CASI Pharmaceuticals, Inc (DE) - Annual Report: 2005 (Form 10-K)
FORM 10-K
SECURITIES AND EXCHANGE COMMISSION
WASHINGTON, D. C., 20549
ANNUAL REPORT PURSUANT TO SECTION 13 OR 15 (d) OF
THE SECURITIES EXCHANGE ACT OF 1934
For the fiscal year ended December 31, 2005
THE SECURITIES EXCHANGE ACT OF 1934
For the fiscal year ended December 31, 2005
Commission file number 0-20713
ENTREMED, INC.
(Exact name of registrant as specified in its charter)
Delaware | 58-1959440 | |
(State of Incorporation) | (I.R.S. Employer Identification No.) | |
9640 Medical Center Drive, Rockville, MD | 20850 | |
(Address of principal executive offices) | (Zip Code) |
Registrants telephone
number, including area code: (240) 864 - 2600
Securities registered pursuant to Section 12 (g) of the Act:
Title
Common Stock, Par Value $.01 Per Share
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
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
As of June 30, 2005, the aggregate market value of the shares of common stock held by
non-affiliates was approximately $100,936,011.
As of March 8, 2006, 64,079,823 shares of the Companys common stock were outstanding.
Documents incorporated by reference
None.
ENTREMED, INC.
FORM 10-K FISCAL YEAR ENDED DECEMBER 31, 2005
FORM 10-K FISCAL YEAR ENDED DECEMBER 31, 2005
Contents and Cross Reference Sheet
Form 10-K | Form 10-K | Form 10-K | ||||||
Part No. | Item No. | Description | Page No. | |||||
I |
1 | Business | 2 | |||||
1A | Risk Factors | 12 | ||||||
1B | Unresolved Staff Comments | 17 | ||||||
2 | Properties | 17 | ||||||
3 | Legal Proceedings | 17 | ||||||
4 | Submission of Matters to a Vote of Security Holders | 17 | ||||||
II |
5 | Market for Registrants Common Equity, | ||||||
Related Stockholder Matters | ||||||||
And Issuer Purchases of Equity Securities | 18 | |||||||
6 | Selected Financial Data | 19 | ||||||
7 | Managements Discussion and Analysis of | |||||||
Financial Condition and Results of Operation | 20 | |||||||
7A | Quantitative and Qualitative Disclosures | |||||||
About Market Risk | 28 | |||||||
8 | Financial Statements and Supplementary Data | 28 | ||||||
9 | Changes in and Disagreements with Accountants | |||||||
on Accounting and Financial Disclosure | 28 | |||||||
9A | Controls and Procedures | 28 | ||||||
III |
10 | Directors and Executive Officers of the Registrant | 30 | |||||
11 | Executive Compensation | 34 | ||||||
12 | Security Ownership of Certain Beneficial Owners and | |||||||
Management and Related Stockholder Matters | 38 | |||||||
13 | Certain Relationships and Related Transactions | 39 | ||||||
14 | Principal Accounting Fees and Services | 39 | ||||||
IV |
15 | Exhibits, Financial Statement Schedules | 40 | |||||
Signatures
|
44 | |||||||
Audited Consolidated Financial Statements | F-1 |
SPECIAL NOTE REGARDING FORWARD-LOOKING STATEMENTS
This report contains certain forward-looking statements within the meaning of Section 27A of the
Securities Exchange Act of 1933, as amended and Section 21E of the Securities Exchange Act of 1934,
as amended. Forward-looking statements also may be included in other statements that we make. All
statements that are not descriptions of historical facts are forward-looking statements. These
statements can generally be identified by the use of forward-looking terminology such as
believes, expects, intends, may, will, should, or anticipates or similar terminology.
These forward-looking statements include, among others, statements regarding the timing of our
clinical trials, our cash position and future expenses, and our future revenues.
