receive approval
before the FDA will permit commercial marketing. The BLA
includes a description of our product development activities,
the results of preclinical studies and clinical trials and
detailed manufacturing information. Unless the FDA gives
expedited review status, this stage of the review process
generally takes at least one year. Should the FDA have concerns
with respect to the potential products safety and
efficacy, it may request additional data, which could delay
product review or approval. The FDA may ultimately decide that
the BLA does not satisfy its criteria for approval and might
require us to do any or all of the following:
| modify the scope of our desired product claims; | |
| add warnings or other safety-related information; and/or | |
| perform additional testing. |
Because the FDA has not yet approved any gene therapy products,
it is not clear what, if any, unforeseen issues may arise during
the approval process. While we expect this regulatory structure
to continue, we also expect the FDAs regulatory approach
to product approval, and its requirements with respect to
product testing, to become more predictable as its scientific
knowledge and experience in the field of gene therapy increases.
Adverse events in the field of gene therapy or other
biotechnology-related fields, however, could result in greater
governmental regulation, stricter labeling requirements and
potential regulatory delays in the testing or approval of gene
therapy products.
Once approved by the FDA, marketed products are subject to
continual FDA review. Later discovery of previously unknown
problems or failure to comply with applicable regulatory
requirements may result in restrictions on marketing a product
or in its withdrawal from the market, as well as potential
criminal penalties or sanctions. In addition, the FDA requires
that manufacturers of a product comply with current Good
Manufacturing Practices requirements, both as a condition to
product approval and on a continuing basis. In complying with
these requirements, we expend significant amounts of time, money
and effort in production, record keeping and quality control.
Our manufacturing facilities are subject to periodic inspections
by the FDA. If major problems are identified during these
inspections that could impact patient safety, the FDA could
subject us to possible action, such as the suspension of product
manufacturing, product seizure, withdrawal of approval or other
regulatory sanctions. The FDA could also require us to recall a
product.
We are also subject to regulation under the Occupational Safety
and Health Act, the Environmental Protection Act, the Toxic
Substances Control Act, the Resource Conservation and Recovery
Act and other federal, state and local regulations. For example,
our controlled use of hazardous materials in our research and
development activities must comply with standards prescribed by
state and federal law.
EMPLOYEES
As of December 31, 2005, we had
95 full-time-equivalent employees. On January 24,
2006, we eliminated 26 of these positions, which left us with
69 full-time-equivalent employees, of which 47 are directly
involved in our research and development activities, including
product development, manufacturing, quality control, quality
assurance, process development, regulatory affairs and clinical
affairs. Nine of our employees
15
Table of Contents
have Ph.D. or M.D. degrees and a significant number of our
management and professional employees have prior experience with
other biotechnology or pharmaceutical companies. We also rely on
a number of temporary staff positions and third party
consultants. None of our employees are covered by a collective
bargaining agreement.
AVAILABLE INFORMATION
We were incorporated in the state of Washington in 1989. Our
executive offices are located at 1100 Olive Way, Suite 100,
Seattle, Washington 98101, and our telephone number is
(206) 623-7612. We file annual, quarterly and current
reports, proxy statements and other information with the SEC. We
make available in the investor relations portion of our website,
free of charge, copies of our annual report on
Form 10-K,
quarterly reports on
Form 10-Q, current
reports on
Form 8-K and
amendments to these reports after filing these reports to the
SEC. Our website is located at www.targetedgenetics.com. You may
also obtain free copies of our periodic reports on the SEC Web
site at http://www.sec.gov.
Item 1A. | Risk Factors |
In addition to the other information contained in this annual
report, you should carefully read and consider the following
risk factors. If any of these risks actually occur, our
business, operating results or financial condition could be
harmed. This could cause the trading price of our stock to
decline, and you could lose all or part of your investment.
Risks Related to Our Business
If we are unable to raise additional capital when needed, we will be unable to conduct our operations and develop our potential products. |
Because internally generated cash flow will not fund development
and commercialization of our product candidates, we will require
substantial additional financial resources. Our future capital
requirements will depend on many factors, including:
| the rate and extent of scientific progress in our research and development programs; | |
| the timing, costs and scope of, and our success in, conducting clinical trials, obtaining regulatory approvals and pursuing patent prosecutions; | |
| competing technological and market developments; | |
| the ability to re-negotiate any lease obligations; | |
| the timing and costs of, and our success in any product commercialization activities and facility expansions, if and as required; and | |
| the expense and outcome of any litigation or administrative proceedings involving our intellectual property, or access to third party intellectual property through licensing agreements. |
We expect that our cash and cash equivalents at
December 31, 2005, plus the funding from our partners and
the proceeds from the sale of common stock in March 2006 will be
sufficient to fund our operations into the first quarter of
2007. This estimate is based on our ability to perform planned
research and development activities and the receipt of planned
funding from our collaborators. In addition, we owe
approximately $8.2 million in aggregate principal amount
under two notes payable to Biogen Idec. The notes terms
require us to make scheduled principal payments of
$3.2 million in August 2007 and $2.5 million in each
of August 2008 and 2009. We will need to raise additional
capital to make the scheduled payments and to repay these notes.
Additional sources of financing could involve one or more of the
following:
| entering into additional product development and funding collaborations or other strategic transactions, or extending or expanding our current collaborations; | |
| issuing equity in the public or private markets; or | |
| issuing debt. |
16
Table of Contents
Additional funding may not be available to us on reasonable
terms, if at all. Our ability to issue equity, and our ability
to issue it at the current market price, may be adversely
affected by the fact that we are presently ineligible under SEC
rules to utilize
Form S-3 for
primary offerings of our securities because the aggregate market
value our outstanding common stock held by non-affiliates is
less than $75 million.
The perceived risk associated with the possible sale of a large
number of shares could cause some of our stockholders to sell
their stock, thus causing the price of our stock to decline. In
addition, actual or anticipated downward pressure on our stock
price due to actual or anticipated sales of stock could cause
some institutions or individuals to engage in short sales of our
common stock, which may itself cause the price of our stock to
decline.
If our stock price declines, we may be unable to raise
additional capital. A sustained inability to raise capital could
force us to go out of business. Significant declines in the
price of our common stock could also impair our ability to
attract and retain qualified employees, reduce the liquidity of
our common stock and result in the delisting of our common stock
from the NASDAQ Capital Market.
The funding that we expect to receive from our collaborations
depends on continued scientific progress under the collaboration
and our collaborators ability and willingness to continue
or extend the collaboration. If we are unable to successfully
access additional capital, we may need to scale back, delay or
terminate one or more of our development programs, curtail
capital expenditures or reduce other operating activities. We
may also be required to relinquish some rights to our technology
or product candidates or grant or take licenses on unfavorable
terms, either of which would reduce the ultimate value to us of
our technology or product candidates.
We expect to continue to operate at a loss and may never become profitable. |
Substantially all of our revenue has been derived under
collaborative research and development agreements relating to
the development of our potential product candidates. We have
incurred, and will continue to incur for the foreseeable future,
significant expense to develop our research and development
programs, conduct preclinical studies and clinical trials, seek
regulatory approval for our product candidates and provide
general and administrative support for these activities. As a
result, we have incurred significant net losses since inception,
and we expect to continue to incur substantial additional losses
in the future. As of December 31, 2005, we had an
accumulated deficit of $250.0 million. We may never
generate profits and, if we do become profitable, we may be
unable to sustain or increase profitability.
All of our product candidates are in early-stage clinical trials or preclinical development, and if we are unable to successfully develop and commercialize our product candidates we will be unable to generate sufficient capital to maintain our business. |
In November 2005, IAVI initiated a Phase II trial for our
HIV/ AIDS vaccine product candidate in South Africa. In October
2005, we initiated a second Phase I trial for our
inflammatory arthritis product candidate in the United States
and Canada. We will not generate any product revenue for at
least several years and then only if we can successfully develop
and commercialize our product candidates. Commercializing our
potential products depends on successful completion of
additional research and development and testing, in both
preclinical development and clinical trials. Clinical trials may
take several years or more to complete. The commencement, cost
and rate of completion of our clinical trials may vary or be
delayed for many reasons, including the risks discussed
elsewhere in this section. If we are unable to successfully
complete preclinical and clinical development of some or all of
our product candidates in a timely manner, we may be unable to
generate sufficient product revenue to maintain our business.
Even if our potential products succeed in clinical trials and
are approved for marketing, these products may never achieve
market acceptance. If we are unsuccessful in commercializing our
product candidates for any reason, including greater
effectiveness or economic feasibility of competing products or
treatments, the failure of the medical community or the public
to accept or use any products based on gene delivery, inadequate
marketing and distribution capabilities or other reasons
discussed elsewhere in this section, we will be unable to
generate sufficient product revenue to maintain our business.
17
Table of Contents
Failure to recruit subjects could delay or prevent clinical trials of our potential products, which could delay or prevent the development of potential products. |
Identifying and qualifying subjects to participate in clinical
trials of our potential products is critically important to our
success. The timing of our clinical trials depends on the speed
at which we can recruit subjects to participate in testing our
product candidates. We have experienced delays in some of our
clinical trials, and we may experience similar delays in the
future. If subjects are unwilling to participate in our gene
therapy trials because of negative publicity from adverse events
in the biotechnology or gene therapy industries or for other
reasons, including competitive clinical trials for similar
patient populations, the timeline for recruiting subjects,
conducting trials and obtaining regulatory approval of potential
products will be delayed. These delays could result in increased
costs, delays in advancing our product development, delays in
testing the effectiveness of our technology or termination of
the clinical trials altogether.
The regulatory approval process for our product candidates is costly, time-consuming and subject to unpredictable changes and delays, and our product candidates may never receive regulatory approval. |
No gene therapy products have received regulatory approval for
marketing from the U.S. Food and Drug Administration, or
FDA. Because our product candidates involve new and unproven
technologies, we believe that the regulatory approval process
may proceed more slowly compared to clinical trials involving
traditional drugs. The FDA and applicable state and foreign
regulators must conclude at each stage of clinical testing that
our clinical data suggest acceptable levels of safety in order
for us to proceed to the next stage of clinical trials. In
addition, gene therapy clinical trials conducted at institutions
that receive funding for recombinant DNA research from the NIH,
are subject to review by the NIHs Office of Biotechnology
Activities RAC. Although NIH guidelines do not have regulatory
status, the RAC review process can impede the initiation of the
trial, even if the FDA has reviewed the trial and approved its
initiation. Moreover, before a clinical trial can begin at an
NIH-funded institution, that institutions Institutional
Biosafety Committee must review the proposed clinical trial to
assess the safety of the trial.
The regulatory process for our product candidates is costly,
time-consuming and subject to unpredictable delays. The clinical
trial requirements of the FDA, NIH and other agencies and the
criteria these regulators use to determine the safety and
efficacy of a product candidate vary substantially according to
the type, complexity, novelty and intended use of the potential
products. In addition, regulatory requirements governing gene
therapy products have changed frequently and may change in the
future. Accordingly, we cannot predict how long it will take or
how much it will cost to obtain regulatory approvals for
clinical trials or for manufacturing or marketing our potential
products. Some or all of our product candidates may never
receive regulatory approval. A product candidate that appears
promising at an early stage of research or development may not
result in a commercially successful product. Our clinical trials
may fail to demonstrate the safety and efficacy of a product
candidate or a product candidate may generate unacceptable side
affects or other problems during or after clinical trials.
Should this occur, we may have to delay or discontinue
development of the product candidate, and the partner, if any,
that supports development of that product candidate may
terminate its support. Delay or failure to obtain, or unexpected
costs in obtaining, the regulatory approval necessary to bring a
potential product to market could decrease our ability to
generate sufficient product revenue to maintain our business.
If we are unable to obtain or maintain licenses for necessary third-party technology on acceptable terms or to develop alternative technology, we may be unable to develop and commercialize our product candidates. |
We have entered into exclusive and nonexclusive license
agreements that give us and our partners rights to use
technologies owned or licensed by commercial and academic
organizations in the research, development and commercialization
of our potential products. For example, we have a gene therapy
technology license agreement with Amgen Inc., or Amgen, as the
successor to Immunex Corporation, or Immunex, under which we
have license rights to certain Immunex proprietary technology
specifically applicable to gene therapy applications. In a
February 2004 letter, Amgen took the position that we are not
licensed, either exclusively or nonexclusively, to use Immunex
intellectual property covering TNFR:Fc or therapeutic uses for
18
Table of Contents
TNFR:Fc. We have responded with a letter confirming our
confidence that the gene therapy technology license agreement
provides us with an exclusive worldwide license to use the gene
construct coding for TNFR:Fc for gene therapy applications. We
have had and continue to have further communications with Amgen
regarding our differences. Notwithstanding our confidence, it is
possible that a resolution of those differences, through
litigation or otherwise, could cause delay or discontinuation of
our development of tgAAC94 or our inability to commercialize any
resulting product.
We believe that we will need to obtain additional licenses to
use patents and unpatented technology owned or licensed by
others for use, compositions, methods, processes to manufacture
compositions, processes to manufacture and purify gene delivery
product candidates and other technologies and processes for our
present and potential product candidates. If we are unable to
maintain our current licenses for third-party technology or
obtain additional licenses on acceptable terms, we may be
required to expend significant time and resources to develop or
license replacement technology. If we are unable to do so, we
may be unable to develop or commercialize the affected product
candidates. In addition, the license agreements for technology
for which we hold exclusive licenses typically contain
provisions that require us to meet minimum development
milestones in order to maintain the license on an exclusive
basis for some or all fields of the license. We also have
license agreements for some of our technologies, which may
require us to sublicense certain of our rights. If we do not
meet these requirements, our licensor may convert all or a
portion of the license to a nonexclusive license or, in some
cases, terminate the license.
In many cases, patent prosecution of our licensed technology is
controlled solely by the licensor. If our licensors fail to
obtain and maintain patent or other protection for the
proprietary intellectual property we license from them, we could
lose our rights to the intellectual property or our exclusivity
with respect to those rights, and our competitors could market
competing products using the intellectual property. Licensing of
intellectual property is of critical importance to our business
and involves complex legal, business and scientific issues and
is complicated by the rapid pace of scientific discovery in our
industry. Disputes may arise regarding intellectual property
subject to a licensing agreement, including:
| the scope of rights granted under the license agreement and other interpretation-related issues; | |
| the extent to which our technology and processes infringe on intellectual property of the licensor that is not subject to the licensing agreement; | |
| the sublicensing of patent and other rights under our collaborative development relationships; | |
| the ownership of inventions and know-how resulting from the joint creation or use of intellectual property by our licensors and us and our partners; and | |
| the priority of invention of patented technology. |
If disputes over intellectual property that we have licensed
prevent or impair our ability to maintain our current licensing
arrangements on acceptable terms, we may be unable to
successfully develop and commercialize the affected product
candidates.
Litigation involving intellectual property, product liability or other claims and product recalls could strain our resources, subject us to significant liability, damage our reputation or result in the invalidation of our proprietary rights. |
As our product development efforts progress, most particularly
in potentially significant markets such as HIV/ AIDS, congestive
heart failure or inflammatory arthritis therapies, the risk
increases that others may claim that our processes and product
candidates infringe on their intellectual property rights. In
addition, administrative proceedings, litigation or both may be
necessary to enforce our intellectual property rights or
determine the rights of others. Defending or pursuing these
claims, regardless of their merit, would be costly and would
likely divert managements attention and resources away
from our operations. If there were to be an adverse outcome in
litigation or an interference proceeding, we could face
potential liability for significant damages or be required to
obtain a license to the patented process or technology at issue,
or both. If we are unable to obtain a license on acceptable
terms, or to develop or obtain alternative technology or
processes, we
19
Table of Contents
may be unable to manufacture or market any product or potential
product that uses the affected process or technology.
Clinical trials and the marketing of any potential products may
expose us to liability claims resulting from the testing or use
of our products. Gene therapy treatments are new and unproven,
and potential known and unknown side effects of gene therapy may
be serious and potentially life-threatening. Product liability
claims may be made by clinical trial participants, consumers,
healthcare providers or other sellers or users of our products.
Although we currently maintain liability insurance, the costs of
product liability and other claims against us may exceed our
insurance coverage. In addition, we may require increased
liability coverage as additional product candidates are used in
clinical trials or commercialized. Liability insurance is
expensive and may not continue to be available on acceptable
terms. A product liability or other claim or product recall not
covered by or exceeding our insurance coverage could
significantly harm our financial condition. In addition, adverse
publicity resulting from a product recall or a liability claim
against us, one of our partners or another gene therapy company
could significantly harm our reputation and make it more
difficult to obtain the funding and collaborative partnerships
necessary to maintain our business.
If we lose our collaborative partners, we may be unable to develop our potential products. |
A portion of our operating expenses are funded through our
collaborative agreements with third parties. We currently have
strategic partnerships with two biotechnology companies, Sirna
Therapeutics and Celladon, one public health organization, IAVI,
and through a contract with a U.S. government agency,
NIAID, that provide for funding, collaborative development,
intellectual property rights or expertise to develop certain of
our product candidates. With limited exceptions, each
collaborator has the right to terminate its obligation to
provide research funding at any time for scientific or business
reasons. In addition, to the extent that funding is provided by
a collaborator for non-program-specific uses, the loss of
significant amounts of collaborative funding could result in the
delay, reduction or termination of additional research and
development programs, a reduction in capital expenditures or
business development and other operating activities, or any
combination of these measures. For example, we have a
collaborative development agreement with IAVI, which expires in
December 2006, that we expect to provide us with funding to
reimburse research and development and manufacturing expenses we
incur in connection with the collaboration. As a result, a
significant portion of our operating expenses are funded through
our collaborative agreements with IAVI. Additionally, IAVI
directly funds the Phase II clinical trial for our HIV/
AIDS vaccine product candidate.
If we do not attract and retain qualified personnel, we may be unable to develop and commercialize some of our potential products. |
Our future success depends in large part on our ability to
attract and retain key technical and management personnel. All
of our employees, including our executive officers, can
terminate their employment with us at any time. We have programs
in place designed to retain personnel, including competitive
compensation packages and programs to create a positive work
environment. Other companies, research and academic institutions
and other organizations in our field compete intensely for
employees, however, and we may be unable to retain our existing
personnel or attract additional qualified employees and
consultants. If we experience significant turnover or difficulty
in recruiting new personnel, our research and development of
product candidates could be delayed and we could experience
difficulty in generating sufficient revenue to maintain our
business.
The reduction in workforce associated with our recent restructuring of operations may impair our ability to develop our product candidates and harm our operations. |
January 24, 2006 we restructured our operations to
concentrate our resources on generating data from the clinical
trials of our inflammatory arthritis product candidate,
maintaining our manufacturing capabilities and advancing our
funded product development efforts. As part of this
restructuring we eliminated 26 positions, which reduced our
workforce by approximately 27%, including scientific, operations
and administrative functions that were not required to support
our programs. This restructuring primarily affected
earlier-stage product discovery and preclinical product
development efforts, small-scale vector manufacturing employees,
20
Table of Contents
management positions and operations and general and
administrative support. Many of the terminated employees possess
specific knowledge or expertise that may later prove to be
important to our operations. As a result of these factors, our
ability to develop our product candidates and respond to
challenges in the future may be impaired and we may not be able
to take advantage of new business opportunities.
If our partners or scientific consultants terminate, reduce or delay our relationships with them, we may be unable to develop our potential products. |
Our partners provide funding, manage regulatory filings, aid and
augment our internal research and development efforts and
provide access to important intellectual property and know-how.
Their activities include, for example, support in processing the
regulatory filings of our product candidates and funding
clinical trials. Our outside scientific consultants and
contractors perform research, develop technology and processes
to advance and augment our internal efforts and provide access
to important intellectual property and know-how. Their
activities include, for example, clinical evaluation of our
product candidates, product development activities performed
under our research collaborations, research under sponsored
research agreements and contract manufacturing services.
Collaborations with established pharmaceutical and biotechnology
companies and academic, research and public health organizations
often provide a measure of validation of our product development
efforts in the eyes of securities analysts, investors and the
medical community. The development of certain of our potential
products, and therefore the success of our business, depends on
the performance of our partners, consultants and contractors. If
they do not dedicate sufficient time, regulatory or other
technical resources to the research and development programs for
our product candidates or if they do not perform their
obligations as expected, we may experience delays in, and may be
unable to continue, the preclinical or clinical development of
those product candidates. Each of our collaborations and
scientific consulting relationships concludes at the end of the
term specified in the applicable agreement unless we and our
partners agree to extend the relationship. Any of our partners
may decline to extend the collaboration, or may be willing to
extend the collaboration only with a significantly reduced
scope, for a number of scientific or business reasons.
Competition for scientific consultants and partners in gene
therapy is intense. We may be unable to successfully maintain
our existing relationships or establish additional relationships
necessary for the development of our product candidates on
acceptable terms, if at all. If we are unable to do so, our
research and development programs may be delayed or we may lose
access to important intellectual property or know-how.
The success of our clinical trials and preclinical studies may not be indicative of results in a large number of subjects of either safety or efficacy. |
The successful results of our technology in preclinical studies
using animal models may not be predictive of the results that we
will see in our clinical trials. In addition, results in
early-stage clinical trials are based on limited numbers of
subjects and generally test for drug safety rather than
efficacy. Our reported progress and results from our early
phases of clinical testing of our product candidates may not be
indicative of progress or results that will be achieved from
larger populations, which could be less favorable. Moreover, we
do not know if the favorable results we have achieved in
clinical trials will have a lasting or repeatable effect. If a
larger group of subjects does not experience positive results or
if any favorable results do not demonstrate a beneficial effect,
our product candidates that we advance to clinical trials, may
not receive approval from the FDA for further clinical trials or
commercialization. For example, in March 2005, we discontinued
the development of tgAAVCF following the analysis of
Phase II clinical trial data in which tgAAVCF failed to
achieve the efficacy endpoints of the trial.
We may be unable to adequately protect our proprietary rights domestically or overseas, which may limit our ability to successfully market any product candidates. |
Our success depends substantially on our ability to protect our
proprietary rights and operate without infringing on the
proprietary rights of others. We own or license patents and
patent applications, and will need to license additional
patents, for genes, processes, practices and techniques critical
to our present and potential product candidates. If we fail to
obtain and maintain patent or other intellectual property
protection for this
21
Table of Contents
technology, our competitors could market competing products
using those genes, processes, practices and techniques. The
patent process takes several years and involves considerable
expense. In addition, patent applications and patent positions
in the field of biotechnology are highly uncertain and involve
complex legal, scientific and factual questions. Our patent
applications may not result in issued patents and the scope of
any patent may be reduced both before and after the patent is
issued. Even if we secure a patent, the patent may not provide
significant protection and may be circumvented or invalidated.
We also rely on unpatented proprietary technology and technology
that we have licensed on a nonexclusive basis. While we take
precautions to protect our proprietary unpatented technology, we
may be unable to meaningfully protect this technology from
unauthorized use or misappropriation by a third party. Our
competitors could also obtain rights to our nonexclusively
licensed proprietary technology. In any event, other companies
may independently develop equivalent proprietary information and
techniques. If our competitors develop and market competing
products using our unpatented or nonexclusively licensed
proprietary technology or substantially similar technology, our
products, if successfully developed, could suffer a reduction in
sales or be forced out of the market.
If we do not develop adequate development, manufacturing, sales, marketing and distribution capabilities, either alone or with our business partners, we will be unable to generate sufficient product revenue to maintain our business. |
Our potential products require significant development of new
processes and design for the advancement of the product
candidate through manufacture, preclinical and clinical testing.
We may be unable to continue development or meet critical
milestones with our partners due to technical or scientific
issues related to manufacturing or development. We currently do
not have the physical capacity to manufacture large-scale
quantities of our potential products. This could limit our
ability to conduct large clinical trials of a product candidate
and to commercially launch a successful product candidate. In
order to manufacture product at such scale, we will need to
expand or improve our current facilities and staff or supplement
them through the use of contract providers. If we are unable to
obtain and maintain the necessary manufacturing capabilities,
either alone or through third parties, we will be unable to
manufacture our potential products in quantities sufficient to
sustain our business. Moreover, we are unlikely to become
profitable if we, or our contract providers, are unable to
manufacture our potential products in a cost-effective manner.
In addition, we have no experience in sales, marketing and
distribution. To successfully commercialize any products that
may result from our development programs, we will need to
develop these capabilities, either on our own or with others. We
intend to enter into collaborations with other entities to
utilize their mature marketing and distribution capabilities,
but we may be unable to enter into marketing and distribution
agreements on favorable terms, if at all. If our current or
future collaborative partners do not commit sufficient resources
to timely marketing and distributing our future products, if
any, and we are unable to develop the necessary marketing and
distribution capabilities on our own, we will be unable to
generate sufficient product revenue to sustain our business.
Post-approval manufacturing or product problems or failure to satisfy applicable regulatory requirements could prevent or limit our ability to market our products. |
Commercialization of any products will require continued
compliance with FDA and other federal, state and local
regulations. For example, our current manufacturing facility,
which is designed for manufacturing our AAV vectors for clinical
and development purposes, is subject to the Good Manufacturing
Practices requirements and other regulations of the FDA, as well
as to other federal, state and local regulations such as the
Occupational Health and Safety Act, the Toxic Substances Control
Act, the Resource Conservation and Recovery Act and the
Environmental Protection Act. Any future manufacturing
facilities that we may construct for large-scale commercial
production will also be subject to regulation. We may be unable
to obtain regulatory approval for or maintain in operation this
or any other manufacturing facility. In addition, we may be
unable to attain or maintain compliance with current or future
regulations relating to manufacture, safety, handling, storage,
record keeping or marketing of potential products. If we fail to
comply with applicable regulatory requirements or discover
previously unknown manufacturing, contamination, product side
effects or
22
Table of Contents
other problems after we receive regulatory approval for a
potential product, we may suffer restrictions on our ability to
market the product or be required to withdraw the product from
the market.
Risks Related to Our Industry
Adverse events in the field of gene therapy could damage public perception of our potential products and negatively affect governmental approval and regulation. |
Public perception of our product candidates could be harmed by
negative events in the field of gene transfer. For example, in
2003, fourteen subjects in a French academic clinical trial
being treated for x-linked severe combined immunodeficiency in a
gene therapy trial using a retroviral vector showed correction
of the disease, although three of the subjects subsequently
developed leukemia. Serious adverse events, including patient
deaths have occurred in clinical trials. Adverse events in our
clinical trials and the resulting publicity, as well as any
other adverse events in the field of gene therapy that may occur
in the future, could result in a decrease in demand for any
products that we may develop. The commercial success of our
product candidates will depend in part on public acceptance of
the use of gene therapy for preventing or treating human
diseases. If public perception is influenced by claims that gene
therapy is unsafe, our product candidates may not be accepted by
the general public or the medical community. The public and the
medical community may conclude that our technology is unsafe.
Future adverse events in gene therapy or the biotechnology
industry could also result in greater governmental regulation,
unfavorable public perception, stricter labeling requirements
and potential regulatory delays in the testing or approval of
our potential products. Any increased scrutiny could delay or
increase the costs of our product development efforts or
clinical trials.
Our use of hazardous materials exposes us to liability risks and regulatory limitations on their use, either of which could reduce our ability to generate product revenue. |
Our research and development activities involve the controlled
use of hazardous materials, including chemicals, biological
materials and radioactive compounds. Our safety procedures for
handling, storing and disposing of these materials must comply
with federal, state and local laws and regulations, including,
among others, those relating to solid and hazardous waste
management, biohazard material handling, radiation and air
pollution control. We may be required to incur significant costs
in the future to comply with environmental or other applicable
laws and regulations. In addition, we cannot eliminate the risk
of accidental contamination or injury from hazardous materials.
If a hazardous material accident were to occur, we could be held
liable for any resulting damages, and this liability could
exceed our insurance and financial resources. Accidents
unrelated to our operations could cause federal, state or local
regulatory agencies to restrict our access to hazardous
materials needed in our research and development efforts, which
could result in delays in our research and development programs.
Paying damages or experiencing delays caused by restricted
access could reduce our ability to generate revenue and make it
more difficult to fund our operations.
The intense competition and rapid technological change in our market may result in failure of our potential products to achieve market acceptance. |
We face increasingly intense competition from a number of
commercial entities and institutions that are developing gene
therapy technologies. Our competitors include early-stage and
more established gene delivery companies, other biotechnology
companies, pharmaceutical companies, universities, research
institutions and government agencies developing gene therapy
products or other biotechnology-based therapies designed to
treat the diseases on which we focus. We also face competition
from companies using more traditional approaches to treating
human diseases, such as surgery, medical devices and
pharmaceutical products. As our product candidates become
commercial gene therapy products that may affect commercial
markets of the analogous protein or traditional pharmaceutical
therapy, disputes including lawsuits, demands, threats or patent
challenges may arise in an effort to slow our development. In
addition, we compete with other companies to acquire products or
technology from research institutions or universities. Many of
our competitors have substantially
23
Table of Contents
more resources, including research and development personnel,
capital and infrastructure, than we do. Many of our competitors
also have greater experience and capabilities than we do in:
| research and development; | |
| clinical trials; | |
| obtaining FDA and other regulatory approvals; | |
| manufacturing; and | |
| marketing and distribution. |
In addition, the competitive positions of other companies,
institutions and organizations, including smaller competitors,
may be strengthened through collaborative relationships.
Consequently, our competitors may be able to develop, obtain
patent protection for, obtain regulatory approval for, or
commercialize new products more rapidly than we do, or
manufacture and market competitive products more successfully
than we do. This could limit the prices we could charge for the
products that we are able to market or result in our products
failing to achieve market acceptance.
Gene therapy is a rapidly evolving field and is expected to
continue to undergo significant and rapid technological change
and competition. Rapid technological development by our
competitors, including development of technologies, products or
processes that are more effective or more economically feasible
than those we have developed, could result in our actual and
proposed technologies, products or processes losing market share
or becoming obsolete.
Healthcare reform measures and the unwillingness of third-party payors to provide adequate reimbursement for the cost of our products could impair our ability to successfully commercialize our potential products and become profitable. |
Sales of medical products and treatments depends substantially,
both domestically and abroad, on the availability of
reimbursement to the consumer from third-party payors. Our
potential products may not be considered cost-effective by
third-party payors, who may not provide coverage at the price
set for our products, if at all. If purchasers or users of our
products are unable to obtain adequate reimbursement, they may
forego or reduce their use of our products. Even if coverage is
provided, the approved reimbursement amount may not be high
enough to allow us to establish or maintain pricing sufficient
to realize a sufficient return on our investment.
Increasing efforts by governmental and third-party payors, such
as Medicare, private insurance plans and managed care
organizations, to cap or reduce healthcare costs will affect our
ability to commercialize our product candidates and become
profitable. We believe that third-party payors will attempt to
reduce healthcare costs by limiting both coverage and level of
reimbursement for new products approved by the FDA. There have
been and will continue to be a number of federal and state
proposals to implement government controls on pricing, the
adoption of which could affect our ability to successfully
commercialize our product candidates. Even if the government
does not adopt any such proposals or reforms, their announcement
could impair our ability to raise capital.
Risks Related to Our Common Stock
If we are unable to comply with the minimum requirements for quotation on the NASDAQ Capital Market and we may be delisted from the NASDAQ Capital Market, the liquidity and market price of our common stock would decline. |
Our stock is listed on the NASDAQ Capital Market. In order to
continue to be listed on the NASDAQ Capital Market, we must meet
specific quantitative standards, including maintaining a minimum
bid price of $1.00 for our common stock. On May 31, 2005,
we received a notice from the NASDAQ Stock Market informing us
that for 30 consecutive business days the bid price of our
common stock had closed below the minimum $1.00 per share
requirement for continued inclusion under Marketplace
Rule 4310(c)(4). The
24
Table of Contents
letter stated that under Marketplace Rule 4310(c)(8)(d), we
were provided with 180 calendar days, or until November 28,
2005, to regain compliance with Marketplace
Rule 4310(c)(4). To regain compliance, the bid price of our
common stock must close at $1.00 per share or more for a
minimum of 10 consecutive business days. As of November 28,
2005, we had not regained compliance with Market place
Rule 4310(c). However, since we met all of the NASDAQ Stock
Markets criteria set forth in Marketplace
Rule 4310(c), except for the bid price requirement, the
NASDAQ provided us with an additional 180 calendar day
compliance period, or until May 26, 2006, to demonstrate
full compliance and maintain our listing on the NASDAQ Capital
Market. To date, we have not achieved the required minimum bid
price. If we have not regained compliance at that time, the
NASDAQ Staff will provide written notification that our
securities will be delisted.
Upon delisting from the NASDAQ Capital Market, trading, if any,
in our shares may continue to be conducted on the OTC
Bulletin Board or in a non-NASDAQ
over-the-counter
market, such as the pink sheets. Delisting of our
shares would result in limited release of the market price of
those shares and limited analyst coverage and could restrict
investors interest in our securities. Also, a delisting
could have a material adverse effect on the trading market and
prices for our shares and our ability to issue additional
securities or to secure additional financing. In addition, if
our shares were not listed and the trading price of our shares
was less than $5 per share, our shares could be subject to
Rule 15g-9 under
the Securities Exchange Act of 1934 which, among other things,
requires that broker/ dealers satisfy special sales practice
requirements, including making individualized written
suitability determinations and receiving a purchasers
written consent prior to any transaction. In such case, our
securities could also be deemed to be a penny stock
under the Securities Enforcement and Penny Stock Reform Act of
1990, which would require additional disclosure in connection
with trades in those shares, including the delivery of a
disclosure schedule explaining the nature and risks of the penny
stock market. Such requirements could severely limit the
liquidity of our securities.
If we sell additional shares, our stock price may decline as a result of the dilution which will occur to existing stockholders. |
Until we are profitable, we will need significant additional
funds to develop our business and sustain our operations. Any
additional sales of shares of our common stock are likely to
have a dilutive effect on our then existing stockholders.
Subsequent sales of these shares in the open market could also
have the effect of lowering our stock price, thereby increasing
the number of shares we may need to issue in the future to raise
the same dollar amount and consequently further diluting our
outstanding shares. These future sales could also have an
adverse effect on the market price of our shares and could
result in additional dilution to the holders of our shares.
The perceived risk associated with the possible sale of a large
number of shares could cause some of our stockholders to sell
their stock, thus causing the price of our stock to decline. In
addition, actual or anticipated downward pressure on our stock
price due to actual or anticipated sales of stock could cause
some institutions or individuals to engage in short sales of our
common stock, which may itself cause the price of our stock to
decline.
If our stock price declines, we may be unable to raise
additional capital. A sustained inability to raise capital could
force us to go out of business. Significant declines in the
price of our common stock could also impair our ability to
attract and retain qualified employees, reduce the liquidity of
our common stock and result in the delisting of our common stock
from the NASDAQ Capital Market.
Concentration of ownership of our common stock may give certain shareholders significant influence over our business. |
A small number of investors own a significant number of shares
of our common stock. As of December 31, 2005, Elan held
approximately 11.6 million and Biogen Idec held
approximately 11.7 million shares of our common stock.
Together these holdings represent approximately 27.2% of our
common shares outstanding as of December 31, 2005. This
concentration of stock ownership may allow these shareholders to
25
Table of Contents
exercise significant control over our strategic decisions and
block, delay or substantially influence all matters requiring
shareholder approval, such as:
| election of directors; | |
| amendment of our charter documents; or | |
| approval of significant corporate transactions, such as a change of control of Targeted Genetics. |
The interests of these shareholders may conflict with the
interests of other holders of our common stock with regard to
such matters. Furthermore, this concentration of ownership of
our common stock could allow these shareholders to delay, deter
or prevent a third party from acquiring control of Targeted
Genetics at a premium over the then-current market price of our
common stock, which could result in a decrease in our stock
price.
Both Biogen Idec and Elan have sold shares of our common stock
and may continue to do so. In accordance with the termination
agreement that we entered into with Elan with in March 2004,
Elan is only permitted to sell quantities of stock our equal to
175% of the volume limitation set forth in Rule 144(e)(1)
promulgated under the Securities Act of 1933, as amended.
Market fluctuations or volatility could cause the market price of our common stock to decline and limit our ability to raise capital. |
The stock market in general and the market for
biotechnology-related companies in particular have experienced
extreme price and volume fluctuations, often unrelated to the
operating performance of the affected companies. The market
price of the securities of biotechnology companies, particularly
companies such as ours without earnings and product revenue, has
been highly volatile and is likely to remain so in the future.
Any report of clinical trial results that are below the
expectations of financial analysts or investors could result in
a decline in our stock price. We believe that in the past,
similar levels of volatility have contributed to the decline in
the market price of our common stock, and may do so again in the
future. Trading volumes of our common stock can increase
dramatically, resulting in a volatile market price for our
common stock. The trading price of our common stock could
decline significantly as a result of sales of a substantial
number of shares of our common stock, or the perception that
significant sales could occur. In addition, the sale of
significant quantities of stock by Elan, Biogen Idec or other
holders of significant amounts of shares of our stock, could
adversely impact the price of our common stock.
Item 2. | Properties. |
We have leased approximately 51,000 square feet of
laboratory, manufacturing and office space in two buildings in
Seattle, Washington. The lease on our primary laboratory,
manufacturing and office space expires in April 2009 and has one
option to renew for a five-year period. The lease on our
administrative office space expires in March 2009 and includes
two options to extend the lease for a total of five additional
years. We have an option to cancel the lease on our
administrative offices at any time between April 2006 and March
2009 with certain early termination penalties. We believe that
our Seattle facilities are sufficient to support our research,
manufacturing and administrative needs under our current
operating plan.
In July 2000, we leased approximately 76,000 square feet of
space in Bothell, Washington, intended for future large-scale
manufacturing of our products. The lease on this facility
expires in September 2015 and includes an option for us to
extend its term for one additional five-year period. While
preliminary design activities have been completed, we have never
occupied this facility and do not currently plan to commence the
construction of this facility unless and until product demands
warrant resumption of construction activities. As a result, we
are trying to sublease all or part of the facility. Any decision
to resume use of the facility will be based on a number of
factors, including the progress of our product candidates in
clinical development, the estimated duration of facility design
and construction, the estimated timing of product manufacturing
requirements, the ability of our current manufacturing
capabilities to meet demand, and the availability of financial
resources.
26
Table of Contents
Item 3. | Legal Proceedings. |
We are not a party to any material legal proceedings.
Item 4. | Submission of Matters to a Vote of Security Holders. |
No matters were submitted to a vote of our security holders
during the fourth quarter of 2005.
PART II
Item 5. | Market for Registrants Common Equity, Related Shareholder Matters and Issuer Purchases of Equity Securities. |
Market Information. Our common stock trades on the NASDAQ
Capital Market under the symbol TGEN. From May 20, 1994
until January 8, 2003, our common stock was traded on the
NASDAQ National Market, under the symbol TGEN.
The following table lists, for each calendar quarter indicated,
the high and low bid quotations for our common stock, as quoted
on the NASDAQ Capital Market or National Market as applicable.
These quotes reflect inter-dealer prices, without retail
mark-up or commission,
and may not necessarily represent actual transactions.
High | Low | ||||||||
2005:
|
|||||||||
4th Quarter
|
$ | 0.72 | $ | 0.48 | |||||
3rd Quarter
|
0.92 | 0.60 | |||||||
2nd Quarter
|
1.25 | 0.50 | |||||||
1st Quarter
|
1.90 | 0.40 | |||||||
2004:
|
|||||||||
4th Quarter
|
$ | 1.98 | $ | 1.13 | |||||
3rd Quarter
|
1.62 | 0.94 | |||||||
2nd Quarter
|
2.22 | 1.24 | |||||||
1st Quarter
|
3.29 | 1.80 |
The last reported bid quotation for our common stock, as quoted
on the NASDAQ Capital Market on March 10, 2006 was
$0.42 per share.
Holders. As of March 7, 2006, we had
364 shareholders of record and approximately 21,000
beneficial holders of our common stock.
Dividends. We have never paid cash dividends and do not
anticipate paying them in the foreseeable future. In addition,
our loan agreement with Biogen restricts the amount of cash
dividends we could pay.
27
Table of Contents
Item 6. | Selected Financial Data. |
Year Ended December 31, | ||||||||||||||||||||||
2005(2) | 2004(2)(3) | 2003(2) | 2002(1)(2) | 2001 | ||||||||||||||||||
Statement of Operations Data
|
||||||||||||||||||||||
Revenue
|
$ | 6,874,000 | $ | 9,652,000 | $ | 14,073,000 | $ | 19,333,000 | $ | 18,880,000 | ||||||||||||
Operating expenses
|
26,221,000 | 24,822,000 | 27,877,000 | 42,074,000 | 47,484,000 | |||||||||||||||||
Loss from operations
|
(19,347,000 | ) | (15,170,000 | ) | (13,804,000 | ) | (22,741,000 | ) | (28,604,000 | ) | ||||||||||||
Net loss applicable to common stock
|
$ | (19,198,000 | ) | $ | (14,257,000 | ) | $ | (14,833,000 | ) | $ | (23,767,000 | ) | $ | (27,170,000 | ) | |||||||
Net loss per basic and diluted common share
|
$ | (0.22 | ) | $ | (0.18 | ) | $ | (0.26 | ) | $ | (0.52 | ) | $ | (0.62 | ) | |||||||
Shares used in computing basic and diluted net loss per common
share
|
85,635,000 | 79,451,000 | 57,486,000 | 45,767,000 | 43,928,000 | |||||||||||||||||
December 31, | |||||||||||||||||||||
2005 | 2004 | 2003 | 2002 | 2001 | |||||||||||||||||
Balance Sheet Data
|
|||||||||||||||||||||
Cash and cash equivalents
|
$ | 14,122,000 | $ | 34,096,000 | $ | 21,057,000 | $ | 12,606,000 | $ | 25,186,000 | |||||||||||
Total assets
|
48,798,000 | 69,965,000 | 57,672,000 | 52,713,000 | 71,038,000 | ||||||||||||||||
Long-term obligations
|
8,177,000 | 10,182,000 | 11,227,000 | 20,494,000 | 16,403,000 | ||||||||||||||||
Preferred stock(4)
|
| | 12,015,000 | 12,015,000 | 12,015,000 | ||||||||||||||||
Total shareholders equity
|
$ | 30,571,000 | $ | 49,762,000 | $ | 33,479,000 | $ | 5,896,000 | $ | 25,386,000 |
(1) | Effective January 1, 2002, we changed our method of accounting for goodwill and other intangible assets. See Note 1 of the notes to our consolidated financial statements. |
(2) | Operating expenses include restructure charges of $2.3 million in 2002, $5.2 million in 2003, $884,000 in 2004 and $1.7 million in 2005. See Note 4 of the notes to our consolidated financial statements. |
(3) | Reflects a $1.0 million gain on the sale of a majority-owned subsidiary in July 2004. See Note 7 of the notes to our consolidated financial statements. |
(4) | As a result of the expiration of an exchange right of the holder in April 2003, we reclassified the Series B preferred stock from mezzanine equity to shareholders equity. The Series B preferred stock was converted by the holder into common stock in March 2004. |
Item 7. | Managements Discussion and Analysis of Financial Condition and Results of Operations. |
OVERVIEW
Targeted Genetics Corporation is a clinical-stage therapeutic
biotechnology company. We are at the forefront of developing,
with the goal of commercializing, a new class of gene
therapeutics. We believe that a wide range of diseases may
potentially be treated or prevented with gene-based products. In
addition to treating diseases which have not had treatments in
the past, we believe that there is also a significant
opportunity to use gene-based products to more effectively treat
diseases that are currently treated using other therapeutic
classes of drugs such as protein-based drugs, monoclonal
antibodies, or small molecule drugs.