Our forward-looking statements are based on information available to us today, and we will not
update these statements. Although we believe that the expectations reflected in such
forward-looking statements are reasonable as of the date thereof, actual results could differ
materially from those currently anticipated due to a number of factors, including risks relating to
the early stage of our product candidates under development operating losses and anticipated future
losses; the value of our common stock; our need for additional capital; intense competition and
rapid technological change in the biopharmaceutical industry; uncertainties relating to our patent
and proprietary rights; uncertainties relating to clinical trials: estimated clinical trial
commencement date; government regulation; and uncertainties of obtaining regulatory approval on a
timely basis or at all. Additional information about the factors and risks that could affect our
business, financial condition and results of operations, are contained in our filings with the U.S.
Securities and Exchange Commission (SEC), which are available at www.sec.gov.
1
PART I
ITEM 1. BUSINESS.
OVERVIEW
EntreMed, Inc. (EntreMed or the Company) (Nasdaq: EMND) is a clinical-stage pharmaceutical
company focused on developing next generation multi-mechanism oncology and antiinflammatory drugs
that target disease cells directly and the blood vessels that nourish them. EntreMed is focused on
developing drugs that are safe and convenient, and provide the potential for improved patient
outcomes. Panzem® (2-methoxyestradiol or 2ME2), one of the Companys lead drug
candidates, is currently in Phase 2 clinical trials for cancer, as well as in preclinical
development for rheumatoid arthritis. MKC-1, a novel cell cycle inhibitor acquired through the
recent acquisition of Miikana Therapeutics, is also in Phase 2 clinical trials for cancer.
ENMD-1198, a novel tubulin binding agent discovered by EntreMed, has an active Investigational New
Drug (IND) application on file with the Food and Drug Administration (FDA). We anticipate that a
Phase 1 clinical trial for ENMD-1198 will commence in the first half of 2006.
In January 2006, we acquired Miikana Therapeutics, Inc., a clinical-stage biopharmaceutical
company with research laboratories in Toronto, Canada. As a result of the transaction, EntreMed
enhanced its pipeline with the addition of a Phase 2 drug candidate, MKC-1, and two preclinical
programs, one in aurora kinase inhibition and one in HDAC inhibition.
Our goal is to develop and commercialize therapeutics based on our scientific expertise in
angiogenesis, cell cycle regulation and inflammation processes vital to the progression of
cancer and other diseases. Our three clinical product candidates are based on these mechanisms.
Our expertise has also led to the identification of new molecules, including new chemical entities
derived from 2ME2, modulators of fibroblast growth factor-2 (FGF-2) activity, proteinase activated
receptor-2 (PAR-2) antagonists, and tissue factor pathway inhibitor (TFPI) peptides.
In
order to further advance our commercial objectives, we may seek strategic alliances,
licensing relationships and co-development partnerships with other companies to develop compounds
for both oncology and non-oncology therapeutic areas.
We were incorporated under Delaware law in 1991. Our principal executive offices are located
at 9640 Medical Center Drive, Rockville, Maryland 20850, and our telephone number is (240)
864-2600.
SCIENTIFIC FOUNDATION
We developed our initial drug pipeline based on comprehensive research into the relationship
between malignancy and the process of angiogenesis (the growth of new blood vessels). This
research led to our focus on drug candidates that act on the cellular pathways that affect
biological processes important in multiple diseases, specifically angiogenesis, inflammation and
cell cycle regulation. Our drug candidates have potential applications in oncology and other
diseases because they are involved with one or more of these pathways.
Angiogenesis. Angiogenesis is a multi-step process in which preexisting blood vessels send
out capillary sprouts to produce new blood vessels. This tightly regulated process involves the
migration, proliferation and differentiation of endothelial cells. In normal physiology,
angiogenesis is a necessary component of the menstrual cycle and wound healing, where the process
is regulated through appropriate shifts in the balance of pro-angiogenic and anti-angiogenic
signals. This tight regulation of angiogenesis in normal physiology is absent or aberrant in
multiple disease settings that are characterized by persistent, inappropriate blood vessel
development.
Angiogenesis occurs in more than 80 diseases, particularly in various cancers where the growth
of new blood vessels is necessary to sustain tumor growth, as well as arthritis, where inflammation
triggers new blood vessel growth and joint erosion. EntreMed scientists, who have studied the
process of angiogenesis in-depth for over a decade, are developing drug candidates to inhibit blood vessel formation and, in turn, control or
stop diseases resulting from inappropriate blood vessel growth.