Gene-based products consist of a delivery vehicle, called a
vector, and genetic material. The role of the vector is to carry
the genetic material into a target cell. Once delivered into the
cell, the gene can express or direct production of the specific
proteins encoded by the gene. Gene-based therapeutics may be
used to treat disease by replacing the missing or defective gene
to facilitate the normal protein production or gene
28
Table of Contents
regulation capabilities of cells. In addition, gene delivery may
be used to enable cells to perform additional roles in the body.
We have four product candidates, two of which are currently in
clinical trials. Our clinical-stage candidates are aimed at
inflammatory arthritis and HIV/ AIDS. Our lead product
candidate, tgAAC94, for the treatment of inflammatory arthritis
is in Phase I clinical trials. Our HIV/ AIDS prophylactic
vaccine product candidate, which we are developing in
collaboration with the International AIDS Vaccine Initiative, or
IAVI, to protect against the progression of HIV infection to
AIDS is in Phase II clinical trials. Our preclinical
product candidates, both in development with collaboration
partners, are aimed at congestive heart failure and
Huntingtons disease.
We have financed the company primarily through proceeds from
public and private sales of our equity securities, through cash
payments received from our collaborative partners for product
development and manufacturing activities, through proceeds from
the issuance of debt and loan funding under equipment financing
arrangements. On March 10, 2006, we sold
12,791,611 shares of our common stock in a public offering
at a price of $0.39 per share and received net proceeds of
approximately $4.8 million.
Our primary expenses are related to the development of our
research and development programs, the conduct of preclinical
studies and clinical trials and general and administrative
support for these activities and accordingly, to an extent, we
are able to reduce expenses and conserve our resources by
reducing these activities. For example, in January 2006 we
restructured our operations to concentrate our resources on
generating data from the clinical trials of our inflammatory
arthritis product candidate, maintaining our manufacturing
capabilities and advancing our funded product development
efforts. As part of this restructuring we eliminated 26
positions, which reduced our workforce by approximately 27%,
including scientific, operations and administrative functions
that were not required to support our programs. This
restructuring primarily affected earlier-stage product discovery
and preclinical product development efforts, small-scale vector
manufacturing employees, management positions and operations and
general and administrative support.
As of December 31, 2005, our accumulated deficit totaled
$250.0 million. We expect to generate substantial
additional losses for the foreseeable future, primarily due to
the costs associated with funding our inflammatory arthritis
clinical development program, developing and maintaining our
manufacturing capabilities and developing our intellectual
property assets.
Notwithstanding our January 2006 cost cutting measures, we will
require access to significantly higher amounts of capital than
we currently have in order to successfully develop our lead
inflammatory arthritis product candidate or our partnered
product candidates. We may be unable to obtain required funding
when needed or on acceptable terms, obtain or maintain corporate
partnerships or complete acquisition transactions necessary or
desirable to complete the development of our product candidates.
CRITICAL ACCOUNTING POLICIES, ESTIMATES AND ASSUMPTIONS
Our discussion and analysis of our financial condition and
results of operations is based upon financial statements that we
have prepared in accordance with accounting principles generally
accepted in the United States. As we prepare our financial
statements we are required to make estimates and judgments that
affect the reported amounts of assets, liabilities, revenues and
expenses, and related disclosures. On an on-going basis, we
evaluate these estimates, including those related to revenue,
accrued restructure charges and goodwill. Estimates are based on
historical experience, information received from third parties
and on various other assumptions that are believed to be
reasonable under the circumstances, the results of which form
the basis for making judgments about the carrying values of
assets and liabilities that are not readily apparent from other
sources. Our actual results may differ from these estimates
under different assumptions or conditions. Note 1 of the
notes to our consolidated financial statements,
Description of Business and Summary of Significant
Accounting Policies, summarizes our significant
accounting policies that we believe are critical to
29
Table of Contents
the presentation of our consolidated financial statements. Our
most critical accounting policies, estimates and assumptions are:
Revenue Recognition Policy |
We generate revenue from technology licenses, collaborative
research arrangements and agreements to provide research,
development and manufacturing services. Revenue under technology
licenses and collaborative agreements typically consists of
nonrefundable, up-front license fees, collaborative research
funding, technology access fees and various other payments.
Revenue from nonrefundable, up-front license fees and technology
access payments is recognized systematically over the related
service period of the collaborative agreement, which is often
the development period. Revenue associated with performance
milestones is recognized as earned, based upon the achievement
of the milestones as defined in the applicable agreements.
Revenue under research and development cost-services contracts
is recognized as the related costs are incurred. Advance
payments received in excess of amounts earned are classified as
deferred revenue.
Estimated Restructuring Charges Associated with the Bothell Facility |
We have adopted the provisions of Statement of Financial
Accounting Standards No. 146, or SFAS No. 146,
Accounting for Costs Associated with Exit or Disposal
Activities, as it relates to our facility in Bothell,
Washington and our former facility in Sharon Hill, Pennsylvania
and we have recorded restructure charges on the related
operating leases. Accrued restructuring charges, and in
particular, those charges associated with exiting a facility,
are subject to many assumptions and estimates. Under
SFAS No. 146, an accrued liability for lease
termination costs is initially measured at fair value, based on
the remaining lease payments due under the lease and other
costs, reduced by sublease rental income that could be
reasonably obtained from the property, and discounted using a
credit-adjusted risk-free interest rate.
The assumptions as to estimated sublease rental income and the
period of time and concessions necessary to enter into a
sublease significantly impact the accrual and may differ from
what actually occurs. We periodically evaluate these
restructuring estimates and assumptions and record additional
restructure charges as necessary to reflect current market
conditions and delays in subleasing the Bothell facility.
Changes to our restructuring estimates and assumptions can be
material. For example in the third quarter of 2005, we revised
our restructuring accrual estimates to account for a change in
approach to leasing the entire Bothell facility, which resulted
in additional restructuring charges of $1.0 million. As a
result of periodic evaluations and updates we have recorded
$6.2 million in additional restructure charges since first
establishing a restructuring reserve for exiting the Bothell
facility. We also record accretion expense based upon changes in
the accrued restructure liability that results from the passage
of time at an assumed discount rate of 10%. We record accretion
expense as a restructuring charge, which totaled $577,000 in
2005, $513,000 in 2004 and $435,000 in 2003.
We will continue to evaluate any additional information that may
become available with respect to the estimates and assumptions
as they relate to these facilities, which may result in further
significant charges to our results of operations. If
circumstances with the lease change, or if we decide to resume
use of this facility, any remaining accrued restructure charges
related to the facility will be reversed. This reversal would be
reflected as a reduction of restructuring expenses and reflected
in the period in which use is resumed. We are unable to
determine the likelihood of any future adjustments to our
accrued restructuring charges.
Valuation of Our Goodwill and Intangible Assets |
In 2000, we acquired Genovo, Inc., a development-stage
biotechnology company, for a purchase price of
$66.4 million. We allocated the excess of the acquisition
cost over the fair value of the identifiable net assets acquired
to goodwill totaling $38.2 million and to other purchased
intangibles totaling $605,000. From 2000 through 2001, we
recorded amortization expenses of $7.1 million of Genovo
goodwill and purchased intangibles. We test goodwill for
impairment at least annually, and more frequently when events or
circumstances indicate the carrying value may be impaired, by
comparing its carrying value to the market value of our shares
outstanding. Events or circumstances which could trigger an
impairment review include a significant adverse change in our
business climate, significant changes in our use of acquired
technology, and
30
Table of Contents
changes to our overall business strategy. In the event that our
valuation tests show an impairment in the recorded value of our
goodwill, we may record a significant non-cash charge to
expense. We have performed annual impairment tests as of
October 1 and periodic tests as we see necessary each year
since the implementation of SFAS No. 142 and concluded
that no impairment in the value of our goodwill had occurred.
Application of New Accounting Standards |
In December 2004, the FASB released its final revised standard,
SFAS No. 123R, Share-Based Payment.
SFAS No. 123R will require us to expense the fair
value of stock options granted over the vesting period.
Currently, we account for stock options under Accounting
Principles Board No. 25, Accounting for Stock
Issued to Employees, which uses the intrinsic value
method and generally recognizes no compensation cost for
employee stock option grants. We are required to adopt of
SFAS No. 123R beginning January 1, 2006. We are
evaluating SFAS No. 123R and believe it will have a
material effect on our consolidated financial statements.
The summary of significant accounting policies should be read in
conjunction with our consolidated financial statements and
related notes and the following discussion of our results of
operations and liquidity and capital resources.
RESULTS OF OPERATIONS
Revenue |
Year Ended December 31, | |||||||||||||
2005 | 2004 | 2003 | |||||||||||
Revenue from collaborative agreements:
|
|||||||||||||
HIV/ AIDS vaccine IAVI
|
$ | 5,588,000 | $ | 8,340,000 | $ | 4,409,000 | |||||||
Congestive heart failure Celladon
|
777,000 | | | ||||||||||
Huntingtons disease Sirna
|
315,000 | | | ||||||||||
Multiple products Biogen
|
| | 5,112,000 | ||||||||||
Hemophilia Wyeth
|
| | 3,894,000 | ||||||||||
Contract manufacturing revenue and other
|
194,000 | 1,312,000 | 658,000 | ||||||||||
Total revenue
|
$ | 6,874,000 | $ | 9,652,000 | $ | 14,073,000 | |||||||
Total revenue in 2005 was $6.9 million, compared to
$9.7 million in 2004. Revenue in 2005 consists primarily of
amounts earned under our HIV/ AIDS vaccine collaboration with
IAVI which decreased to $5.6 million in 2005 from
$8.3 million in 2004. This decrease in revenue reflects the
completion of certain development activities as the program
progresses into the Phase I and Phase II clinical
trials. During 2005 we incurred $2.8 million to support
development activities under the Celladon collaboration, which
consisted primarily of internal development and manufacturing
efforts. We agreed to contribute up to $2.0 million to
support these development activities and we are reimbursed for
efforts over that amount. During the current year, we were
reimbursed $0.8 million for those labor hours and those
outside costs associated with the Celladon collaboration.
Revenue from the Sirna collaboration represents our program
costs that exceed more than half of the total collaboration
costs. The decrease in total revenue for 2004 compared to 2003
is the result of revenues earned in 2003 under our former
development collaborations with Biogen and Wyeth. Revenue in
2003 includes $3.9 million of revenue related to the
termination of our collaboration with Wyeth and
$5.1 million of revenue from our collaboration with Biogen
including $2.6 million in revenue we recognized upon the
completion of this development collaboration in September 2003.
The increase in 2004 revenue earned under our IAVI HIV/ AIDS
vaccine collaboration as compared to 2003 revenue reflects
higher 2004 manufacturing and development activities supporting
the addition of a second vaccine candidate.
We expect that substantially all of our 2006 revenue will
consist of research and development revenue from our
collaborations with IAVI, Celladon, and a NIAID funded
subcontract with Childrens Hospital of
31
Table of Contents
Philadelphia. We expect revenues associated with IAVI will be
lower in 2006 than in 2005 due to the completion of planned
development activities and scheduled product manufacturing.
Whereas, we expect revenue associate with Celladon will increase
as Celladon works towards beginning a Phase I clinical
trial with their congestive heart failure product in early 2007.
In addition, we are expecting revenue from our recently
initiated HIV/ AIDS vaccine collaboration with NIAID, CCRI and
Childrens Hospital of Philadelphia. Our revenue for the
next several years will be dependent on the continuation of our
current collaborations reaching milestones on our congestive
heart failure, NIAID and HIV/ AIDS vaccine product development
efforts and whether we enter into any new collaborations.
Operating Expenses |
Research and Development. Research and development
expenses totaled $18.2 million in 2005, compared to
$17.3 million in 2004. This increase primarily reflects the
costs associated with our programs in preclinical development
which was $8.1 million in 2005 as compared to
$5.3 million in 2004. The increase is primarily due to the
continued progress of our inflammatory arthritis program and
initiation of the Celladon and Sirna collaborations in early
2005. Costs of our inflammatory arthritis program increased due
to the clinical costs associated with progress on our
Phase I clinical trials. These increases were offset by
decreased HIV/ AIDS vaccine development costs as the project
progresses through the clinical stages of development. While
total research and development expenses in 2004 were comparable
to 2003, the costs associated with our programs in clinical
development increased from $1.5 million in 2003 to
$12.0 million in 2004 reflecting the initiation of clinical
trials for our HIV/ AIDS vaccine program in December 2003 and
our inflammatory arthritis program in March 2004.
We currently expect our research and development expenses in
2006 to decrease somewhat as compared to 2005, as the result of
decreased levels of development and manufacturing activities
generated by our January 2006 reduction in force and related
re-alignment of our product development resources. Cost savings
generated by this re-alignment will be partially offset by costs
incurred to support the NIAID HIV/ AIDS vaccine program awarded
in 2005. Our research and development expenses fluctuate due to
the timing of expenditures for the varying stages of our
research, product development and clinical development programs
and the availability of capital resources. We expect that our
expenses will continue to fluctuate as we proceed with our
current development collaborations, enter into potential new
development collaborations and licensing agreements, and
potentially earn milestone payments.
The following is an allocation of our total research and
development expenses between our programs in clinical
development and those that are in research or preclinical stages
of development:
Year Ended December 31, | |||||||||||||
2005 | 2004 | 2003 | |||||||||||
Programs in clinical development:
|
|||||||||||||
Inflammatory arthritis (initiated Phase I clinical trial in
March 2004)
|
$ | 2,444,000 | $ | 1,771,000 | $ | | |||||||
HIV/ AIDS vaccine (initiated Phase I clinical trial in
December 2003)
|
2,836,000 | 3,704,000 | 19,000 | ||||||||||
Cystic fibrosis
|
534,000 | 823,000 | 566,000 | ||||||||||
Cancer products
|
| | 14,000 | ||||||||||
Indirect costs
|
4,246,000 | 5,660,000 | 888,000 | ||||||||||
Total programs in clinical development
|
10,060,000 | 11,958,000 | 1,487,000 | ||||||||||
Programs in research and preclinical development
|
8,139,000 | 5,330,000 | 15,710,000 | ||||||||||
Total research and development expense
|
$ | 18,199,000 | $ | 17,288,000 | $ | 17,197,000 | |||||||
Research and development costs attributable to programs in
clinical development include costs of salaries, benefits,
clinical trial sites, outside services, materials and supplies
incurred to support the clinical programs. Indirect costs
allocated to clinical programs include facility and occupancy
costs, research and
32
Table of Contents
development administrative costs, and license and royalty
payments. These costs are further allocated between clinical and
preclinical programs based on relative levels of program
activity. IAVI separately manages and funds the clinical trial
costs of our HIV/ AIDS vaccine program and the Cystic Fibrosis
Foundation had separately funded the external costs of the
tgAAVCF clinical trials. As a result, we do not include those
costs in our research and development expenses.
Costs attributed to programs in research and preclinical
development represent our earlier-stage development activities
including costs incurred on programs prior to their transition
into clinical trials. Because we conduct multiple research
projects and utilize resources across several programs, the
majority of our research and preclinical development costs are
not directly assigned to individual programs, but are instead
allocated among multiple programs. For purposes of reimbursement
from our collaboration partners, we capture the level of effort
expended on a program through our project management system,
which is based primarily on human resource time allocated to
each program, supplemented by an allocation of indirect costs
and other specifically identifiable costs, if any. As a result,
the costs allocated to a program do not necessarily reflect the
actual costs of the program.
We initiated clinical testing of our HIV/ AIDS vaccine product
candidate in December 2003 and our inflammatory arthritis
product candidate in March 2004. All related development
activities associated with our inflammatory arthritis and HIV/
AIDS vaccine programs are reflected as costs associated with
programs in clinical development as of the date of initiation of
clinical testing. Therefore, during 2004 our research and
development expenses associated with programs under clinical
development increased reflecting the transition of these
programs from research and development into clinical testing.
General and Administrative. We incurred general and
administrative expenses of $6.3 million in 2005 compared to
$6.7 million in 2004. This decrease primarily reflects
costs eliminated as a result of our July 2004 sale of CellExSys.
We incurred general and administrative expenses of
$6.7 million in 2004 compared to $5.5 million in 2003.
This increase primarily reflects increased patent and
intellectual property costs, personnel costs and administrative
compliance costs. We expect our general and administrative
expenses in 2006 to decrease somewhat as compared to 2005,
primarily as the result of decreased staffing levels and lower
intellectual property costs.
Restructure Charges. Accrued restructuring charges
represent our best estimate of the fair value of the liability
to exit the Bothell facility as determined under
SFAS No. 146 and are computed as the fair value of the
difference between the remaining lease payments due on these
leases and estimated sub-lease costs and rentals. During 2005,
we adjusted our assumptions in March and September to reflect
our updated assumptions surrounding the Bothell facility.
Restructuring charges consist of the following:
Year Ended December 31, | ||||||||||||
2005 | 2004 | 2003 | ||||||||||
Charges related to changes in assumptions
|
$ | 1,132,000 | $ | 371,000 | $ | 4,718,000 | ||||||
Accretion expense
|
577,000 | 513,000 | 435,000 | |||||||||
Employee termination and other
|
| | 37,000 | |||||||||
Total restructuring charges
|
$ | 1,709,000 | $ | 884,000 | $ | 5,190,000 | ||||||
As of December 31, 2005, our accrued restructure liability
related to our Bothell facility was $7.1 million. In
January 2006, we incurred approximately $208,000 of
restructuring charges related to a reduction in force.
Evaluation of Goodwill and Other Intangible Assets. We
periodically and annually on October 1 evaluate the
carrying value of our goodwill in accordance with
SFAS No. 142, Goodwill and Other Intangible
Assets and if there is evidence of a impairment in
value, we reduce the carrying value of the asset. As of
December 31, 2005, we have concluded that there is no
impairment in the carrying value of our goodwill.
Investment Income. Investment income was $661,000 in 2005
compared to $383,000 in 2004, as a result of higher yields on
our investments during 2005 due to rising interest rates. In
2004, investment income was
33
Table of Contents
$383,000 as compared to $183,000 in 2003. This increase is
primarily the result of a higher level of invested funds
resulting from our common stock placement in February 2004 which
resulted in net proceeds of $23.7 million, and to a lesser
degree from higher yields on our investments.
Interest Expense. Interest expense relates to interest on
outstanding loans from our collaborative partners and
obligations under equipment financing arrangements that we used
to finance purchases of laboratory and computer equipment,
furniture and leasehold improvements. Interest expense increased
to $512,000 in 2005 from $476,000 in 2004. The increase is due
to rising interest rates, which increased the interest due on
the Biogen note payable. Interest expense decreased from
$1.2 million in 2003 due to lower debt balances resulting
from the conversion of $9.4 million owed to Elan into
equity in September 2003.
Gain on sale of majority-owned subsidiary. In July 2004,
Chromos Molecular Systems, Inc., or Chromos, acquired all of the
outstanding shares of our majority-owned subsidiary, CellExSys,
through a merger between CellExSys and Chromos Inc., a
wholly-owned subsidiary of Chromos. Under the terms of the
merger agreement, Chromos issued CellExSys shareholders
1,500,000 shares of Chromos common stock and a secured
convertible debenture totaling approximately $3.4 million
Canadian (approximately $2.5 million at the time of close).
As a result, we recorded a gain of $1.0 million
representing the deposits received from Chromos to fund
pre-closing operating costs, the fair value of our share of the
stock and debenture, and the net liabilities assumed by Chromos.
Liquidity and Capital Resources
We had cash and cash equivalents balances of $14.1 million
as of December 31 2005, $34.1 million as of
December 31, 2004 and $21.1 million as of
December 31, 2003. Our cash and cash equivalents decreased
in 2005 primarily reflecting our net loss of $19.2 million
and the resulting cash used in operations of $16.4 million.
Our 2005 cash flow results also include an early loan repayment
to Biogen of $2.5 million. Our cash and cash equivalents
increased in 2004 reflecting net proceeds of $29.8 million
from sales of our common stock, offset by our net loss for the
year of $14.3 million and the resulting cash used in
operations of $15.5 million.
To date, our primary sources of capital are from public and
private sales of our equity securities, through cash payments
received from our collaborative partners and proceeds from the
issuance of debt. To a lesser degree, we have also financed our
operations through interest earned on cash and short-term
investments, loan funding under equipment leasing agreements and
research grants. Our primary expenses are related to the
development of our research and development programs, the
conduct of preclinical studies and clinical trials and general
and administrative support for these activities.
Substantially all of our revenue to date has been derived under
collaborative research and development agreements relating to
the development of our potential product candidates. We do not
expect the revenue generated from our current or future
collaborative research and development arrangement to be
sufficient to fully fund the development and commercialization
of our product candidates. As a result, we do not expect to
generate positive cash flow from our operations for the
foreseeable future and our ability to generate any sustained
positive cash flow is dependent upon our success at developing
and commercializing our product candidates.
We will require substantial additional financial resources to
fund the development and commercialization of our lead product
candidate in inflammatory arthritis.
Since we implemented a reduction in force in January 2006, we
are currently focusing our development funding on our
inflammatory arthritis product candidate which is in
Phase I clinical trials. During 2005 we spent approximately
$2.4 million on this program in outside costs and allocated
staff costs to support research and development activities and
clinical trial costs, and we expect to spend approximately
$3 million to $4 million in 2006 on this program,
largely for clinical trial expenses. We currently fund all costs
of this program from our working capital and expect to do so for
the foreseeable future, although our strategy is to ultimately
seek a partner to fund later-stage development of this program.
34
Table of Contents
Our operating cash flows are primarily influenced by our losses
from operations, but are also affected by depreciation and
amortization of our property and equipment, accounts receivable,
deferred revenue and restructuring activity. Depreciation and
amortization charges for 2005 were $1.3 million, which were
flat with 2004 and down from $2.4 million in 2003. This
decrease in 2005 and 2004 as compared to 2003 primarily reflects
the lower manufacturing facility amortization. Accounts
receivable was a significant source of cash in 2003 as we had a
payment due from a collaborative partner at the end of 2002 that
was paid in 2003. Deferred revenue activity in 2005 and 2004
reflects changes in the levels of pre-funded work under our
collaborative agreements. In 2003, we recognized
$5.1 million of deferred revenue under our Biogen
collaboration. Increases in our restructure reserve, offset by
payments of rent for our Bothell facility and former Sharon Hill
facility resulted in net increases in reserves of $802,000 in
2005 and $3.4 million in 2003. In 2004, rent payments were
partially offset by charges, which resulted in a net decrease of
$523,000.