2
Cell Cycle Regulation. Cell cycle regulation is the replication, differentiation and death of
cells. One specific aspect of cell cycle regulation is the programmed control of cell death
(apoptosis). In certain diseases, such as cancer, the balance between cell proliferation and cell
death is altered, resulting in inappropriate cell growth. Our compounds impact biochemical pathways
in cells that result in their death via apoptosis. We believe that the selective induction of
apoptosis through drugs that block cell cycle activities can either stabilize or cause the
regression of cancer, inflammation and other disease processes characterized by inappropriate cell
growth. Our preclinical studies have demonstrated induction of endothelial cell and tumor cell
apoptosis.
Inflammation. Inflammation is the process involving the reaction of tissue to injury or
disease. The condition may be either local or systemic and can be divided into acute (immediate)
and chronic (prolonged) patterns. The endothelial cell and angiogenesis (formation of new blood
vessels) are involved in inflammatory diseases. In contrast to acute inflammation, which is
defined by vascular changes, edema, and white blood cell accumulation (neutrophils), chronic
inflammation is characterized by additional white blood cell changes (macrophages and lymphocytes),
tissue destruction, angiogenesis, and scaring. As a result, the cellular pathways involved in
acute and chronic inflammation can be overlapping or distinct. Inflammation is a process that is
associated with many diseases, including cancer and arthritis. Many of EntreMeds compounds have
demonstrated both anti-inflammatory and anti-tumor properties in preclinical models.
PIPELINE STRENGTH
We believe that our pipeline offers promising candidates for successful commercialization for
the following reasons:
Multiple Mechanisms of Action. Our compounds work through multiple mechanisms of action
(MOA). Therefore, a single compound can attack a disease through multiple cellular pathways, as
well as impact different diseases. For example, 2ME2s MOAs include the inhibition of: 1)
angiogenesis; 2) microtubule (cell structure) formation; and 3) hypoxia inducible factor-1 alpha
(HIF-1á), a protein required for cell survival under stress. Apoptosis (cell death) can also be
induced by 2ME2. Working through multiple mechanisms of action, 2ME2 has the potential to attack
cancer cells through multiple pathways that affect the formation and replication of tumor cells, as
well as interrupt the formation of blood vessels that nourish tumor cells and sustain tumor growth.
Versatility. Our compounds are versatile in terms of possible therapeutic applications.
While our preclinical and clinical efforts continue to focus on oncology and inflammatory diseases,
we believe that other diseases characterized by angiogenesis represent future opportunities.
However, our efforts are currently focused entirely on our core therapeutic programs in oncology
and inflammation. We will evaluate non-core programs on a case-by-case basis, and then only in the
context of external license or development alliances.
Convenient Dosing. We are developing drug candidates that we believe will be easy to use with
minimal interruption to the patients daily routine as compared to other modes of drug
administration. We are focusing specifically on oral drug delivery technologies, as well as other
convenient administration routes.
Intellectual Property Position. All of our pipeline programs, with the exception
of 2ME2 (licensed from Childrens Medical Center Corporation), the tubulin inhibitor program
licensed from Celgene Corporation, and MKC-1 (developed originally by Roche), were discovered and
developed internally and, as a result, are our sole property. Our in-house discoveries have the
potential to significantly reduce its obligation to make future milestone, royalty and/or licensing
payments. We plan to continue supporting our internally-generated pipeline and development efforts.
Economic Viability. We believe that our current focus on the development of small molecule
drug candidates may translate into lower product development, process development, and other
associated costs when compared to biological products. Additionally, greater economic potential may
be possible because each of our compounds may offer several therapeutic applications.
3
DEVELOPMENT PIPELINE
Our pipeline contains a balanced portfolio of clinical product candidates and promising
preclinical compounds. The portfolio contains compounds that work primarily through multiple
mechanisms-of-action and have demonstrated the potential in both oncology and inflammation.
In 2006, our pipeline will consist of two product candidates (Panzem® NCD and
MKC-1) in multiple Phase 2 oncology studies, a Phase 1 clinical trial for ENMD-1198, plus a
preclinical rheumatoid arthritis program that is headed toward IND-directed studies.