Our cash from investing activities included $3.0 million of
loan repayments including $2.5 million to Biogen and
$499,000 to financing companies in payment of our obligations
under equipment financing arrangements. This compares to
$866,000 of payments under our equipment financing arrangements
in 2004 and $1.3 million of payments in 2003.
Sales of common stock were significant contributors to our cash
flows from investing activities in 2004 and in 2003. Our
financial results in 2004 include $29.8 million of proceeds
as a result of the sale of 14,931,332 shares of common
stock. Our financial results in 2003 include $21.0 million
of proceeds as a result of the sale of 10,436,638 shares of
common stock.
Our primary source of capital historically has been from public
and private sales of our equity securities. On March 10,
2006, we sold 12,791,611 shares of our common stock in a
public offering at a price of $0.39 per share and received
net proceeds of approximately $4.8 million. We intend to
continue to seek appropriate opportunities to access the public
and private capital markets, however, our ability to issue
equity securities at the current market price will likely be
adversely affected by the fact that we are presently ineligible
under SEC rules to utilize
Form S-3 for
primary offerings of our securities because the aggregate market
value of our outstanding common stock held by non-affiliates is
less than $75 million.
Our near-term financing strategy includes leveraging our
development capabilities and intellectual property assets into
additional capital raising opportunities, advancing our clinical
development programs and accessing the public and private
capital markets at appropriate times. In addition, we intend to
manage our cash by focusing on advancing our inflammatory
product candidate through clinical testing.
Our development collaborations have typically provided us with
funding in several forms, including purchases of our equity
securities, loans, payments for reimbursement of research and
development costs and milestone fees and payments. We and our
partners typically agree on a target disease and create a
development plan for the product candidate, which often extends
for multiple years and is subject to termination or extension.
For example, when the IAVI collaboration was initiated in 2000,
it originally had a three-year term and our most recent
extension to the IAVI collaboration, extended the agreement
through December 2006. In November, 2005 we announced that we
extended the scope of our HIV/ AIDS vaccine program via a
collaboration between Childrens Hospital of Philadelphia,
CCRI and us under a contract awarded by the NIAID. Our portion
of the funding for this new collaboration could be up to
$18.2 million over the next five years. The funding from
each of our collaborative partners fully offsets our incremental
program costs from each collaboration and also partially funds
development of our inflammatory arthritis product candidate,
overhead and fixed costs. Our revenue from collaborative
agreements totaled $6.9 million in 2005, $9.7 million
in 2004 and $14.1 million in 2003 and assuming that we
complete all of the planned development activities for each of
these funded projects, we expect to earn revenue of up to
approximately $9 million in 2006.
Each of our collaborations has provisions that allow our
partners the right to terminate the underlying collaboration and
the obligation to provide research funding at any time with as
few as 90 days notice. If we were to lose the collaborative
funding expected from the IAVI, Celladon or NIAID collaborations
and were unable to obtain alternative sources of funding, we
would be unable to continue our research and development program
for that product candidate and our cash horizon would be
shortened.
35
Table of Contents
In addition to the funding necessary to advance our product
development and fund our ongoing operating costs, we also have
significant outstanding debt, lease commitments and long-term
obligations which draw on our cash resources. The following
table presents our contractual commitments:
Payments Due Through Year Ended December 31: | |||||||||||||||||||||||||||||
Contractual Obligations | 2006 | 2007 | 2008 | 2009 | 2010 | Thereafter | Total | ||||||||||||||||||||||
Long-term debt
obligations |
$ | | $ | 3,150,000 | $ | 2,500,000 | $ | 2,500,000 | $ | | $ | | $ | 8,150,000 | |||||||||||||||
Interest related to long-term debt obligations
|
388,000 | 334,000 | 205,000 | 75,000 | | | 1,002,000 | ||||||||||||||||||||||
Equipment financing obligations
|
155,000 | 26,000 | 1,000 | | | | 182,000 | ||||||||||||||||||||||
Interest related to equipment financing obligations
|
8,000 | 1,000 | | | | | 9,000 | ||||||||||||||||||||||
Operating lease obligations:
|
|||||||||||||||||||||||||||||
Seattle facilities occupied
|
974,000 | 1,002,000 | 1,030,000 | 259,000 | | | 3,265000 | ||||||||||||||||||||||
Bothell facility not occupied
|
1,362,000 | 1,362,000 | 1,362,000 | 1,362,000 | 1,431,000 | 7,770,000 | 14,649,000 | ||||||||||||||||||||||
Purchase obligations
|
708,000 | | | | | | 708,000 | ||||||||||||||||||||||
Total
|
$ | 3,595,000 | $ | 5,875,000 | $ | 5,098,000 | $ | 4,196,000 | $ | 1,431,000 | $ | 7,770,000 | $ | 27,965,000 | |||||||||||||||
In 2001, we borrowed $10 million from Biogen (now Biogen
Idec) to fund our general operations. This note was due in
August 2006. In September 2005, restructured the repayment of
this $10 million of debt and a separate $650,000 loan owed
to Biogen we incurred as part of our acquisition of Genovo.
Under the amended terms of these loans, we paid
$2.5 million of the outstanding debt in September 2005 and
agreed to make additional payments of $2.5 million in each
of August 2007, 2008 and August 2009. In addition, we agreed to
repay the $650,000 loan in August 2007. In addition, as part of
the restructuring agreement, we agreed to apply one-third of
certain up-front or milestone payments received from potential
corporate collaborations to repayment of the outstanding debt,
first to the payment of any accrued and unpaid interest on the
principal being repaid, and second to the payment of outstanding
principal in reverse order of maturity.
Operating lease obligations represent our commitments for our
facilities in Seattle and Bothell, Washington. Lease payments
for our Seattle facilities will total $3.3 million between
January 1, 2006 and the end of the lease in 2009. Lease
payments on the Bothell facility will total $14.6 million
between January 1, 2006 and the end of the lease in 2015.
We are pursuing efforts to sublease or otherwise reduce the
costs of our Bothell facility, and we may seek to sublease the
surplus portion of our office and laboratory space in our
Seattle facility.
Our research and development expenses fluctuate due to the
timing of expenditures for the varying stages of our research,
product development and clinical development programs and the
availability of capital resources. Because a significant portion
of our revenue and expense is directly tied to our research and
development activities, our revenue will fluctuate with the
level of future research and development activities. We expect
that our revenue and expense will continue to fluctuate as we
proceed with our current development collaborations, enter into
potential new development collaborations and licensing
agreements, and potentially earn milestone payments.
Over the past several years, we have scaled our development
activities to the level of available cash resources and
financial support from collaboration partners. Research and
development and general and administrative expenses increased by
approximately 2% in 2005 compared to 2004, increased by
approximately 6% in 2004 compared to 2003 and are expected to
decrease modestly in 2006 as a result of cost reduction efforts
implemented in January 2006 largely offset by additional
clinical trial costs to support the advancement of our
inflammatory arthritis program and certain incremental costs
necessary to support our new NIAID HIV/ AIDS vaccine
project. Assuming that our product development programs progress
at the rates
36
Table of Contents
currently planned, we believe that our net cash requirements
during 2006 will range from $13 million to
$16 million. This amount is subject to change as the result
of the outcome of our product development and business
development initiatives and other efforts.
We need to raise additional capital to complete our current
inflammatory arthritis clinical trial, evaluate the trial
results, and assuming satisfactory results, to plan and initiate
further clinical testing of our potential inflammatory arthritis
product. We expect that our cash and cash equivalents at
December 31, 2005, plus the funding expected from our
collaborative partners to fund 2006 work activities and proceeds
from the sale of our common stock in March 2006 will be
sufficient to fund our operations into the first quarter of 2007.
We are seeking avenues to exploit our intellectual property,
product development and manufacturing capabilities and
evaluating other opportunities to obtain additional capital to
fund our future operations. Additional sources of capital could
involve one or more of the following:
| entering into additional product development and funding collaborations or other strategic transactions, or extending or expanding our current collaborations; | |
| selling or licensing our technology or product candidates; | |
| issuing equity in the public or private markets; or | |
| issuing debt. |
Additional funding may not be available to us on reasonable
terms, if at all. Our ability to issue equity, and our ability
to issue it at the current market price, will likely be
adversely affected by the fact that we are presently ineligible
under SEC rules to utilize
Form S-3 for
primary offerings of our securities because the aggregate market
value of our outstanding common stock held by non-affiliates is
less than $75 million.
The perceived risk associated with the possible sale of a large
number of shares could cause some of our stockholders to sell
their stock, thus causing the price of our stock to decline. In
addition, actual or anticipated downward pressure on our stock
price due to actual or anticipated sales of stock could cause
some institutions or individuals to engage in short sales of our
common stock, which may itself cause the price of our stock to
decline.
If our stock price declines, we may be unable to raise
additional capital. A sustained inability to raise capital could
force us to go out of business. Significant declines in the
price of our common stock could also impair our ability to
attract and retain qualified employees, reduce the liquidity of
our common stock and result in the delisting of our common stock
from the NASDAQ Capital Market.
We expect the level of our future operating expenses to be
driven by the needs of our product development programs offset
by the availability of funds through partner-funded
collaborations, equity offerings or other financing activities.
The size, scope and pace of our development activities depend on
the availability of these resources. Our future cash
requirements will depend on many factors, including:
| the rate and extent of scientific progress in our research and development programs; | |
| the timing, costs and scope of, and our success in clinical trials, obtaining regulatory approvals and filing, prosecuting and enforcing patents; | |
| competing technological and market developments; | |
| the timing and costs of, and our success in any product commercialization activities and facility expansions, if and as required; and | |
| the expense and outcome of any litigation or administrative proceedings involving our intellectual property, or access to third party intellectual property through licensing agreements. |
Depending on our ability to successfully access additional
funding, we may be forced to implement additional cost reduction
measures, such as the reduction in force we implemented in
January 2006. Further adjustments may include scaling back or
delaying our inflammatory arthritis development program,
additional staff reductions, scaling back our intellectual
property prosecution, subleasing portions of our lab facilities,
curtailing capital expenditures or reducing other operating
activities. We may also be required to relinquish some rights to
our technology or product candidates or grant licenses on
unfavorable terms, either of which would reduce the ultimate
value to us of the technology or product candidates.
37
Table of Contents
OFF-BALANCE SHEET ARRANGEMENTS
Although we do not have any joint ventures or other similar
off-balance sheet items, in the ordinary course of business we
enter into agreements that require us to indemnify
counterparties against third-party claims. These may include:
agreements with vendors and suppliers, under which we may
indemnify them against claims arising from our use of their
products or services; agreements with clinical investigators,
under which we may indemnify them against claims arising from
their use of our product candidates; real estate and equipment
leases, under which we may indemnify lessors against third-party
claims relating to use of their property; agreements with
licensees or licensors, under which we may indemnify the
licensee or licensor against claims arising from their use of
our intellectual property or our use of their intellectual
property; and agreements with initial purchasers and
underwriters of our securities, under which we may indemnify
them against claims relating to their participation in the
transactions.
The nature and terms of these indemnifications vary from
contract to contract, and generally a maximum obligation is not
stated. Because we are unable to estimate our potential
obligation, and because management does not expect these
indemnifications to have a material adverse effect on our
consolidated financial position, results of operations or cash
flows, no related liabilities are recorded at December 31,
2005 or 2004. We hold insurance policies that mitigate potential
losses arising from certain indemnifications and, historically,
we have not incurred significant costs related to performance
under these obligations.
IMPACT OF NEW ACCOUNTING PRONOUNCEMENTS
In December 2004, the FASB issued SFAS No. 123R
Share Based Payment. This statement is a
revision to SFAS No. 123, supersedes APB No. 25,
Accounting for Stock Issued to Employees, and
amends SFAS No. 95, Statement of Cash
Flows. This statement will require us to expense the
cost of employee services received in exchange for an award of
equity instruments. This statement also provides guidance on
valuing and expensing these awards, as well as disclosure
requirements, and is effective for us beginning January 1,
2006.
As permitted by SFAS No. 123, we currently account for
share-based payments to employees using the APB No. 25
intrinsic value method and recognize no compensation cost for
employee stock options. At this time, we cannot predict the
impact of the adoption of SFAS No. 123R because the
impact will depend on levels of share-based payments granted in
the future. However, valuation of employee stock options under
SFAS No. 123R is similar to SFAS No. 123,
with minor exceptions. For information about what our reported
results of operations and earnings per share would have been had
we adopted SFAS No. 123, see the discussion under the
heading Stock Compensation in Note 1 of the
notes to our consolidated financial statements. The adoption of
SFAS No. 123Rs fair value method will have a
significant impact on our results of operations, although it
will have no impact on our overall financial position. We will
adopt the modified prospective method to implement the new
accounting standard.
Item 7A. | Quantitative and Qualitative Disclosures About Market Risk |
Items with interest rate risk: |
| Short term investments: Because of the short-term nature of our investments, we believe that our exposure to market rate fluctuations on our investments is minimal. Currently, we do not use any derivative or other financial instruments or derivative commodity instruments to hedge any market risks and do not plan to employ these instruments in the future. At December 31, 2005, we held $14.1 million in cash and cash equivalents, which are primarily invested in money market funds and denominated in U.S. dollars. An analysis of the impact on these securities of a hypothetical 10% change in short-term interest rates from those in effect at December 31, 2005, indicates that such a change in interest rates would not have a significant impact on our financial position or on our expected results of operations in 2006. | |
| Notes payable: Our results of operations are affected by changes in short-term interest rates as a result of a loan from Biogen which contains a variable interest rate. Interest payments on this loan are |
38
Table of Contents
established quarterly based upon the one-year London Interbank Offered Rate, or LIBOR, plus 1%. As of December 31, 2005, we were accruing interest on the note at a rate of 5.68%. The carrying amount of the note payable approximates fair value because the interest rate on this instrument changes with, or approximates, market rates. The following table provides information as of December 31, 2005, about our obligations that are sensitive to changes in interest rate fluctuations: |
Expected Maturity Date | ||||||||||||||||
2006 | 2007 | 2008 | 2009 | 2010 | Total | |||||||||||
Variable rate note
|
$ | | $2,500,000 | $2,500,000 | $2,500,000 | $ | | $7,500,000 |
Items with market and foreign currency exchange risk: |
| Investment in Chromos Molecular Systems, Inc.: At December 31, 2005, we held 2.7 million shares of Chromos Molecular Systems, Chromos, common shares with a market value of $0.16 per common share denominated in Canadian dollars. As of December 31, 2005 the Canadian to US exchange rate was US $0.8580 per CA $1.00. As of December 31, 2005, the investment is recorded at $376,000 with a $21,000 unrealized loss and is classified within prepaid expenses and other. We hold these shares of common stock as available-for-sale securities as we periodically sell them on the Toronto Stock Exchange. As a result of selling 370,000 shares of Chromos stock in 2005, we recorded $32,000 of net realized losses and received $57,000 in cash. The amount of potential realizable value in this investment will be determined by the market, the exchange rate between the Canadian and US dollar and our ability to sell the shares in the open market. |
39
Table of Contents
Item 8. | Financial Statements and Supplementary Data. |
Page | ||||
41 | ||||
42 | ||||
43 | ||||
44 | ||||
45 | ||||
46 |
40
Table of Contents
REPORT OF INDEPENDENT REGISTERED PUBLIC ACCOUNTING FIRM
The Board of Directors and Shareholders of Targeted Genetics
Corporation
We have audited the accompanying consolidated balance sheets of
Targeted Genetics Corporation as of December 31, 2005 and
2004, and the related consolidated statements of operations,
preferred stock and shareholders equity, and cash flows
for each of the three years in the period ended
December 31, 2005. These financial statements are the
responsibility of the Companys management. Our
responsibility is to express an opinion on these financial
statements based on our audits.
We conducted our audits in accordance with the standards of the
Public Company Accounting Oversight Board (United States). Those
standards require that we plan and perform the audit to obtain
reasonable assurance about whether the financial statements are
free of material misstatement. An audit includes examining, on a
test basis, evidence supporting the amounts and disclosures in
the financial statements. An audit also includes assessing the
accounting principles used and significant estimates made by
management, as well as evaluating the overall financial
statement presentation. We believe that our audits provide a
reasonable basis for our opinion.
In our opinion, the financial statements referred to above
present fairly, in all material respects, the consolidated
financial position of Targeted Genetics Corporation at
December 31, 2005 and 2004, and the consolidated results of
its operations and its cash flows for each of the three years in
the period ended December 31, 2005, in conformity with
U.S. generally accepted accounting principles.
We also have audited, in accordance with the standards of the
Public Company Accounting Oversight Board (United States), the
effectiveness of Targeted Genetics Corporations internal
control over financial reporting as of December 31, 2005,
based on criteria established in Internal Control-Integrated
Framework issued by the Committee of Sponsoring Organizations of
the Treadway Commission and our report dated March 14, 2006
expressed an unqualified opinion thereon.
/s/ Ernst & Young LLP |
Seattle, Washington
March 14, 2006
41
Table of Contents
TARGETED GENETICS CORPORATION
CONSOLIDATED BALANCE SHEETS
December 31, | |||||||||||
2005 | 2004 | ||||||||||
ASSETS | |||||||||||
Current assets:
|
|||||||||||
Cash and cash equivalents
|
$ | 14,122,000 | $ | 34,096,000 | |||||||
Accounts receivable
|
380,000 | 404,000 | |||||||||
Prepaid expenses and other
|
683,000 | 653,000 | |||||||||
Total current assets
|
15,185,000 | 35,153,000 | |||||||||
Property and equipment, net
|
1,747,000 | 2,495,000 | |||||||||
Goodwill
|
31,649,000 | 31,649,000 | |||||||||
Other assets
|
217,000 | 668,000 | |||||||||
Total assets
|
$ | 48,798,000 | $ | 69,965,000 | |||||||
LIABILITIES AND SHAREHOLDERS EQUITY | |||||||||||
Current liabilities:
|
|||||||||||
Accounts payable and accrued expenses
|
$ | 1,770,000 | $ | 1,436,000 | |||||||
Accrued employee expenses
|
587,000 | 1,030,000 | |||||||||
Current portion of accrued restructure charges
|
1,838,000 | 407,000 | |||||||||
Deferred revenue
|
278,000 | | |||||||||
Current portion of long-term obligations
|
155,000 | 1,122,000 | |||||||||
Total current liabilities
|
4,628,000 | 3,995,000 | |||||||||
Accrued restructure charges and deferred rent
|
5,422,000 | 6,026,000 | |||||||||
Long-term obligations
|
8,177,000 | 10,182,000 | |||||||||
Commitments and Contingencies (Notes 2 and 6)
|
|||||||||||
Shareholders equity:
|
|||||||||||
Preferred stock, $0.01 par value, 6,000,000 shares
authorized:
|
|||||||||||
Series A preferred stock, 800,000 shares designated,
none issued and outstanding
|
| | |||||||||
Common stock, $0.01 par value, 180,000,000 shares
authorized, 85,694,244 shares issued and outstanding at
December 31, 2005 and 120,000,000 shares authorized,
85,626,326 shares issued and outstanding at
December 31, 2004
|
857,000 | 856,000 | |||||||||
Additional paid-in capital
|
279,772,000 | 279,745,000 | |||||||||
Accumulated deficit
|
(250,037,000 | ) | (230,839,000 | ) | |||||||
Accumulated other comprehensive loss
|
(21,000 | ) | | ||||||||
Total shareholders equity
|
30,571,000 | 49,762,000 | |||||||||
Total liabilities and shareholders equity
|
$ | 48,798,000 | $ | 69,965,000 | |||||||
See accompanying notes to consolidated financial statements
42
Table of Contents
TARGETED GENETICS CORPORATION
CONSOLIDATED STATEMENTS OF OPERATIONS
Year Ended December 31, | ||||||||||||||
2005 | 2004 | 2003 | ||||||||||||
Revenue:
|
||||||||||||||
Collaborative revenue
|
$ | 6,874,000 | $ | 9,652,000 | $ | 14,073,000 | ||||||||
Operating expenses:
|
||||||||||||||
Research and development
|
18,199,000 | 17,288,000 | 17,197,000 | |||||||||||
General and administrative
|
6,313,000 | 6,650,000 | 5,490,000 | |||||||||||
Restructure charges
|
1,709,000 | 884,000 | 5,190,000 | |||||||||||
Total operating expenses
|
26,221,000 | 24,822,000 | 27,877,000 | |||||||||||
Loss from operations
|
(19,347,000 | ) | (15,170,000 | ) | (13,804,000 | ) | ||||||||
Investment income
|
661,000 | 383,000 | 183,000 | |||||||||||
Interest expense
|
(512,000 | ) | (476,000 | ) | (1,212,000 | ) | ||||||||
Gain on sale of majority-owned subsidiary
|
| 1,006,000 | | |||||||||||
Net loss
|
$ | (19,198,000 | ) | $ | (14,257,000 | ) | $ | (14,833,000 | ) | |||||
Net loss per common share (basic and diluted)
|
$ | (0.22 | ) | $ | (0.18 | ) | $ | (0.26 | ) | |||||
Shares used in computation of basic and diluted net loss per
common share
|
85,635,000 | 79,451,000 | 57,486,000 | |||||||||||
See accompanying notes to consolidated financial statements
43
Table of Contents
TARGETED GENETICS CORPORATION
CONSOLIDATED STATEMENTS OF PREFERRED STOCK
AND SHAREHOLDERS EQUITY
Series B | Accumulated | ||||||||||||||||||||||||||||||||
Preferred Stock | Common Stock | Other | Total | ||||||||||||||||||||||||||||||
Additional | Accumulated | Comprehensive | Shareholders | ||||||||||||||||||||||||||||||
Shares | Amount | Shares | Amount | Paid-In-Capital | Deficit | Loss | Equity | ||||||||||||||||||||||||||
Balance at December 31, 2002
|
12,015 | $ | 12,015,000 | 50,566,348 | $ | 506,000 | $ | 207,139,000 | $ | (201,749,000 | ) | $ | | $ | 5,896,000 | ||||||||||||||||||
Net loss and comprehensive loss 2003
|
| | | | | (14,833,000 | ) | | (14,833,000 | ) | |||||||||||||||||||||||
Reclassification of Series B convertible preferred stock
|
| (12,015,000 | ) | | | 12,015,000 | | | 12,015,000 | ||||||||||||||||||||||||
Issuance of shares for cash, net of issue costs of $1,387,000
|
| | 7,777,778 | 78,000 | 16,037,000 | | | 16,115,000 | |||||||||||||||||||||||||
Issuance of shares to Biogen for cash, net of issue costs of
$2,000
|
| | 2,515,843 | 25,000 | 4,768,000 | | | 4,793,000 | |||||||||||||||||||||||||
Issuance of shares to Elan for debt conversion
|
| | 5,203,244 | 52,000 | 9,315,000 | | | 9,367,000 | |||||||||||||||||||||||||
Exercise of stock options
|
| | 143,017 | 1,000 | 125,000 | | | 126,000 | |||||||||||||||||||||||||
Balance at December 31, 2003
|
12,015 | | 66,206,230 | 662,000 | 249,399,000 | (216,582,000 | ) | | 33,479,000 | ||||||||||||||||||||||||
Net loss and comprehensive loss 2004
|
| | | | | (14,257,000 | ) | | (14,257,000 | ) | |||||||||||||||||||||||
Conversion of Series B convertible preferred stock
|
(12,015 | ) | | 4,330,000 | 43,000 | (43,000 | ) | | | | |||||||||||||||||||||||
Issuance of shares for cash, net of issue costs of $1,742,000
|
| | 10,854,257 | 109,000 | 23,657,000 | | | 23,766,000 | |||||||||||||||||||||||||
Issuance of shares for cash, net of issue costs of $28,000
|
| | 3,954,132 | 39,000 | 5,933,000 | | | 5,972,000 | |||||||||||||||||||||||||
Issuance of shares to acquire minority interest in
majority-owned subsidiary
|
| | 158,764 | 2,000 | 748,000 | | | 750,000 | |||||||||||||||||||||||||
Exercise of stock options
|
| | 122,943 | 1,000 | 51,000 | | | 52,000 | |||||||||||||||||||||||||
Balance at December 31, 2004
|
| | 85,626,326 | 856,000 | 279,745,000 | (230,839,000 | ) | | 49,762,000 | ||||||||||||||||||||||||
Net loss 2005
|
| | | | | (19,198,000 | ) | | (19,198,000 | ) | |||||||||||||||||||||||
Unrealized loss on available-for-sale securities
|
| | | | | | (21,000 | ) | (21,000 | ) | |||||||||||||||||||||||
Comprehensive net loss 2005
|
| | | | | | (21,000 | ) | (19,219,000 | ) | |||||||||||||||||||||||
Exercise of stock options
|
| | 67,918 | 1,000 | 27,000 | | | 28,000 | |||||||||||||||||||||||||
Balance at December 31, 2005
|
| $ | | 85,694,244 | $ | 857,000 | $ | 279,772,000 | $ | (250,037,000 | ) | $ | (21,000 | ) | $ | 30,571,000 | |||||||||||||||||
See accompanying notes to consolidated financial statements
44
Table of Contents
TARGETED GENETICS CORPORATION
CONSOLIDATED STATEMENTS OF CASH FLOWS
Year Ended December 31, | |||||||||||||||
2005 | 2004 | 2003 | |||||||||||||
Operating activities:
|
|||||||||||||||
Net loss
|
$ | (19,198,000 | ) | $ | (14,257,000 | ) | $ | (14,833,000 | ) | ||||||
Adjustments to reconcile net loss to net cash used in operating
activities:
|
|||||||||||||||
Gain on sale of majority-owned subsidiary
|
| (1,006,000 | ) | | |||||||||||
Depreciation and amortization
|
1,319,000 | 1,289,000 | 2,420,000 | ||||||||||||
Non cash interest expense
|
27,000 | 63,000 | 822,000 | ||||||||||||
Gain on investments
|
(1,000 | ) | | | |||||||||||
Loss (gain) on sale/disposal of fixed assets
|
98,000 | (51,000 | ) | | |||||||||||
Changes in assets and liabilities:
|
|||||||||||||||
Decrease (increase) in accounts receivable
|
24,000 | (155,000 | ) | 1,004,000 | |||||||||||
Decrease (increase) in prepaid expenses and other
|
(107,000 | ) | 104,000 | 43,000 | |||||||||||
Decrease in other assets
|
451,000 | 341,000 | 348,000 | ||||||||||||
Increase (decrease) in current liabilities
|
(109,000 | ) | (193,000 | ) | 394,000 | ||||||||||
Increase (decrease) in deferred revenue
|
278,000 | (1,180,000 | ) | (4,861,000 | ) | ||||||||||
Increase (decrease) in accrued restructure charges and deferred
rent
|
827,000 | (478,000 | ) | 3,433,000 | |||||||||||
Net cash used in operating activities
|
(16,391,000 | ) | (15,523,000 | ) | (11,230,000 | ) | |||||||||
Investing activities:
|
|||||||||||||||
Purchases of property and equipment
|
(669,000 | ) | (408,000 | ) | (316,000 | ) | |||||||||
Proceeds from sales of investments
|
57,000 | | | ||||||||||||
Net cash used in investing activities
|
(612,000 | ) | (408,000 | ) | (316,000 | ) | |||||||||
Financing activities:
|
|||||||||||||||
Net proceeds from sales of capital stock
|
28,000 | 29,790,000 | 21,028,000 | ||||||||||||
Repayment of debt
|
(2,500,000 | ) | | | |||||||||||
Proceeds from equipment financing arrangements
|
| 46,000 | 229,000 | ||||||||||||
Payments under equipment financing arrangements
|
(499,000 | ) | (866,000 | ) | (1,260,000 | ) | |||||||||
Net cash provided by (used in) financing activities
|
(2,971,000 | ) | 28,970,000 | 19,997,000 | |||||||||||
Net increase (decrease) in cash and cash equivalents
|
(19,974,000 | ) | 13,039,000 | 8,451,000 | |||||||||||
Cash and cash equivalents, beginning of year
|
34,096,000 | 21,057,000 | 12,606,000 | ||||||||||||
Cash and cash equivalents, end of year
|
$ | 14,122,000 | $ | 34,096,000 | $ | 21,057,000 | |||||||||
Supplemental information:
|
|||||||||||||||
Cash paid for interest
|
$ | 441,000 | $ | 413,000 | $ | 459,000 |
See accompanying notes to consolidated financial statements
45
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL STATEMENTS
1. | Description of Business and Summary of Significant Accounting Policies |
Description of Business |
Targeted Genetics was incorporated in the state of Washington in
March 1989. We conduct research and development of gene therapy
products and technologies for treating both acquired and
inherited diseases. We develop these programs on our own and
under various collaborative agreements with others.
Basis of Presentation |
Our consolidated financial statements include the accounts of
Targeted Genetics, our wholly owned subsidiaries Genovo, Inc.
(inactive) and TGCF Manufacturing Corporation
(inactive), and until its sale in July 2004, our
majority-owned subsidiary, CellExSys, Inc., or CellExSys. All
significant intercompany transactions have been eliminated in
consolidation.
Cash Equivalents |
Cash equivalents include short-term investments that have a
maturity at the time of purchase of three months or less, are
readily convertible into cash and we believe have an
insignificant level of valuation risk attributable to potential
changes in interest rates. Our cash equivalents are recorded at
cost, which approximates fair market value, and consist
primarily of money market accounts and shares in a limited-term
bond fund.
Fair Value of Financial Instruments |
We believe that the carrying amounts of financial instruments
such as cash and cash equivalents, available-for-sale
securities, accounts receivable and accounts payable approximate
fair value because of the short-term nature of these items. We
believe that the carrying amounts of the notes payable and
equipment financing obligations approximate fair value because
the interest rates on these instruments change with, or
approximate, market interest rates.
Property and Equipment |
Property and equipment is stated at cost less accumulated
depreciation. We compute depreciation of property and equipment
using the straight-line method over the assets estimated
useful life. The useful lives of our furniture and equipment
ranges from three to ten years, and our leasehold improvements
are amortized over the shorter of the assets estimated
useful life or the remainder of the lease term. Our leasehold
improvements are currently being amortized over useful lives
which range from four to seven years. Depreciation and
amortization expense was $1.3 million in 2005 and 2004 and
$2.4 million in 2003. Depreciation expense includes
depreciation of property and equipment purchased under capital
leases.
Goodwill and Purchased Intangibles |
In 2000, we acquired Genovo, Inc., a development-stage
biotechnology company, for a purchase price of
$66.4 million. We allocated the excess of the acquisition
cost over the fair value of the identifiable net assets acquired
to goodwill totaling $38.2 million and to other purchased
intangibles totaling $605,000. From 2000 through 2001, we
recorded amortization expenses of $7.1 million of goodwill
and purchased intangibles. We test goodwill for impairment at
least annually, and more frequently when events or circumstances
indicate the carrying value may be impaired, by comparing its
carrying value to the market value of our shares outstanding.
Events or circumstances which could trigger an impairment review
include a significant adverse change in our business climate,
significant changes in our use of acquired technology, and
changes to our overall business strategy. In the event that our
valuation tests show an impairment in the recorded value of our
goodwill, we may record a significant non-cash charge to
expense. We have performed annual impairment
46
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
tests as of October 1 each year since our January 1,
2002 implementation of SFAS No. 142 Goodwill
and Other Intangible Assets and concluded that no
impairment in the value of our goodwill has occurred.
Prepaid Expenses and Other |
Other assets consists primarily of the Chromos Inc., or Chromos,
securities that we received as payment on the debenture was and
as consideration for the sale of CellExSys in July 2004. In
accordance with SFAS No. 115, Accounting for
Certain Investments in Debt and Equity Securities,
these investment securities are classified as available-for-sale
and reported at fair value, with unrealized gains and losses
excluded from earnings and reported as accumulated other
comprehensive loss within stockholders equity. Upon the
sales of Chromos securities, realized gains and losses are
computed using the difference between the sales price and the
book value which is determined on a specific identification
basis. We periodically evaluate the investment securities for
other-than-temporary declines in value and record any losses
through an adjustment to earnings.
Other Assets |
In accordance with SFAS No. 144, Accounting
for the Impairment or Disposal of Long-Lived Assets,
we review the carrying value and fair value of long-lived
assets whenever events or changes in circumstances indicate that
there may be impairment in value. Conditions that would
necessitate an impairment assessment include a significant
decline in the observable market value of an asset, a
significant change in the extent or manner in which an asset is
used, or a significant adverse change that would indicate that
the carrying amount of an asset or group of assets is not
recoverable.
Accrued Restructure Charges |
We apply the provisions of SFAS No. 146,
Accounting for Costs Associated with Exit or Disposal
Activities, as it relates to our facilities in
Bothell, Washington and our former facility in Sharon Hill,
Pennsylvania. As a result, we have recorded restructuring
charges on the operating leases for these facilities. Accrued
restructuring charges, and in particular, those charges
associated with exiting a facility, are subject to many
assumptions and estimates. Under SFAS No. 146, an
accrued liability for lease termination costs is initially
measured at fair value, based on the remaining lease payments
due under the lease and other costs, reduced by sublease rental
income that could be reasonably obtained from the property, and
discounted using a credit-adjusted risk-free interest rate. We
use a risk-free annual interest rate of 10%. The assumptions as
to estimated sublease rental income, the period of time to
execute a sublease and the costs and concessions necessary to
enter into a sublease significantly impact the accrual and may
differ from what actually occurs. We review these estimates
periodically and adjust the accrual when necessary.
Series B Convertible Preferred Stock |
In July 1999, we issued shares of our Series B convertible
exchangeable preferred stock, valued at $12 million, to
Elan International Services Ltd., or Elan, in exchange for our
80.1% interest in Emerald Gene Systems, Ltd., or Emerald.
Emerald was our then majority-owned research and development
joint venture with Elan that was terminated during 2002 and
dissolved in January 2005. The Series B preferred stock and
accrued dividends were convertible at Elans option into
shares of our common stock, at a conversion price of
$3.32 per share. Compounding dividends accrued
semi-annually at 7% per year on the $1,000 per share
face value of the preferred stock. Dividends were not paid in
cash, but rather resulted in an increase to the number of shares
of common stock issued upon conversion.
Elan was entitled to exchange the Series B preferred stock
for all shares of preferred stock that we held in Emerald until
this exchange right expired in April 2003. Prior to the
expiration of the exchange right, the carrying value of the
Series B preferred stock was reflected as mezzanine equity
in our financial statements.
47
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Upon expiration of the exchange right, we reclassified the
Series B preferred stock from mezzanine equity to
shareholders equity. Elan converted the Series B
preferred stock into 4,330,000 shares of common stock in
March 2004.
Stock Compensation |
As permitted by the provisions of SFAS No. 123,
Accounting for Stock-Based Compensation, we
have elected to follow Accounting Principles Board, or APB,
No. 25, Accounting for Stock Issued to
Employees, which uses the intrinsic value method and
generally results in the recognition of no compensation cost for
employee stock option grants when priced at the fair value per
share on the date of grant. We do not recognize any compensation
expense for options granted to employees or directors because we
grant all options at fair market value on the date of grant. The
adoption of SFAS No. 123R, Share-Based
Payments (a revision of SFAS No. 123) on
January 1, 2006 will require us to record expense stock
option grants.
If we had elected to recognize compensation expense based on the
fair market value at the grant dates for the stock options
granted, the pro forma net loss and net loss per common share
would have been as follows:
Year Ended December 31, | |||||||||||||
2005 | 2004 | 2003 | |||||||||||
Net loss:
|
|||||||||||||
As reported
|
$ | (19,198,000 | ) | $ | (14,257,000 | ) | $ | (14,833,000 | ) | ||||
Add: Stock-based compensation under SFAS 123
|
(1,372,000 | ) | (1,564,000 | ) | (780,000 | ) | |||||||
Pro forma
|
$ | (20,570,000 | ) | $ | (15,821,000 | ) | $ | (15,613,000 | ) | ||||
Basic net loss per share:
|
|||||||||||||
As reported
|
$ | (0.22 | ) | $ | (0.18 | ) | $ | (0.26 | ) | ||||
Pro forma
|
(0.24 | ) | (0.20 | ) | (0.27 | ) |
Revenue Recognition under Collaborative Agreements |
We generate revenue from technology licenses, collaborative
research arrangements and agreements to provide research,
development and manufacturing services. Revenue under technology
licenses and collaborative agreements typically consists of
nonrefundable, up-front license fees, collaborative research
funding, technology access fees and various other payments.
Revenue from nonrefundable, up-front license fees and technology
access payments is initially deferred and then recognized
systematically over the service period of the collaborative
agreement, which is often the development period. Revenue
associated with performance milestones is recognized as earned,
based upon the achievement of the milestones defined in the
applicable agreements. Revenue under research and development
contracts is recognized as the related costs are incurred.
Payments received in excess of amounts earned are classified as
deferred revenue in the accompanying consolidated balance sheets.
Significant Revenue Relationships and Concentration of Risk |
Our primary source of revenue is from our collaborative
agreements. During 2005 and 2004, revenues under our
collaboration with the International AIDS Vaccine Initiative, or
IAVI, accounted for a substantial portion our revenue. Revenue
in 2003 included revenue from our collaboration with IAVI and
also included revenues from our collaborations with Biogen and
Wyeth. For 2006 we expect to earn significant revenue from our
three primary collaborators: International AIDS Vaccine
Initiative, or IAVI, Celladon Corporation, or Celladon, and
National Institute of Allergy and Infectious Diseases, or NIAID.
During 2005, we were named
48
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
as a subcontractor to receive up to $18 million of a five
year, $22 million NIAID contract with Columbus
Childrens Research Institute and The Childrens
Hospital of Philadelphia to develop AAV-based HIV/AIDS vaccine
for use in the developed world. A significant change in the
level of work or timing of work activities and the funding
received from any of these collaborations could disrupt our
business and adversely affect our cash flow and results of
operations.
Research and Development Costs |
Research and development costs include salaries, costs of
outside collaborators and outside services, clinical trial
expenses, royalty and license costs and allocated facility,
occupancy and utility expenses. We expense research and
development costs as incurred. Costs and expenses related to
programs conducted under collaborative agreements that result in
collaborative revenue totaled approximately $8.2 million in
2005, $7.1 million in 2004 and in 2003.
Operating Leases |
We have operating leases for our office space and lab facilities
located in Seattle and Bothell, Washington. These lease
agreements contain rent escalation clauses and rent holidays.
For scheduled rent escalation clauses during the lease terms or
for rental payments commencing at a date other than the date of
initial occupancy, we record minimum rental expenses on a
straight-line basis over the terms of the leases in the
consolidated statement of operations. When we make leasehold
improvements to our facilities, we amortize them over the
shorter of the useful life or the remaining term of the lease.
We currently have $689,000 of leasehold improvements related to
our Seattle facilities that we are amortizing over the remainder
of the current lease terms, which expire near the end of the
first quarter of 2009.
Net Loss per Common Share |
Net loss per common share is based on net loss divided by the
weighted average number of common shares outstanding during the
period. Our diluted net loss per share is the same as our basic
net loss per share because all stock options, warrants and other
potentially dilutive securities are antidilutive and therefore
excluded from the calculation of diluted net loss per share. The
total number of shares that we excluded from the calculations of
net loss per share were 9,040,870 shares in 2005,
8,093,058 shares in 2004 and 10,867,013 shares in 2003.
Use of Estimates |
The preparation of financial statements in conformity with
U.S. generally accepted accounting principles requires
management to make estimates and assumptions that affect the
amounts reported in the financial statements and accompanying
notes. Our actual results may differ from those estimates.
Recently Issued Accounting Standards |
In December 2004, the FASB issued SFAS No. 123R
Share Based Payment. This statement is a
revision to SFAS No. 123, supersedes APB No. 25,
Accounting for Stock Issued to Employees, and
amends SFAS No. 95, Statement of Cash
Flows. This statement will require us to expense the
cost of employee services received in exchange for awards of
equity instruments. This statement also provides guidance on
valuing and expensing these awards, as well as disclosure
requirements, and is effective for us as of January 1, 2006.
SFAS No. 123R permits public companies to choose
between the following two adoption methods:
1. A modified prospective method in which compensation cost is recognized beginning with the effective date (a) based on the requirements of SFAS No. 123R for all share-based payments granted |
49
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
after the effective date and (b) based on the requirements of SFAS No. 123 for all awards granted to employees prior to the effective date of SFAS No. 123R that remain unvested on the effective date, or | |
2. A modified retrospective method which includes the requirements of the modified prospective method described above, but also permits entities to restate based on the amounts previously recognized under SFAS No. 123 for purposes of pro forma disclosures either (a) all prior periods presented or (b) prior interim periods of the year of adoption. |
As permitted by SFAS No. 123, we currently account for
share-based payments to employees using the APB No. 25
intrinsic value method and recognize no compensation cost for
employee stock options. At this time, we cannot predict the
impact of the adoption of SFAS No. 123R because the
impact will depend on levels of share-based payments granted in
the future. However, valuation of employee stock options under
SFAS No. 123R is similar to SFAS No. 123,
with minor exceptions. For information about what our reported
results of operations and earnings per share would have been had
we adopted SFAS No. 123, see the discussion under the
heading Stock Compensation in this note. The
adoption of SFAS No. 123Rs fair value method
will have a significant impact on our results of operations,
although it will have no impact on our overall financial
position. We will adopt the modified prospective method to
implement the new accounting standard.
Reclassifications |
Certain reclassifications have been made to conform prior year
classifications to the current year presentation.
2. | Liquidity and Managements Plans |
We have prepared the accompanying financial statements on a
going concern basis, which assumes that we will realize our
assets and satisfy our liabilities in the normal course of
business. The accompanying financial statements do not include
any adjustments that might be required should we be unable to
recover the value of our assets or satisfy our liabilities.
As reflected in the accompanying consolidated financial
statements, we have incurred net losses since our inception and
we have an accumulated deficit of $250.0 million as of
December 31, 2005. Our cash balance as of December 31,
2005 was $14.1 million and our accounts receivable balance
was $380,000. As described Note 13 of the notes to our
consolidated financial statements on March 10, 2006 we
raised $4.8 million of working capital through the sale of
common stock. We project 2006 net operating cash flow
deficits of approximately $13 million to $16 million.
This projection assumes that we complete all of the planned
development activities for each of our funded projects,
resulting in approximately $9 million of 2006 funding from
our collaborative partners, and assumes that we achieve our
current operating plan, which includes achieving cost reductions
and cash flow benefits of $2 million in 2006 from reducing
our patent expenses, paying certain discretionary expenses in
stock rather than cash and reductions in compensation expenses.
We believe that our current resources, combined with the capital
raised in March 2006, are sufficient to fund our planned
operations, including our clinical trials, into the first
quarter of 2007. We also intend to seek additional financing in
order to fund our operations further into or through 2007;
however, we can not provide assurances that we will be
successful in obtaining additional financing when and as needed
in the future.
50
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
3. | Property and Equipment |
Property and equipment consisted of the following:
December 31, | ||||||||
2005 | 2004 | |||||||
Furniture and equipment
|
$ | 6,783,000 | $ | 6,410,000 | ||||
Leasehold improvements
|
9,756,000 | 9,739,000 | ||||||
16,539,000 | 16,149,000 | |||||||
Less accumulated depreciation and amortization
|
(14,792,000 | ) | (13,654,000 | ) | ||||
$ | 1,747,000 | $ | 2,495,000 | |||||
We finance a portion of our equipment through equipment
financing arrangements, which include extensions and purchase
options, and require us to pledge the equipment as security for
the financing. The cost of equipment that has been pledged under
financing arrangements totaled $1.0 million at
December 31, 2005 and $2.6 million at
December 31, 2004.