CLINICAL PROGRAMS
PanzemÒ. Panzem, 2-methoxyestradiol or 2ME2, is our lead clinical candidate.
2ME2 has multiple mechanisms of action (MOA), including inhibiting angiogenesis, disrupting
microtubule (cell structure) formation, down regulating hypoxia inducible factor-one alpha (HIF-1α,
a survival protein), and inducing apoptosis (cell death). The 2ME2 mechanisms that are
particularly relevant to the treatment of cancer involve inhibiting endothelial cell growth
(anti-angiogenic activity) and killing tumor cells directly (pro-apoptotic activity). Additionally,
preclinical models show that 2ME2 has potential therapeutic applications in inflammatory diseases
such as rheumatoid arthritis. 2ME2 has activity in cell lines that are resistant to various
chemotherapy agents including taxanes (microtubule stabilizing agents), etoposide, adriamycin and
methotrexate (DNA synthesis interfering agents), and tamoxifen (anti-estrogen agent).
PanzemÒ NCD. We have reformulated 2ME2 as an orally-administered liquid
suspension (Panzem® NCD) to increase plasma drug levels in patients.
PanzemÒ NCD uses Elan Drug Deliverys (Elan) NanoCrystalÒ
Colloidal Dispersion (NCD) technology, a proprietary technology that is being used successfully in
marketed pharmaceuticals. The NCD technology produces nanometer-sized particles, which are up to
500 times smaller than particles manufactured by conventional milling techniques. In preclinical
studies, the liquid suspension formulation demonstrated antitumor activity with no additional
toxicity. Phase 1 and 2 clinical trials in 171 patients with a prior capsule formulation showed
evidence of biological activity, even though there was insufficient bioavailability to achieve the
level of antitumor activity seen in preclinical studies.
4
PanzemÒ
Clinical Development Overview in Cancer Patients
Clinical Development Overview in Cancer Patients
# | ||||||||||||
OF | ||||||||||||
TRIAL TYPE | PANZEM FORMULATION | INDICATION | PTS | STATUS | COMMENTS | |||||||
Phase 2 (single agent) |
NCD | Glioblastoma | 32 | Enrolling | Assess safety, pharmacokinetics and efficacy. | |||||||
Phase 1b (single agent) |
NCD | Advanced Cancers | 16 | Closed | Well tolerated. Steady state 2ME2 levels achieved. Phase 2 dose determined. | |||||||
Phase 1* (single agent) |
Capsule | Breast | 31 | Complete | No MTD identified no grade III/IV toxicities Clinical benefit and bone pain relief. | |||||||
Phase 1 (with TaxotereÒ ) |
Capsule | Breast | 15 | Complete | One complete response; 20% overall response rate. No additive toxicity. | |||||||
Phase 1* (single agent) |
Capsule | Advanced solid tumors | 20 | Complete | Partial response in ovarian cancer patient. No MTD identified; well tolerated. | |||||||
Phase 1* (single agent) |
Capsule | Advanced solid tumors | 12 | Complete | Well tolerated. Data under evaluation. | |||||||
Phase 2 (single agent) |
Capsule | Prostate | 33 | Complete | Well tolerated; evidence of disease stabilization. | |||||||
Phase 2 (single agent) |
Capsule | Multiple Myeloma | 60 | Closed | Well tolerated; some patients with stable disease. |
* NCI-sponsored trial. EntreMed provided study drug for this trial.
In November 2005, we announced interim results for two Phase 1b studies for Panzem®
NCD in patients with advanced cancer. The studies were conducted at the University of Wisconsin
Comprehensive Cancer Center and the Indiana University Cancer Center. The data were presented at
the combined American Association for Cancer Research, National Cancer Institute, European
Organization for Research and Treatment of Cancer (AACR-NCI-EORTC) International Conference on
Molecular Targets and Cancer Therapeutics.
Results from both dose-escalation studies concluded that the pharmacokinetic profile of
Panzem® NCD met the study objective. Specifically, a dose and schedule of
administration were determined that provided constant exposure of 2-methoxyestradiol allowing the
selection of a Phase 2 dose. Results from the University of Wisconsin have determined a maximum
tolerated dose with fatigue as the dose limiting toxicity, which was above the recommended Phase 2
dose.