4. | Restructure Charges |
In December 2002, we began to pursue options to sublease, or
terminate, our lease on the Bothell facility and in February
2003, we closed our facility in Sharon Hill, Pennsylvania that
we acquired when we acquired Genovo, Inc. We record accrued
restructure charges as they relate to the leases on these
facilities. Accrued restructure charges represent our best
estimate of the fair value of the liability remaining under the
lease and are computed as the present value of the difference
between the remaining lease payments due less the net of
sublease income and expense. These assumptions are periodically
reviewed and adjustments are made to the accrued restructure
charge when necessary. We record accretion expense based upon
changes in the accrued restructure liability that result from
the passage of time and the assumed discount rate of 10%.
Accretion expense is recorded on an ongoing basis through the
end of the lease term in September 2015 and is reflected as a
restructuring charge in the accompanying consolidated statements
of operations.
The tables below present our total estimated restructure charges
and a reconciliation of the associated liability:
Employee | Contract | Other | |||||||||||||||
Termination | Termination | Associated | |||||||||||||||
Benefits | Costs | Costs | Total | ||||||||||||||
Incurred in 2002
|
$ | 725,000 | $ | 1,602,000 | $ | | $ | 2,327,000 | |||||||||
Incurred in 2003
|
5,000 | 5,153,000 | 32,000 | 5,190,000 | |||||||||||||
Incurred in 2004
|
| 884,000 | | 884,000 | |||||||||||||
Incurred in 2005
|
| 1,709,000 | | 1,709,000 | |||||||||||||
Cumulative incurred to date
|
730,000 | 9,348,000 | 32,000 | 10,110,000 | |||||||||||||
Estimated future charges
|
| 7,329,000 | | 7,329,000 | |||||||||||||
Total expected to be incurred
|
$ | 730,000 | $ | 16,677,000 | $ | 32,000 | $ | 17,439,000 | |||||||||
51
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Contract | |||||
Termination | |||||
Costs | |||||
December 31, 2004 accrued liability
|
$ | 6,348,000 | |||
Charges related to changes in assumptions
|
1,132,000 | ||||
Accretion expense
|
577,000 | ||||
Amount paid in 2005
|
(908,000 | ) | |||
December 31, 2005 accrued liability
|
$ | 7,149,000 | |||
For the year ended December 31, 2005, we recorded
additional restructure charges of $1.1 million as a result
of updating our estimates of costs and sublease income
associated with exiting the Bothell facility. During the year,
we changed real estate brokers and based upon their consultation
and advice, we modified our approach to subleasing our Bothell
facility and altered our marketing strategy to market the full
facility as available for sublease. Previously, we had intended
to sublease approximately one-third of the facility. Our current
expectations are that we will not use the Bothell facility for
our manufacturing needs. We recorded a $1.1 million charge
to reflect the changes in our assumptions related to:
| an increase in the square footage available for sublease and related increase in sublease income; | |
| an increase in the amount of time necessary to find a sublease tenant; | |
| an increase in broker commissions; and | |
| a decrease in estimated costs of tenant improvements. |
Adjustments to the accrued restructure liability for year also
reflect $577,000 of accretion expense for the period. The amount
paid during the year represents lease payments made to the
landlord for the portion of the lease we intended to sublease.
The total of these charges and adjustments to the liability are
reflected as restructure charges in the accompanying
consolidated statements of operations.
We periodically evaluate our restructuring estimates and
assumptions and record additional restructure charges as
necessary. Because restructure charges are estimates based upon
assumptions regarding the timing and amounts of future events,
significant adjustments to the accrual may be necessary in the
future based on the actual outcome of events and as we become
aware of new facts and circumstances. If we decide to resume use
of the Bothell facility, any remaining accrued restructure
charges related to the facility will be reversed. This reversal
would be reflected as a reduction of restructuring expenses and
reflected in the period in which use is resumed. We are unable
to determine the likelihood of any future adjustments to our
accrued restructuring charges. We have a $200,000 certificate of
deposit recorded within other assets that is pledged as
collateral for the Bothell facility lease.
5. | Long-Term Obligations |
Long-term obligations consisted of the following:
December 31, | ||||||||
2005 | 2004 | |||||||
Loans payable to Biogen Idec
|
$ | 8,150,000 | $ | 10,624,000 | ||||
Equipment financing obligations
|
182,000 | 680,000 | ||||||
8,332,000 | 11,304,000 | |||||||
Less current portion
|
(155,000 | ) | (1,122,000 | ) | ||||
$ | 8,177,000 | $ | 10,182,000 | |||||
52
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Future aggregate principal payments related to long-term
obligations are $155,000 in 2006, $3.2 million in 2007,
$2.5 million in 2008 and in 2009 and zero in 2010.
We have two loans payable to Biogen Idec, a beneficial owner of
approximately 13.5% of our outstanding common shares. During
2001, we borrowed $10.0 million from Biogen Idec to fund
our general operations under the terms of an unsecured loan
agreement. Outstanding borrowings under this unsecured loan
agreement bear interest at the one-year London Interbank Offered
Rate, or LIBOR, plus 1%, which is reset quarterly. The loan
contains financial covenants establishing limits on our ability
to declare or pay cash dividends and it was originally scheduled
to mature in August 2006. The second loan is a promissory note
payable to Biogen Idec which we assumed in 2000 as part of our
acquisition of Genovo. This promissory note has an outstanding
principal amount of $650,000, bears no interest, and was
originally scheduled to mature in September 2005.
In September 2005, we modified the terms of our loans payable to
Biogen Idec. In connection with these modifications, we made a
$2.5 million cash payment to Biogen Idec on
September 1, 2005 to reduce the outstanding principal on
the $10.0 million loan to $7.5 million and agreed to
make scheduled payments of $2.5 million of principal plus
accrued interest on each of August 1, 2007, 2008 and 2009.
In addition, we have agreed to apply one-third of certain
up-front payments received from potential future corporate
collaborations to the outstanding balance on this loan payable,
first to repayment of any accrued and unpaid interest on the
principal being repaid, and second to the repayment of
outstanding principal in reverse order of maturity. In addition,
the maturity date of the $650,000 promissory note to Biogen Idec
was extended until August 1, 2007.
Equipment financing obligations relate to secured financing for
the purchase of capital equipment and leasehold improvements.
These obligations bear interest at rates ranging from 8.15% to
9.09% and mature from June 2006 to February 2008.
6. | Commitments |
We lease our research and office facilities in Seattle,
Washington under two non-cancelable operating leases. The lease
on our primary laboratory, manufacturing and office space
expires in April 2009 and contains an option to renew the lease
for a five-year period. The lease on our administrative office
space expires in March 2009, includes two options to extend the
lease for a total of five additional years and includes an
option to cancel the lease at any time between April 2006 and
March 2009 with certain early termination penalties. We lease a
facility in Bothell, Washington under a non-cancelable operating
lease that expires in September 2015, which was intended to
accommodate future manufacturing of our product candidates. We
have applied SFAS No. 146 as it relates to our Bothell
facility lease and have recorded accrued restructure charges of
$7.1 million as of December 31, 2005. This accrual
represents the present value of future lease payments, net of
assumed sublease payments. Future lease payments on our facility
in Bothell will reduce the amount of the accrued restructure
charges and are included in future minimum lease payments under
non-cancelable operating leases which are as follows:
Year Ending December 31, | |||||
2006
|
$ | 2,336,000 | |||
2007
|
2,364,000 | ||||
2008
|
2,392,000 | ||||
2009
|
1,622,000 | ||||
2010
|
1,431,000 | ||||
Thereafter
|
7,770,000 | ||||
Total minimum lease payments
|
$ | 17,915,000 | |||
53
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
We recognized rent expense under operating leases of
$1.4 million in 2005, $1.6 million in 2004 and
$1.7 million in 2003.
7. Investment
in Chromos Molecular Systems, Inc.
In July 2004, our majority-owned subsidiary, CellExSys Inc., was
acquired by and merged into Chromos Inc., a wholly-owned
subsidiary of Chromos Molecular Systems Inc., or Chromos.
Chromos is a publicly traded company whose common stock is
traded on the Toronto Stock Exchange. At the time of the sale,
we owned approximately 79% of CellExSys. As a result of the sale
of CellExSys, we received consideration of approximately
1.2 million shares of Chromos common stock and a 79% share
of any payments made by Chromos under an interest bearing
debenture issued by Chromos.
As a result of the sale of our share of CellExSys, we recorded a
gain in 2004 which was comprised of the following:
Deposits received from Chromos to fund pre-closing operating
costs
|
$ | 502,000 | ||
Estimated fair value of consideration received
|
453,000 | |||
Net liabilities assumed by Chromos
|
51,000 | |||
$ | 1,006,000 | |||
Chromos funded $502,000 of CellExSys operating costs
through the closing of the merger and assumed CellExSys
net liabilities as of the merger date of $51,000 consisting
primarily of trade and employee payables partially offset by an
employee note receivable. We estimated the fair value of the
stock and debenture we received based on several factors
including the market price and trading volume of Chromos common
stock and Chromos financial and business condition.
As of December 31, 2004, we valued our Chromos securities
and interest in the debenture at $453,000. Based upon our first
quarter 2005 assessment of Chromos financial condition we
recorded a $63,000 charge to investment income to reduce the
carrying value of the debenture to zero. In the second quarter
of 2005, Chromos stock price declined and we recorded an
$181,000 other-than-temporary impairment charge relating to the
decline in value of the Chromos securities. During the second
half of the year, Chromos issued us approximately
1.0 million shares of its common stock in connection with
the scheduled debt and interest payment under the debenture and
also later in 2005, Chromos issued us an additional
approximately 0.9 million shares of its common stock as an
early payment of the debenture. As the carrying value of the
debenture was zero, we recorded the market value of the shares
received as investment income. During 2005, we sold
370,000 shares of Chromos and recognized net losses of
$32,000. These securities are sold on a specific identification
method based on when we received the shares. This 2005 activity
nets to a $1,000 gain as follows:
2005 | ||||
Impact of Chromos securities activity on investment income
|
||||
Write down of debenture in the first quarter
|
$ | (63,000 | ) | |
Other-than-temporary loss on Chromos securities in the second
quarter
|
(181,000 | ) | ||
Gain on receipt of debenture payments in the third and fourth
quarters
|
277,000 | |||
Realized losses on sales of Chromos securities, net
|
(32,000 | ) | ||
Increase in investment income attributable to Chromos activity
|
$ | 1,000 | ||
As of December 31, 2005, we hold approximately
2.7 million shares of Chromos common stock with a fair
value of $376,000, which includes $21,000 of unrealized losses.
Our Chromos securities was in an unrealized loss position as of
December 31, 2005, but shortly after December 31, 2005
the stock price
54
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
increased over our carrying value. We will continue to record
our common stock investment in Chromos at fair market value and
record changes in the fair market value of this stock in
accumulated other comprehensive loss. We periodically evaluate
our Chromos stock for signs of impairment that may be
other-than-temporary which would necessitate a reduction in the
carrying value of the investment and charge to expense. One of
the members of our Board of Directors is also a member of the
Board of Directors of Chromos.
8. | Other Comprehensive Loss |
Comprehensive loss is the total of net loss and all other
non-owner changes in equity. Comprehensive loss includes
unrealized gains and losses from investments and foreign
currency translations, as presented in the following table:
December 31, | ||||||||
2005 | 2004 | |||||||
Net loss as reported
|
$ | (19,198,000 | ) | $ | (14,257,000 | ) | ||
Other comprehensive loss:
|
||||||||
Unrealized loss on available-for-sale securities
|
(37,000 | ) | | |||||
Foreign currency translation adjustment
|
16,000 | | ||||||
Other comprehensive loss
|
$ | (19,219,000 | ) | $ | (14,257,000 | ) | ||
9. | Shareholders Equity |
Stock Purchase Warrants |
In 1999, in connection with a technology license agreement, we
issued to Alkermes, Inc. a warrant to
purchase 1,000,000 shares of our common stock at an
exercise price of $2.50 per share, expiring in June 2007,
and a warrant to purchase 1,000,000 shares of our
common stock at an exercise price of $4.16 per share,
expiring in June 2009. Both of these warrants were outstanding
at December 31, 2005.
Shareholder Rights Plan |
In 1996, our Board of Directors adopted a shareholder rights
plan. Under our rights plan, each holder of a share of
outstanding common stock is also entitled to one preferred stock
purchase right. We adopted the rights plan to guard against
partial tender offers and other abusive tactics that might be
used in an attempt to gain control of Targeted Genetics without
paying all shareholders a fair price for their shares. The
rights plan will not prevent a change of control, but is
designed to deter coercive takeover tactics and to encourage
anyone attempting to acquire us to first negotiate with our
Board of Directors. Generally, if any person or group becomes
the beneficial owner of more than 15% of our outstanding common
stock (an acquiring person), then each preferred stock purchase
right not owned by the acquiring person or its affiliates would
entitle its holder to purchase a share of our common stock at a
50% discount, which would result in a significant dilution of
the acquiring persons interest in Targeted Genetics. If we
or 50% or more of our assets or earnings are thereafter
acquired, each right will entitle its holder to purchase a share
of common stock of the acquiring entity for a 50% discount.
The shareholder rights plan expires in October 2006. Our Board
of Directors will generally be entitled to redeem the rights for
$0.01 per right at any time before a person or group
acquires more than 15% of our common stock. In addition, at any
time after an acquiring person crosses the 15% threshold but
before it acquires us or 50% of our assets or earnings, the
Board may exchange all or part of the rights (other than those
held by the acquiring person) for one share of common stock per
right. Upon its expiration, we do not expect to renew this
shareholder rights plan.
55
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Stock Options |
We have various stock option plans (Option Plans) that provide
for the issuance of nonqualified and incentive stock options to
acquire up to 12,979,444 shares of our common stock. These
stock options may be granted by our Board of Directors to our
employees, directors and officers and to consultants, agents,
advisors and independent contractors who provide services to us.
The exercise price for incentive stock options shall not be less
than the fair market value of the shares on the date of grant.
Options granted under our Option Plans expire no later than ten
years from the date of grant and generally vest and become
exercisable over a four-year period following the date of grant.
However in 2003, we granted options to
purchase 655,000 shares of our common stock with
vesting periods which ranged from twelve to eighteen months.
Each year, every non-employee member of our Board of Directors
receives an annual nonqualified stock option grant to
purchase 20,000 shares, the chairman of our Board of
Directors receives an additional 10,000 nonqualified stock
options and the chairman of each of the audit, compensation or
nominating and corporate governance committees of our Board of
Directors each receive an additional 5,000 nonqualified stock
options. These options vest over a 12 month period provided
that they provide continued service to us.
The following table summarizes activity related to our Option
Plans:
Weighted | |||||||||||||
Average | |||||||||||||
Exercise | Options | ||||||||||||
Shares | Price | Exercisable | |||||||||||
Balance, December 31, 2002
|
4,439,407 | $ | 3.80 | 2,389,393 | |||||||||
Granted
|
1,060,250 | 0.54 | |||||||||||
Exercised
|
(143,017 | ) | 0.88 | ||||||||||
Expired
|
(4,400 | ) | 0.55 | ||||||||||
Forfeited
|
(1,254,553 | ) | 3.78 | ||||||||||
Balance, December 31, 2003
|
4,097,687 | 3.07 | 2,706,127 | ||||||||||
Granted
|
2,517,950 | 1.35 | |||||||||||
Exercised
|
(122,943 | ) | 0.43 | ||||||||||
Expired
|
(97,666 | ) | 4.81 | ||||||||||
Forfeited
|
(301,970 | ) | 3.52 | ||||||||||
Balance, December 31, 2004
|
6,093,058 | 2.36 | 3,416,598 | ||||||||||
Granted
|
1,528,200 | 0.88 | |||||||||||
Exercised
|
(67,918 | ) | 0.41 | ||||||||||
Expired
|
(84,501 | ) | 3.91 | ||||||||||
Forfeited
|
(427,969 | ) | 1.61 | ||||||||||
Balance, December 31, 2005
|
7,040,870 | 2.09 | 4,320,432 | ||||||||||
56
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
The following table summarizes information regarding our
outstanding and exercisable options at December 31, 2005:
Outstanding | Exercisable | ||||||||||||||||||||
Weighted | Weighted Average | Weighted | |||||||||||||||||||
Average | Remaining | Average | |||||||||||||||||||
Number of | Exercise | Contractual Life | Number of | Exercise | |||||||||||||||||
Range of Exercise Prices | Option Shares | Price | (Years) | Option Shares | Price | ||||||||||||||||
$0.29 - $ 0.49
|
572,117 | $ | 0.38 | 7.23 | 532,429 | $ | 0.37 | ||||||||||||||
0.60 - 0.84
|
677,604 | 0.75 | 8.01 | 311,534 | 0.78 | ||||||||||||||||
0.91 - 1.28
|
1,300,418 | 0.97 | 8.76 | 261,574 | 1.08 | ||||||||||||||||
1.31 - 1.31
|
1,947,200 | 1.31 | 8.38 | 837,071 | 1.31 | ||||||||||||||||
1.32 - 2.53
|
1,076,454 | 1.90 | 4.42 | 958,528 | 1.93 | ||||||||||||||||
2.57 - 14.88
|
1,467,077 | 5.53 | 4.78 | 1,419,296 | 5.63 | ||||||||||||||||
Balance, December 31, 2005
|
7,040,870 | 2.09 | 6.97 | 4,320,432 | 2.70 |
For purposes of our pro forma employee stock-based compensation
disclosures, we calculate the pro forma compensation costs for
stock options granted using the accelerated expense attribution
method and estimate the fair value of each option on the date of
grant using the Black-Scholes-Merton pricing model with the
following weighted average assumptions:
2005 | 2004 | 2003 | ||||||||||
Expected dividend rate
|
Nil | Nil | Nil | |||||||||
Expected stock price volatility range
|
1.09-1.12 | 1.12-1.47 | 1.47-1.51 | |||||||||
Risk-free interest rate range
|
3.63-4.36% | 2.71-4.47% | 1.62-4.19% | |||||||||
Expected life of options range
|
4-5 years | 4 years | 4 years | |||||||||
Weighted average fair value (per share) of options granted
|
$0.75 | $1.27 | $0.49 |
Reserved Shares |
As of December 31, 2005, we had reserved shares of our
common stock for future issuance as follows:
Stock options granted
|
7,040,870 | |||
Available for future stock option grants under Option Plans
|
2,993,541 | |||
Stock purchase warrants
|
2,000,000 | |||
Total shares reserved
|
12,034,411 | |||
57
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
10. | Collaborative and Other Agreements |
We have entered into various relationships with pharmaceutical
and biotechnology companies, a non-profit organization and with
the U.S. government through a contract with the NIAID to
develop our product candidates. Under these partnerships, we
typically are reimbursed for research and development activities
we perform and in certain cases we received milestone and
upfront payments. Revenues earned under our research and
development collaborations are as follows:
Year Ended December 31, | |||||||||||||
2005 | 2004 | 2003 | |||||||||||
IAVI
|
$ | 5,588,000 | $ | 8,340,000 | $ | 4,409,000 | |||||||
Celladon
|
777,000 | | | ||||||||||
Sirna
|
315,000 | | | ||||||||||
Former collaborations:
|
|||||||||||||
Biogen
|
| | 5,112,000 | ||||||||||
Wyeth
|
| | 3,894,000 | ||||||||||
Other
|
194,000 | 1,312,000 | 658,000 | ||||||||||
$ | 6,874,000 | $ | 9,652,000 | $ | 14,073,000 | ||||||||
International AIDS Vaccine Initiative Agreement |
In 2000, we entered into a three-year development collaboration
with IAVI and Columbus Childrens Research Institute at
Childrens Hospital in Columbus, Ohio to develop a vaccine
to protect against the progression of HIV infection to AIDS. In
2004, this collaboration was extended through December 2006.
Under the terms of the collaboration, IAVI provides funding to
us to support development, preclinical studies and manufacturing
of product for clinical trials on a cost reimbursement basis.
IAVI independently monitors and funds clinical development costs
under the collaboration.
Under the terms of the IAVI agreement, we have retained
exclusive rights to commercialize any product that results from
the collaboration in developed countries, and we have agreed to
manufacture vaccines that result from the collaboration for IAVI
to distribute in developing countries. If we decline, or are
unable, to produce the vaccine for developing countries in
reasonable quantities and at a reasonable price, IAVI has the
right to contract with other manufacturers to make the vaccine
for use in those countries.
Celladon Collaboration |
In December 2004, we established a collaboration with Celladon
focused on the development of AAV-based drugs for the treatment
of congestive heart failure. In connection with the formation of
this collaboration, certain of Celladons investors
purchased 3,954,132 shares of our common stock at
$1.52 per share resulting in net proceeds to us of
$6.0 million. We recorded the proceeds as equity at the
fair value of the common stock, which approximated market value.
During 2005 we incurred $2.8 million to support development
activities under the Celladon collaboration, which consisted
primarily of internal development and manufacturing efforts. We
agreed to contribute up to $2.0 million to support these
development activities and we are reimbursed for efforts over
that amount. During the current year, we were reimbursed
$0.8 million for those labor hours and outside costs
associated with the Celladon collaboration. We are also entitled
to receive milestone payments during the development of product
candidates under the collaboration as well as royalties and
manufacturing profits from the commercialization of product
candidates developed under the collaboration.
58
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Sirna Therapeutics Collaboration |
In January 2005, we established a collaboration with Sirna to
develop AAV-based approaches to treating Huntingtons
disease. We, and Sirna, have agreed to co-develop product
candidates under the collaboration, whereby we share the costs
of development and any potential future revenues that result
from the collaboration. We split the development costs evenly
with Sirna. We record revenue and Sirna reimburses us for our
program costs that exceed more than half of the total
collaboration costs. One of the members of our Board of
Directors is also a member of the Board of Directors of Sirna.
Former Biogen Collaboration |
In 2000, we established a three-year multiple-product
development and commercialization collaboration with Biogen.