In January 2006, a Phase 2 clinical trial with PanzemÒ NCD commenced in
glioblastoma multiforme (GBM) patients at the Duke University Medical Center Brain Tumor Center.
We expect to commence additional Phase 2 studies with Panzem® NCD during the first half
of 2006. Additional Phase 2 studies under consideration include carcinoid, breast, prostate,
ovarian, renal cell, multiple myeloma and non-small cell lung (NSCLC) cancer.
MKC-1. Through our acquisition of Miikana Therapeutics, we enhanced our oncology pipeline
with the addition of MKC-1, a Phase 2 drug candidate. MKC-1 is an orally-active, small molecule,
cell cycle inhibitor with a unique mechanism of action. Specifically, MKC-1 arrests cellular
mitosis by inhibiting a novel intracellular target important in cellular trafficking that has been
shown to be involved in cell division. A Phase 2 clinical trial of MKC-1 in metastatic breast
cancer patients commenced in January 2006. We anticipate that a second Phase 2 trial with MKC-1 in
non-small cell lung cancer will commence in the second half of 2006.
ENMD-1198. EntreMed discovered a New Chemical Entity (NCE) that inhibits tumor growth based
on modifying the chemical structure of 2-methoxyestradiol (2ME2) to increase its anti-tumor and
anti-angiogenic
5
properties, as well as decrease its rate of metabolism. The lead compound from this program,
ENMD-1198, demonstrated improved pharmacokinetic parameters and a reduced rate of metabolism when
compared to 2ME2. Additionally, ENMD-1198 showed excellent anti-tumor activity in several
preclinical animal models. We own the intellectual property for ENMD-1198.
Preclinical data demonstrated that oral administration of ENMD-1198 leads to pronounced in
vivo antitumor activity in cancer models, resulting in a reduction of tumor burden and/or an
increase in survival when compared to controls. Oral daily treatment with ENMD-1198 in an
orthotopic animal model of human breast cancer led to the disruption of microtubules within tumor
cells and a substantial decrease in tumor cell proliferation and angiogenesis. Results from the
toxicology studies demonstrate that ENMD-1198 affects cell populations with a high proliferative
rate, including bone marrow, gastrointestinal tract and lymphoid organs. These effects are common
with approved cancer agents and can be monitored in the clinic. In November 2005, the Company
received FDA acceptance of an IND for ENMD-1198. EntreMed plans to enter into a Phase 1 oncology
study in the first half of 2006.
PRECLINICAL PIPELINE
2ME2 for Rheumatoid Arthritis. The activities ascribed to 2ME2, namely antiangiogenesis,
pro-apoptosis, down regulation of HIF-1á, and inhibition of bone resorption, suggest it may be
effective in treating diseases with inflammatory components, such as rheumatoid arthritis.
EntreMed and its collaborators have now established the dose-dependent, anti-arthritic activity of
2ME2 following oral administration in four distinct animal models of rheumatoid arthritis. This
activity has been manifested as an inhibition in 1) the infiltration of inflammatory cells, 2)
pannus formation, 3) cartilage lesions, and 4) bone resorption.
Treatment with 2ME2 has resulted in a dose-dependent decrease in the severity of rheumatoid
arthritis in preclinical models, strongly suggesting disease-modifying anti-rheumatic drug (DMARD)
activity the potential to treat the underlying pathology of rheumatoid arthritis, rather than
merely treating symptoms such as pain. Based on these results, we plan to conduct IND-enabling
toxicology studies for 2ME2 in rheumatoid arthritis. The use of PanzemÒ for
rheumatoid arthritis opens the possibility to cross over with 2ME2 into a second therapeutic area
with a large, still underserved market.
MKC-1693. Through our acquisition of Miikana Therapeutics, we strengthened our preclinical
pipeline with the addition of a program in aurora kinase inhibition. Specifically, one of these
compounds, MKC-1693, is an aurora kinase inhibitor with a unique kinase profile and mechanism of
action that is currently in preclinical oncology studies. Aurora kinases are known to be involved
in the process of mitosis, or cell division, which is critical in the development of human cancers.