Upon initiation of the collaboration in 2000, Biogen paid us
$8.0 million, which included an upfront technology license
of $5.0 million and up-front prepaid research and
development funding of $3.0 million. Under this agreement,
Biogen provided $3.0 million of additional research and
development funding, paid at a minimum rate of $1.0 million
per year. We amortized the $8.0 million upfront fee paid by
Biogen over the initial research and development collaboration
period which ended on September 30, 2003. We recognized
revenue on the $1.0 million minimum annual project funding
as we performed specified research and development.
As part of this collaboration, Biogen also agreed to provide us
with loans of up to $10.0 million and committed to
purchase, at our discretion, up to $10.0 million of our
common stock. In 2001, we borrowed $10.0 million under the
loan commitment. In 2002, we issued 5,804,673 shares of our
common stock to Biogen at a price of approximately
$0.69 per share and received proceeds of $4.0 million
and in 2003, we issued 2,515,843 shares of our common stock
to Biogen at a price of $1.91 per share and received
proceeds of $4.8 million. As of December 31, 2005,
Biogen owned approximately 11.7 million shares of our
common stock representing approximately 13.7% of our total
common shares outstanding.
Former Wyeth Collaboration |
In 2000, we entered into a collaboration to develop gene therapy
products for treating hemophilia with Wyeth. Under the terms of
a research and development funding agreement, Wyeth paid us
upfront payments of $5.6 million and ongoing payments for
research and development activities performed under the
collaboration. In 2002, Wyeth elected to terminate this
collaboration and related agreements. In 2003, we entered into a
termination agreement with Wyeth that provided for a cash
termination settlement of $2.6 million, and we recognized
$1.3 million in previously received up front cash payments
as revenue.
11. | Employee Retirement Plan |
We sponsor an employee retirement plan under Section 401(k)
of the Internal Revenue Code. All of our employees who meet the
minimum eligibility requirements are eligible to participate in
the plan. Our matching contributions to the 401(k) plan are made
at the discretion of our Board of Directors and were $165,000 in
2005, $133,000 in 2004, and zero in 2003. We suspended matching
contributions effective February 1, 2006.
12. | Income Taxes |
At December 31, 2005, we had net operating loss
carry-forwards of approximately $168.2 million and research
tax credit carry-forwards of $7.5 million. The
carry-forwards will begin to expire in 2007 if not utilized, and
may be further subject to the application of Section 382 of
the Internal Revenue Code of 1986, as amended, as discussed
further below. We have provided a valuation allowance to offset
the deferred tax assets, due to the uncertainty of realizing the
benefits of the net deferred tax asset.
59
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
Significant components of our deferred tax assets and
liabilities were as follows:
December 31, | |||||||||
2005 | 2004 | ||||||||
Deferred tax assets
|
|||||||||
Net operating loss carry-forwards
|
$ | 57,190,000 | $ | 51,230,000 | |||||
Capital loss carry-forwards
|
1,910,000 | 2,080,000 | |||||||
Research and orphan drug credit carry-forwards
|
7,460,000 | 7,000,000 | |||||||
Depreciation and amortization
|
3,340,000 | 3,270,000 | |||||||
Restructure and other
|
2,810,000 | 2,430,000 | |||||||
Gross deferred tax assets
|
72,710,000 | 66,010,000 | |||||||
Valuation allowance for deferred tax assets
|
(72,710,000 | ) | (66,010,000 | ) | |||||
Net deferred tax asset
|
$ | | $ | | |||||
The change in the valuation allowance was $6.7 million for
2005 and $5.4 million for 2004. The capital loss generated
by the sale of CellExSys shares in 2004 may only be carried
forward to offset future capital gains and will expire after
2009 if not utilized. Our valuation allowances as of
December 31, 2005 and December 31, 2004 include an
allowance for this capital loss carry-forward.
Our past sales and issuances of stock have likely resulted in
ownership changes as defined by Section 382 of the Internal
Revenue Code of 1986, as amended. As a result, the utilization
of our net operating losses and tax credits will be limited and
a portion of the carry-forwards may expire unused.
13. | Subsequent Events |
In January 2006, we announced a restructuring in order to reduce
expenses and realign resources to advance our product through
clinical trials. We reduced our workforce by 26 employees,
primarily in early-stage research and development groups and in
operations and general and administrative functions. We incurred
severance expenses of approximately $208,000 related to the
reduction in force, which will be recorded as a restructuring
charge in the first quarter of 2006.
On March 10, 2006, we sold 12,791,611 shares of our
common stock in a public offering at a price of $0.39 per
share and received net proceeds of approximately
$4.8 million.
60
Table of Contents
TARGETED GENETICS CORPORATION
NOTES TO CONSOLIDATED FINANCIAL
STATEMENTS (Continued)
14. | Condensed Quarterly Financial Information (unaudited) |
The following tables present our unaudited quarterly results for
2005 and 2004. The net loss in the third quarter of 2004
reflects a $1.0 million gain on the sale of a
majority-owned subsidiary. We believe that the following
information reflects all normal recurring adjustments for a fair
presentation of the information for the periods presented. The
operating results for any quarter are not necessarily indicative
of results for any future period.
Quarter Ended | ||||||||||||||||
March 31, | June 30, | September 30, | December 31, | |||||||||||||
2005 | 2005 | 2005 | 2005 | |||||||||||||
Revenue
|
$ | 2,000,000 | $ | 1,462,000 | $ | 1,468,000 | $ | 1,944,000 | ||||||||
Restructure charges
|
219,000 | 119,000 | 1,188,000 | 183,000 | ||||||||||||
Loss from operations
|
(4,643,000 | ) | (5,125,000 | ) | (5,863,000 | ) | (3,716,000 | ) | ||||||||
Net loss
|
(4,672,000 | ) | (5,294,000 | ) | (5,683,000 | ) | (3,549,000 | ) | ||||||||
Basic and diluted net loss per common share
|
(0.05 | ) | (0.06 | ) | (0.07 | ) | (0.04 | ) |
Quarter Ended | ||||||||||||||||
March 31, | June 30, | September 30, | December 31, | |||||||||||||
2004 | 2004 | 2004 | 2004 | |||||||||||||
Revenue
|
$ | 1,320,000 | $ | 2,761,000 | $ | 2,388,000 | $ | 3,183,000 | ||||||||
Restructure charges
|
195,000 | 221,000 | 381,000 | 87,000 | ||||||||||||
Loss from operations
|
(4,862,000 | ) | (4,354,000 | ) | (3,737,000 | ) | (2,217,000 | ) | ||||||||
Net loss
|
(4,858,000 | ) | 4,450,000 | ) | (2,724,000 | ) | (2,225,000 | ) | ||||||||
Basic and diluted net loss per common share
|
(0.07 | ) | (0.05 | ) | (0.03 | ) | (0.03 | ) |
61
Table of Contents
Item 9. | Changes in and Disagreements with Accountants on Accounting and Financial Disclosure. |
None.
Item 9A. | Controls and Procedures. |
Evaluation of disclosure controls and procedures. Our
management evaluated, with the participation of our Chief
Executive Officer and our Chief Financial Officer, the
effectiveness of our disclosure controls and procedures as of
the end of the period covered by this Annual Report on
Form 10-K. Based
on this evaluation, our Chief Executive Officer and our Chief
Financial Officer have concluded that our disclosure controls
and procedures are effective to ensure that information we are
required to disclose in reports that we file or submit under the
Securities Exchange Act of 1934 is recorded, processed,
summarized and reported within the time periods specified in
Securities and Exchange Commission rules and forms.
Managements annual report on internal control over
financial reporting. We are responsible for establishing and
maintaining an adequate internal control structure and
procedures our financial reporting. We have assessed the
effectiveness of internal control over financial reporting as of
December 31, 2005. Our assessment was based on criteria set
forth by the Committee of Sponsoring Organizations of the
Treadway Commission, or COSO, Internal Control-Integrated
Framework.
Our internal control over financial reporting is a process
designed to provide reasonable assurance regarding the
reliability of financial reporting and the preparation of
financial statements for external purposes in accordance with
generally accepted accounting principles. Our internal control
over financial reporting includes those policies and procedures
that:
(1) pertain to the maintenance of records that, in reasonable detail, accurately and fairly reflect our transactions and dispositions of the assets; | |
(2) provide reasonable assurance that transactions are recorded as necessary to permit preparation of financial statements in accordance with generally accepted accounting principles, and that our receipts and expenditures are being made only in accordance with authorizations of our management and Board of Directors; and | |
(3) provide reasonable assurance regarding prevention or timely detection of unauthorized acquisition, use, or disposition of our assets that could have a material effect on the financial statements. |
Because of its inherent limitations, internal control over
financial reporting may not prevent or detect misstatements.
Also, projections of any evaluation of effectiveness to future
periods are subject to the risk that controls may become
inadequate because of changes in conditions, or that the degree
of compliance with the policies or procedures may deteriorate.
Based on using the COSO criteria, we believe our internal
control over financial reporting as of December 31, 2005
was effective.
Our independent registered public accounting firm has audited
the consolidated financial statements included in this Annual
Report on
Form 10-K and has
issued a report on managements assessment of our internal
control over financial reporting as well as on the effectiveness
of our internal control over financial reporting, as stated in
their report which is included elsewhere herein.
Changes in internal control over financial reporting.
There was no change in our internal control over financial
reporting that occurred during the period covered by this Annual
Report on
Form 10-K that has
materially affected, or is reasonably likely to materially
affect, our internal control over financial reporting.
62
Table of Contents
Report of Independent Registered Public Accounting Firm
The Board of Directors and Shareholders of Targeted Genetics
Corporation
We have audited managements assessment, included in the
accompanying managements annual report on internal control
over financial reporting, that Targeted Genetics maintained
effective internal control over financial reporting as of
December 31, 2005, based on criteria established in
Internal Control Integrated Framework issued by the
Committee of Sponsoring Organizations of the Treadway Commission
(the COSO criteria). Targeted Genetics Corporations
management is responsible for maintaining effective internal
control over financial reporting and for its assessment of the
effectiveness of internal control over financial reporting. Our
responsibility is to express an opinion on managements
assessment and an opinion on the effectiveness of the
companys internal control over financial reporting based
on our audit.
We conducted our audit in accordance with the standards of the
Public Company Accounting Oversight Board (United States). Those
standards require that we plan and perform the audit to obtain
reasonable assurance about whether effective internal control
over financial reporting was maintained in all material
respects. Our audit included obtaining an understanding of
internal control over financial reporting, evaluating
managements assessment, testing and evaluating the design
and operating effectiveness of internal control, and performing
such other procedures as we considered necessary in the
circumstances. We believe that our audit provides a reasonable
basis for our opinion.
A companys internal control over financial reporting is a
process designed to provide reasonable assurance regarding the
reliability of financial reporting and the preparation of
financial statements for external purposes in accordance with
generally accepted accounting principles. A companys
internal control over financial reporting includes those
policies and procedures that (1) pertain to the maintenance
of records that, in reasonable detail, accurately and fairly
reflect the transactions and dispositions of the assets of the
company; (2) provide reasonable assurance that transactions
are recorded as necessary to permit preparation of financial
statements in accordance with generally accepted accounting
principles, and that receipts and expenditures of the company
are being made only in accordance with authorizations of
management and directors of the company; and (3) provide
reasonable assurance regarding prevention or timely detection of
unauthorized acquisition, use, or disposition of the
companys assets that could have a material effect on the
financial statements.
Because of its inherent limitations, internal control over
financial reporting may not prevent or detect misstatements.
Also, projections of any evaluation of effectiveness to future
periods are subject to the risk that controls may become
inadequate because of changes in conditions, or that the degree
of compliance with the policies or procedures may deteriorate.
In our opinion, managements assessment that Targeted
Genetics Corporation maintained effective internal control over
financial reporting as of December 31, 2005, is fairly
stated, in all material respects, based on the COSO criteria.
Also, in our opinion, Targeted Genetics Corporation maintained,
in all material respects, effective internal control over
financial reporting as of December 31, 2005, based on the
COSO criteria.
We also have audited, in accordance with the standards of the
Public Company Accounting Oversight Board (United States), the
consolidated balance sheets of Targeted Genetics Corporation as
of December 31, 2005 and 2004, and the related consolidated
statements of operations, preferred stock and shareholders
equity, and cash flows for each of the three years in the period
ended December 31, 2005, and our report dated
March 14, 2006 expressed an unqualified opinion thereon.
/s/ Ernst & Young LLP |
Seattle, Washington
March 14, 2006
63
Table of Contents
PART III
Item 10. | Directors and Executive Officers of Registrant |
The information required by this Item is incorporated by
reference to the sections captioned
Proposal One Election of Directors,
Executive Officers, and Section 16(a)
Beneficial Ownership Reporting Compliance in the proxy
statement for our annual meeting of shareholders to be held on
May 8, 2006.
Code of Ethics
We have a Code of Conduct, which applies to all employees,
officers and directors of Targeted Genetics. Our Code of Conduct
meets the requirements of a code of ethics as
defined by Item 406 of
Regulation S-K,
and applies to our Chief Executive Officer, Chief Financial
Officer (who is both our principal financial and principal
accounting officer), as well as all other employees. Our Code of
Conduct also meets the requirements of a code of conduct under
Marketplace Rule 4350(n) of the National Association of
Securities Dealers, Inc. Our Code of Conduct is posted on our
website at
http://www.targetedgenetics.com/investor/corp-info.php under the
heading Corporate Governance.
Item 11. | Executive Compensation. |
The information required by this Item with respect to executive
compensation is incorporated by reference to the section
captioned Executive Compensation in the proxy
statement for our annual meeting of shareholders to be held on
May 8, 2006.
Item 12. | Security Ownership of Certain Beneficial Owners and Management and Related Shareholder Matters. |
The information required by this Item with respect to beneficial
ownership is incorporated by reference to the section captioned
Principal Shareholders and Securities
Authorized for Issuance Under Equity Compensation Plans in
the proxy statement for our annual meeting of shareholders to be
held on May 8, 2006.
Securities Authorized for Issuance under Equity Compensation
Plans. In March 2004, our Board of Directors approved an
increase in the number of shares available for issuance under
our 1999 Stock Option Plan from 6,000,000 shares to
9,500,000 shares. The following table lists our equity
compensation plans, including individual compensation
arrangements, under which equity securities are authorized for
issuance as of December 31, 2005:
Number of Securities | |||||||||||||
to be Issued Upon | Weighted-Average | Number of Securities | |||||||||||
Exercise of Outstanding | Exercise Price of | Remaining Available for | |||||||||||
Options, Warrants and | Outstanding Options, | Future Issuance Under | |||||||||||
Rights | Warrants and Rights | Equity Compensation Plans | |||||||||||
Equity compensation plans approved by security holders
|
7,040,870 | $ | 2.09 | 2,993,541 | |||||||||
Equity compensation plans not subject to approval by security
holders
|
2,000,000 | 3.33 | | ||||||||||
Total
|
9,040,870 | 2.60 | 2,993,541 | ||||||||||
In 1999, in connection with a technology license agreement, we
issued to Alkermes, Inc. a warrant to
purchase 1,000,000 shares of our common stock at an
exercise price of $2.50 per share, expiring in June 2007,
and a warrant to purchase 1,000,000 shares of our
common stock at an exercise price of $4.16 per share,
expiring in June 2009. These warrants are presented in the table
above as Equity compensation plans not subject to approval
by security holders.
64
Table of Contents
Item 13. | Certain Relationships and Related Transactions. |
The information required by this Item with respect to certain
relationships and related-party transactions is incorporated by
reference to the sections captioned Executive
Compensation Change of Control Arrangements
and Executive Compensation Arrangements with
Management in the proxy statement for our annual meeting
of shareholders to be held on May 8, 2006.
Item 14. | Principal Accountant Fees and Services. |
The information required by this Item with respect to principal
accountant fees and services is incorporated by reference to the
section captioned Proposal Three
Ratification of Independent Auditors in the proxy
statement for our annual meeting of shareholders to be held on
May 8, 2006.
PART IV
Item 15. | Exhibits, Financial Statement Schedules and Reports on Form 8-K. |
1. | Financial Statements |
The following consolidated financial statements are submitted in
Part II, Item 8 of this annual report:
Page | ||||
Report of Independent Registered Public Accounting Firm
|
39 | |||
Consolidated Balance Sheets as of December 31, 2005 and 2004
|
40 | |||
Consolidated Statements of Operations for the years ended
December 31, 2005, 2004 and 2003
|
41 | |||
Consolidated Statements of Preferred Stock and
Shareholders Equity for the years ended December 31,
2005, 2004 and 2003
|
42 | |||
Consolidated Statements of Cash Flows for the years ended
December 31, 2005, 2004 and 2003
|
43 | |||
Notes to Consolidated Financial Statements
|
44 |
2. | Financial Statement Schedules |
All financial statement schedules have been omitted because the
required information is either included in the consolidated
financial statements or the notes thereto or is not applicable.
3. | Exhibits |
65
Table of Contents
INDEX TO EXHIBITS
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
3 | .1 | Amended and Restated Articles of Incorporation | 8-K | 05/26/2005 | 3.1 | |||||||||||||
3 | .2 | Amended and Restated Bylaws | 10-K | 12/31/1996 | 3.2 | |||||||||||||
4 | .1 | Rights Agreement, dated as of October 17, 1996, between Targeted Genetics and ChaseMellon Shareholder Services | 8-A | 10/22/1996 | 2.1 | |||||||||||||
4 | .2 | First Amendment of Rights Agreement, dated July 21, 1999, between Targeted Genetics and ChaseMellon Shareholder Services | 8-K | 8/4/1999 | 1.9 | |||||||||||||
4 | .3 | Second Amendment to Rights Agreement, dated September 25, 2002, between Targeted Genetics and Mellon Investor Services LLC (formerly known as ChaseMellon Investor Services L.L.C.) | 8-K | 10/10/2002 | 10.1 | |||||||||||||
4 | .4 | Third Amendment to Rights Agreement, dated January 23, 2003, between Targeted Genetics and Mellon Investor Services LLC | 10-K | 12/31/2002 | 4.4 | |||||||||||||
4 | .5 | Fourth Amendment to Rights Agreement, dated as of September 2, 2003, between Targeted Genetics and Mellon Investor Services LLC | 8-K | 10/1/2003 | 4.1 | |||||||||||||
10 | .1 | Form of Indemnification Agreement between Targeted Genetics and its officers and directors | 10-K | 12/31/1999 | 10.1 | |||||||||||||
10 | .2 | Form of Senior Management Employment Agreement between the registrant and its executive officers | 10-K | 12/31/1996 | 10.2 | |||||||||||||
10 | .3 | Gene Transfer Technology License Agreement, dated as of February 18, 1992, between Immunex Corporation and Targeted Genetics* | 10-K | 12/31/1999 | 10.3 | |||||||||||||
10 | .4 | PHS Patent License Agreement Non-Exclusive, dated as of July 13, 1993, between National Institutes of Health Centers for Disease Control and Targeted Genetics* | 10-K | 12/31/1999 | 10.4 | |||||||||||||
10 | .5 | PHS Patent License Agreement Exclusive, dated as of March 10, 1994, between National Institutes of Health Centers for Disease Control and Targeted Genetics* | 10-K | 12/31/1997 | 10.10 | |||||||||||||
10 | .6 | Patent License Agreement, dated as of December 25, 1993, between The University of Florida Research Foundation, Inc. and Targeted Genetics* | 10-K | 12/31/1999 | 10.5 | |||||||||||||
10 | .7 | First Amended and Restated License Agreement, effective as of October 12, 1995, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | S-1 | 4/16/1996 | 10.30 |
66
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .8 | Amendment to First Amended and Restated License Agreement, dated as of June 19, 1996, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | 10-Q | 6/30/1996 | 10.1 | |||||||||||||
10 | .9 | Second Amendment to First Amended and Restated License Agreement, dated as of April 17, 1998, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | 10-K | 12/31/1998 | 10.16 | |||||||||||||
10 | .10 | Exclusive Sublicense Agreement, dated June 9, 1999, between Targeted Genetics and Alkermes, Inc.* | 8-K | 1/6/1999 | 10.36 | |||||||||||||
10 | .11 | Amendment Agreement to Exclusive Sublicense Agreement, dated as of March 12, 2002, between Targeted Genetics and Alkermes, Inc.* | 10-K | 12/31/2003 | 10.52 | |||||||||||||
10 | .12 | Amendment No. 2 to Exclusive Sublicense Agreement, dated as of May 29, 2003, between Targeted Genetics and Alkermes, Inc.* | 8-K | 10/1/2003 | 10.1 | |||||||||||||
10 | .13 | Common Stock and Warrants Issuance Agreement, dated June 9, 1999, by and between Targeted Genetics and Alkermes, Inc. | 10-Q | 6/30/1999 | 10.1 | |||||||||||||
10 | .14 | Warrant Agreements, dated June 9, 1999, by and between Targeted Genetics and Alkermes, Inc. | 10-Q | 6/30/1999 | 10.2 | |||||||||||||
10 | .15 | Funding Agreement, dated as of July 21, 1999, among Targeted Genetics, Elan International Services, Ltd., and Elan Corporation, plc | 8-K | 8/4/1999 | 1.3 | |||||||||||||
10 | .16 | Subscription, Joint Development and Operating Agreement, dated as of July 21, 1999, among Elan Corporation, plc, Elan International Services, Ltd., Targeted Genetics and Targeted Genetics Newco, Ltd.* | 8-K | 8/4/1999 | 1.4 | |||||||||||||
10 | .17 | Convertible Promissory Note, dated July 21, 1999, issued by Targeted Genetics to Elan International Services, Ltd. | 8-K | 8/4/1999 | 1.5 | |||||||||||||
10 | .18 | License Agreement dated July 21, 1999, between Targeted Genetics Newco, Ltd. and Targeted Genetics* | 8-K | 8/4/1999 | 1.6 | |||||||||||||
10 | .19 | License Agreement, dated July 21, 1999, between Targeted Genetics Newco, Ltd. and Elan Pharmaceutical Technologies, a division of Elan Corporation, plc* | 8-K | 8/4/1999 | 1.7 |
67
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .20 | Termination Agreement, dated March 31, 2004, among Targeted Genetics, Elan Corporation PLC, Elan Pharma International Limited, Elan International Services, Ltd. and Emerald Gene Systems, Ltd.* | 8-K | 4/06/2004 | 99.2 | |||||||||||||
10 | .21 | Agreement and Plan of Merger dated as of August 8, 2000, among Targeted Genetics, Inc., Genovo, Inc., TGC Acquisition Corporation and Biogen, Inc.* | 8-K | 8/23/2000 | 2.1 | |||||||||||||
10 | .22 | Development and Marketing Agreement, dated as of August 8, 2000, between Targeted Genetics, Genovo, Inc. and Biogen, Inc. | 8-K | 9/13/200 | 10.1 | |||||||||||||
10 | .23 | Funding Agreement dated as of August 8, 2000, between Targeted Genetics and Biogen, Inc. | 8-K | 9/13/2000 | 10.2 | |||||||||||||
10 | .24 | Amendment to Funding Agreement, dated as of July 14, 2003, between Targeted Genetics and Biogen, Inc. | 8-K | 7/22/2003 | 10.3 | |||||||||||||
10 | .25 | Amendment No. 2 to Funding Agreement, dated September 1, 2005, between Targeted Genetics and Biogen Idec | 8-K | 9/1/05 | 10.1 | |||||||||||||
10 | .26 | Amended and Restated Promissory Note of Targeted Genetics dated September 1, 2005 (issued to Biogen Idec in principal amount of $10,000,000) | 8-K | 9/1/05 | 10.2 | |||||||||||||
10 | .27 | Amended and Restated Promissory Note of Targeted Genetics dated September 1, 2005 (issued to Biogen Idec in principal amount of $650,000) | 8-K | 9/1/05 | 10.3 | |||||||||||||
10 | .28 | Industrial Collaboration Agreement, dated as of February 1, 2000, between the International Aids Vaccine Initiative, Inc., Childrens Research Institute and Targeted Genetics* | 10-Q | 9/30/2002 | 10.1 | |||||||||||||
10 | .29 | Amendment No. 1 to Industrial Collaboration Agreement, dated as of March 14, 2003, among the International Aids Vaccine Initiative, Inc., Childrens Research Institute and Targeted Genetics* | 10-K | 12/31/2002 | 10.42 | |||||||||||||
10 | .29 | Amendment No. 2 to Industrial Collaboration Agreement, dated August 1, 2003, among Targeted Genetics, International Aids Vaccine Initiative, Inc. and Childrens Research Institute* | 10-Q | 9/30/2003 | 10.2 | |||||||||||||
10 | .30 | Amendment No. 3 to Industrial Collaboration Agreement, dated December 2, 2003, among Targeted Genetics, International Aids Vaccine Initiative, Inc. and Childrens Research Institute* | 8-K | 1/13/2004 | 10.2 |
68
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .31 | Study Funding Agreement, dated as of April 23, 2003, between Targeted Genetics and Cystic Fibrosis Foundation Therapeutics, Inc.* | 10-Q | 3/31/2003 | 10.2 | |||||||||||||
10 | .32 | Collaboration Agreement, dated December 31, 2004, between Targeted Genetics and Celladon Corporation.* | 10-K | 12/31/2004 | 10.56 | |||||||||||||
10 | .34 | Manufacturing Agreement, dated December 31, 2004, between Targeted Genetics and Celladon Corporation.* | 10-K | 12/31/2004 | 10.57 | |||||||||||||
10 | .35 | Common Stock Purchase Agreement, dated December 31, 2004, by and among Targeted Genetics, Enterprise Partners and Venrock Partners | 10-K | 12/31/2004 | 10.58 | |||||||||||||
10 | .35 | Biological Processing Services Agreement, dated as of March 28, 2003, between GenVec, Inc. and Targeted Genetics* | 10-Q | 3/31/2003 | 10.1 | |||||||||||||
10 | .36 | Agreement Under an NIH Prime Award, dated February 8, 2006, between The Childrens Hospital of Philadelphia and Targeted Genetics* | 10-K | X | ||||||||||||||
10 | .37 | Office Lease, dated as of October 7, 1996, between Benaroya Capital Company, LLC and Targeted Genetics | 10-K | 12/31/1996 | 10.26 | |||||||||||||
10 | .39 | Second Lease Amendment, dated February 25, 2000, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.2 | |||||||||||||
10 | .40 | Third Lease Amendment, dated April 19, 2000, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.3 | |||||||||||||
10 | .41 | Fourth Lease Amendment, dated March 28, 2001, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.4 | |||||||||||||
10 | .42 | Fifth Lease Amendment, dated January 2, 2004, between Targeted Genetics and Benaroya Capital Company, LLC* | 8-K | 1/13/2004 | 10.3 | |||||||||||||
10 | .43 | Canyon Park Building Lease, dated as of June 30, 2000, between Targeted Genetics and CarrAmerica Corporation | 10-Q | 6/30/2000 | 10.1 | |||||||||||||
10 | .44 | Olive Way Building Lease, dated as of November 20, 1993, as amended, between Targeted Genetics and Ironwood Apartments, Inc. (successor in interest to Metropolitan Federal Savings and Loan Association) | 10-K | 12/31/1999 | 10.29 | |||||||||||||
10 | .45 | Fifth Amendment to Lease Agreement, dated as of June 20, 2003, between Targeted Genetics and Ironwood Apartments, Inc. | 8-K | 7/22/2003 | 10.2 |
69
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .46 | Sixth Amendment to Lease Agreement, dated as of November 1, 2003, between Targeted Genetics and Ironwood Apartments, Inc.* | 8-K | 1/13/2004 | 10.1 | |||||||||||||
10 | .47 | 1992 Restated Stock Option Plan | S-8 | 7/10/1998 | 99.1 | |||||||||||||
10 | .48 | Stock Option Plan for Nonemployee Directors | 10-Q | 3/31/2001 | 10.34 | |||||||||||||
10 | .49 | 1999 Stock Option Plan, as amended and restated March 22, 2004 | S-8 | 6/17/2004 | 10.1 | |||||||||||||
10 | .50 | 2000 Genovo Inc. Roll-Over Stock Option Plan | S-8 | 10/19/2000 | 99.1 | |||||||||||||
21 | .1 | Subsidiaries of Targeted Genetics | X | |||||||||||||||
23 | .1 | Consent of Independent Registered Public Accounting Firm | X | |||||||||||||||
31 | .1 | Certification of Chief Executive Officer pursuant to Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934, as amended | X | |||||||||||||||
31 | .2 | Certification of Chief Financial Officer pursuant to Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934, as amended | X | |||||||||||||||
32 | .1 | Certification of Chief Executive Officer pursuant to 18 U.S.C. Section 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002 | X | |||||||||||||||
32 | .2 | Certification of Chief Financial Officer pursuant to 18 U.S.C. Section 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002 | X |
* | Portions of these exhibits have been omitted based on a grant of or application for confidential treatment from the SEC. The omitted portions of these exhibits have been filed separately with the SEC. |
70
Table of Contents
SIGNATURES
Pursuant to the requirements of Section 13 or 15(d) of the
Securities Exchange Act of 1934, as amended, the registrant has
duly caused this report to be signed on its behalf by the
undersigned, thereunto duly authorized in the city of Seattle,
state of Washington, on March 14, 2006.
TARGETED GENETICS CORPORATION |
By: | /s/ H. Stewart Parker |
|
|
H. Stewart Parker | |
President and Chief Executive Officer |
POWER OF ATTORNEY
Each person whose individual signature appears below hereby
authorizes and appoints H. Stewart Parker and David J. Poston,
and each of them, with full power of substitution and
resubstitution and full power to act without the other, as his
or her true and lawful
attorney-in-fact and
agent to act in his or her name, place and stead and to execute
in the name and on behalf of each person, individually and in
each capacity stated below, and to file, any and all amendments
to this report, and to file the same, with all exhibits thereto,
and other documents in connection therewith, with the Securities
and Exchange Commission, granting unto said
attorneys-in-fact and
agents, and each of them, full power and authority to do and
perform each and every act and thing, ratifying and confirming
all that said
attorneys-in-fact and
agents or any of them or their or his or her substitute or
substitutes may lawfully do or cause to be done by virtue
thereof.
Pursuant to the requirements of the Securities Exchange Act of
1934, this report has been signed below by the following persons
on behalf of the registrant and in the capacities and on the
dates indicated.
Signature | Title | Date | ||||
/s/ H. Stewart Parker H. Stewart Parker |
President, Chief Executive Officer and Director (Principal Executive Officer) |
March 14, 2006 | ||||
/s/ David J. Poston David J. Poston |
Vice President, Finance, Chief Financial Officer and Treasurer
(Principal Financial and Accounting Officer) |
March 14, 2006 | ||||
/s/ Jeremy L. Curnock
Cook Jeremy L. Curnock Cook |
Chairman of the Board | March 14, 2006 | ||||
/s/ Jack L. Bowman Jack L. Bowman |
Director | March 14, 2006 | ||||
/s/ Joseph M.
Davie, Ph.D., M.D. Joseph M. Davie, Ph.D., M.D. |
Director | March 14, 2006 | ||||
/s/ Roger L. Hawley Roger L. Hawley |
Director | March 14, 2006 | ||||
/s/ Louis P. Lacasse Louis P. Lacasse |
Director | March 14, 2006 | ||||
Nelson L. Levy, Ph.D., M.D. |
Director | March , 2006 | ||||
Michael S. Perry, D.V.M, Ph.D. |
Director | March , 2006 |
71
Table of Contents
EXHIBIT INDEX
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
3 | .1 | Amended and Restated Articles of Incorporation | 8-K | 05/26/2005 | 3.1 | |||||||||||||
3 | .2 | Amended and Restated Bylaws | 10-K | 12/31/1996 | 3.2 | |||||||||||||
4 | .1 | Rights Agreement, dated as of October 17, 1996, between Targeted Genetics and ChaseMellon Shareholder Services | 8-A | 10/22/1996 | 2.1 | |||||||||||||
4 | .2 | First Amendment of Rights Agreement, dated July 21, 1999, between Targeted Genetics and ChaseMellon Shareholder Services | 8-K | 8/4/1999 | 1.9 | |||||||||||||
4 | .3 | Second Amendment to Rights Agreement, dated September 25, 2002, between Targeted Genetics and Mellon Investor Services LLC (formerly known as ChaseMellon Investor Services L.L.C.) | 8-K | 10/10/2002 | 10.1 | |||||||||||||
4 | .4 | Third Amendment to Rights Agreement, dated January 23, 2003, between Targeted Genetics and Mellon Investor Services LLC | 10-K | 12/31/2002 | 4.4 | |||||||||||||
4 | .5 | Fourth Amendment to Rights Agreement, dated as of September 2, 2003, between Targeted Genetics and Mellon Investor Services LLC | 8-K | 10/1/2003 | 4.1 | |||||||||||||
10 | .1 | Form of Indemnification Agreement between Targeted Genetics and its officers and directors | 10-K | 12/31/1999 | 10.1 | |||||||||||||
10 | .2 | Form of Senior Management Employment Agreement between the registrant and its executive officers | 10-K | 12/31/1996 | 10.2 | |||||||||||||
10 | .3 | Gene Transfer Technology License Agreement, dated as of February 18, 1992, between Immunex Corporation and Targeted Genetics* | 10-K | 12/31/1999 | 10.3 | |||||||||||||
10 | .4 | PHS Patent License Agreement Non-Exclusive, dated as of July 13, 1993, between National Institutes of Health Centers for Disease Control and Targeted Genetics* | 10-K | 12/31/1999 | 10.4 | |||||||||||||
10 | .5 | PHS Patent License Agreement Exclusive, dated as of March 10, 1994, between National Institutes of Health Centers for Disease Control and Targeted Genetics* | 10-K | 12/31/1997 | 10.10 | |||||||||||||
10 | .6 | Patent License Agreement, dated as of December 25, 1993, between The University of Florida Research Foundation, Inc. and Targeted Genetics* | 10-K | 12/31/1999 | 10.5 | |||||||||||||
10 | .7 | First Amended and Restated License Agreement, effective as of October 12, 1995, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | S-1 | 4/16/1996 | 10.30 |
72
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .8 | Amendment to First Amended and Restated License Agreement, dated as of June 19, 1996, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | 10-Q | 6/30/1996 | 10.1 | |||||||||||||
10 | .9 | Second Amendment to First Amended and Restated License Agreement, dated as of April 17, 1998, between The University of Tennessee Research Corporation and RGene Therapeutics, Inc.* | 10-K | 12/31/1998 | 10.16 | |||||||||||||
10 | .10 | Exclusive Sublicense Agreement, dated June 9, 1999, between Targeted Genetics and Alkermes, Inc.* | 8-K | 1/6/1999 | 10.36 | |||||||||||||
10 | .11 | Amendment Agreement to Exclusive Sublicense Agreement, dated as of March 12, 2002, between Targeted Genetics and Alkermes, Inc.* | 10-K | 12/31/2003 | 10.52 | |||||||||||||
10 | .12 | Amendment No. 2 to Exclusive Sublicense Agreement, dated as of May 29, 2003, between Targeted Genetics and Alkermes, Inc.* | 8-K | 10/1/2003 | 10.1 | |||||||||||||
10 | .13 | Common Stock and Warrants Issuance Agreement, dated June 9, 1999, by and between Targeted Genetics and Alkermes, Inc. | 10-Q | 6/30/1999 | 10.1 | |||||||||||||
10 | .14 | Warrant Agreements, dated June 9, 1999, by and between Targeted Genetics and Alkermes, Inc. | 10-Q | 6/30/1999 | 10.2 | |||||||||||||
10 | .15 | Funding Agreement, dated as of July 21, 1999, among Targeted Genetics, Elan International Services, Ltd., and Elan Corporation, plc | 8-K | 8/4/1999 | 1.3 | |||||||||||||
10 | .16 | Subscription, Joint Development and Operating Agreement, dated as of July 21, 1999, among Elan Corporation, plc, Elan International Services, Ltd., Targeted Genetics and Targeted Genetics Newco, Ltd.* | 8-K | 8/4/1999 | 1.4 | |||||||||||||
10 | .17 | Convertible Promissory Note, dated July 21, 1999, issued by Targeted Genetics to Elan International Services, Ltd. | 8-K | 8/4/1999 | 1.5 | |||||||||||||
10 | .18 | License Agreement dated July 21, 1999, between Targeted Genetics Newco, Ltd. and Targeted Genetics* | 8-K | 8/4/1999 | 1.6 | |||||||||||||
10 | .19 | License Agreement, dated July 21, 1999, between Targeted Genetics Newco, Ltd. and Elan Pharmaceutical Technologies, a division of Elan Corporation, plc* | 8-K | 8/4/1999 | 1.7 |
73
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .20 | Termination Agreement, dated March 31, 2004, among Targeted Genetics, Elan Corporation PLC, Elan Pharma International Limited, Elan International Services, Ltd. and Emerald Gene Systems, Ltd.* | 8-K | 4/06/2004 | 99.2 | |||||||||||||
10 | .21 | Agreement and Plan of Merger dated as of August 8, 2000, among Targeted Genetics, Inc., Genovo, Inc., TGC Acquisition Corporation and Biogen, Inc.* | 8-K | 8/23/2000 | 2.1 | |||||||||||||
10 | .22 | Development and Marketing Agreement, dated as of August 8, 2000, between Targeted Genetics, Genovo, Inc. and Biogen, Inc. | 8-K | 9/13/200 | 10.1 | |||||||||||||
10 | .23 | Funding Agreement dated as of August 8, 2000, between Targeted Genetics and Biogen, Inc. | 8-K | 9/13/2000 | 10.2 | |||||||||||||
10 | .24 | Amendment to Funding Agreement, dated as of July 14, 2003, between Targeted Genetics and Biogen, Inc. | 8-K | 7/22/2003 | 10.3 | |||||||||||||
10 | .25 | Amendment No. 2 to Funding Agreement, dated September 1, 2005, between Targeted Genetics and Biogen Idec | 8-K | 9/1/05 | 10.1 | |||||||||||||
10 | .26 | Amended and Restated Promissory Note of Targeted Genetics dated September 1, 2005 (issued to Biogen Idec in principal amount of $10,000,000) | 8-K | 9/1/05 | 10.2 | |||||||||||||
10 | .27 | Amended and Restated Promissory Note of Targeted Genetics dated September 1, 2005 (issued to Biogen Idec in principal amount of $650,000) | 8-K | 9/1/05 | 10.3 | |||||||||||||
10 | .28 | Industrial Collaboration Agreement, dated as of February 1, 2000, between the International Aids Vaccine Initiative, Inc., Childrens Research Institute and Targeted Genetics* | 10-Q | 9/30/2002 | 10.1 | |||||||||||||
10 | .29 | Amendment No. 1 to Industrial Collaboration Agreement, dated as of March 14, 2003, among the International Aids Vaccine Initiative, Inc., Childrens Research Institute and Targeted Genetics* | 10-K | 12/31/2002 | 10.42 | |||||||||||||
10 | .29 | Amendment No. 2 to Industrial Collaboration Agreement, dated August 1, 2003, among Targeted Genetics, International Aids Vaccine Initiative, Inc. and Childrens Research Institute* | 10-Q | 9/30/2003 | 10.2 | |||||||||||||
10 | .30 | Amendment No. 3 to Industrial Collaboration Agreement, dated December 2, 2003, among Targeted Genetics, International Aids Vaccine Initiative, Inc. and Childrens Research Institute* | 8-K | 1/13/2004 | 10.2 |
74
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .31 | Study Funding Agreement, dated as of April 23, 2003, between Targeted Genetics and Cystic Fibrosis Foundation Therapeutics, Inc.* | 10-Q | 3/31/2003 | 10.2 | |||||||||||||
10 | .32 | Collaboration Agreement, dated December 31, 2004, between Targeted Genetics and Celladon Corporation.* | 10-K | 12/31/2004 | 10.56 | |||||||||||||
10 | .34 | Manufacturing Agreement, dated December 31, 2004, between Targeted Genetics and Celladon Corporation.* | 10-K | 12/31/2004 | 10.57 | |||||||||||||
10 | .35 | Common Stock Purchase Agreement, dated December 31, 2004, by and among Targeted Genetics, Enterprise Partners and Venrock Partners. | 10-K | 12/31/2004 | 10.58 | |||||||||||||
10 | .35 | Biological Processing Services Agreement, dated as of March 28, 2003, between GenVec, Inc. and Targeted Genetics* | 10-Q | 3/31/2003 | 10.1 | |||||||||||||
10 | .36 | Agreement Under an NIH Prime Award, dated February 8, 2006, between The Childrens Hospital of Philadelphia and Targeted Genetics* | 10-K | X | ||||||||||||||
10 | .37 | Office Lease, dated as of October 7, 1996, between Benaroya Capital Company, LLC and Targeted Genetics | 10-K | 12/31/1996 | 10.26 | |||||||||||||
10 | .38 | First Lease Amendment, dated May 12, 1997, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.1 | |||||||||||||
10 | .39 | Second Lease Amendment, dated February 25, 2000, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.2 | |||||||||||||
10 | .40 | Third Lease Amendment, dated April 19, 2000, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.3 | |||||||||||||
10 | .41 | Fourth Lease Amendment, dated March 28, 2001, between Targeted Genetics and Benaroya Capital Company, LLC | 10-Q | 6/30/2001 | 10.4 | |||||||||||||
10 | .42 | Fifth Lease Amendment, dated January 2, 2004, between Targeted Genetics and Benaroya Capital Company, LLC* | 8-K | 1/13/2004 | 10.3 | |||||||||||||
10 | .43 | Canyon Park Building Lease, dated as of June 30, 2000, between Targeted Genetics and CarrAmerica Corporation | 10-Q | 6/30/2000 | 10.1 | |||||||||||||
10 | .44 | Olive Way Building Lease, dated as of November 20, 1993, as amended, between Targeted Genetics and Ironwood Apartments, Inc. (successor in interest to Metropolitan Federal Savings and Loan Association) | 10-K | 12/31/1999 | 10.29 | |||||||||||||
10 | .45 | Fifth Amendment to Lease Agreement, dated as of June 20, 2003, between Targeted Genetics and Ironwood Apartments, Inc. | 8-K | 7/22/2003 | 10.2 |
75
Table of Contents
Incorporated by Reference | ||||||||||||||||||
Exhibit No. | Exhibit Description | Form | Date | Number | Filed Herewith | |||||||||||||
10 | .46 | Sixth Amendment to Lease Agreement, dated as of November 1, 2003, between Targeted Genetics and Ironwood Apartments, Inc.* | 8-K | 1/13/2004 | 10.1 | |||||||||||||
10 | .47 | 1992 Restated Stock Option Plan | S-8 | 7/10/1998 | 99.1 | |||||||||||||
10 | .48 | Stock Option Plan for Nonemployee Directors | 10-Q | 3/31/2001 | 10.34 | |||||||||||||
10 | .49 | 1999 Stock Option Plan, as amended and restated March 22, 2004 | S-8 | 6/17/2004 | 10.1 | |||||||||||||
10 | .50 | 2000 Genovo Inc. Roll-Over Stock Option Plan | S-8 | 10/19/2000 | 99.1 | |||||||||||||
21 | .1 | Subsidiaries of Targeted Genetics | X | |||||||||||||||
23 | .1 | Consent of Independent Registered Public Accounting Firm | X | |||||||||||||||
31 | .1 | Certification of Chief Executive Officer pursuant to Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934, as amended | X | |||||||||||||||
31 | .2 | Certification of Chief Financial Officer pursuant to Rules 13a-15(e) and 15d-15(e) under the Securities Exchange Act of 1934, as amended | X | |||||||||||||||
32 | .1 | Certification of Chief Executive Officer pursuant to 18 U.S.C. Section 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002 | X | |||||||||||||||
32 | .2 | Certification of Chief Financial Officer pursuant to 18 U.S.C. Section 1350, as adopted pursuant to Section 906 of the Sarbanes-Oxley Act of 2002 | X |
* | Portions of these exhibits have been omitted based on a grant of or application for confidential treatment from the SEC. The omitted portions of these exhibits have been filed separately with the SEC. |
76