We anticipate that IND-directed studies leading to the filing of an IND will commence in 2006.
Tubulin Inhibitors. Tubulin inhibitors comprise a broad family of compounds that bind to
tubulin and disrupt microtubules, resulting in programmed cell death (apoptosis). In March 2005,
we in-licensed Celgenes tubulin inhibitor program. We have assumed responsibility for the
preclinical and clinical development of the tubulin inhibitors for oncology applications.
Results from in vitro and in vivo studies have shown that Celgenes tubulin inhibitors inhibit
tumor cell proliferation in a dose-dependent manner and, based on in vitro studies, to inhibit
angiogenesis. Our goal for 2006 is to select a lead compound and commence IND-enabling preclinical
studies.
Proteinase Activated Receptor-2 (PAR-2) Inhibitor. PAR-2 is a cell surface receptor that is
known to play a critical role in acute and chronic inflammation. We discovered a peptide that
blocks PAR-2, the first such compound identified as an antagonist of PAR-2. The anti-PAR-2 peptide
inhibits tumor growth and formation of new blood vessels in preclinical models. Our PAR-2
antagonist has also been shown to be an inhibitor of inflammation in preclinical models.
6
Multiple small molecule PAR-2 antagonists have been synthesized to identify compounds with
increased activity, and we are currently studying PAR-2s potential therapeutic applications in oncology and
inflammatory diseases. We own all intellectual property associated with the PAR-2 antagonist
program and its resulting compounds.
Tissue Factor Pathway Inhibitor (TFPI). TFPI is a naturally occurring anticoagulant protein
that has been shown to inhibit tumor progression in preclinical models. We discovered a peptide
fragment of TFPI that blocks tumor growth and angiogenesis in animal models. In preclinical
studies, the peptide does not affect normal blood clotting, a risk long associated with the
development of coagulation inhibitors for oncology applications. The TFPI peptides antiangiogenic
mechanism of action has now been verified and shown to be independent of TFPIs anticoagulant
activity. The TFPI peptide binds to a very low density lipoprotein receptor and induces apoptosis
(cell death) in endothelial cells, the cells that form blood vessels.
We have entered into a joint research collaboration with Affymax, a leader in the discovery
and development of novel peptide drugs, to identify lead drug candidates for the treatment of
cancer. The resulting drug candidates will subsequently be screened for their therapeutic
potential in oncology, as well as inflammatory diseases, with the goal of bringing one or more lead
compounds forward into clinical development. Our goal for 2006 is to identify one or more such
lead compounds.
FGF-2 Cancer Vaccine (ENMD-0996). EntreMed scientists are currently developing a cancer
vaccine that targets fibroblast growth factor-2 (FGF-2), a potent stimulator of angiogenesis.
ENMD-0996 consists of a specific peptide fragment of FGF-2 in an adjuvant formulation. In
preclinical studies, ENMD-0996 inhibited tumor development in multiple preclinical models with no
evidence of toxicity or any negative effects on wound healing or reproduction. Preclinical
oncology studies are continuing, with the goal to advance ENMD-0996 into IND-directed studies. We
own all intellectual property associated with this program.
HDAC Inhibitors. Developed by Miikana, a histone deacetylase (HDAC) inhibitor program is
currently in preclinical evaluation. HDAC inhibitors have been shown to arrest cancer cell
growth and/or induce apoptosis both in vivo and in vitro. Our goal for 2006 is to identify a
lead molecule and commence IND-enabling studies.
BUSINESS DEVELOPMENT STRATEGY
Oncology is EntreMeds principal clinical and commercial focus. Our scientific research,
however, has provided data that support the preclinical development of its compounds in certain
non-oncology applications. Our strategy is to continue developing compounds for oncology and
inflammatory diseases, while exploring strategic alliances selectively for its compounds in other
therapeutic areas.
At the appropriate time during clinical development, we may pursue co-development partners for
its core pipeline product candidates to help accelerate their development and strengthen the
development program with complementary expertise. Likewise, we can provide our co-development
partners with substantial know-how relating to small molecules, and peptides that inhibit
angiogenesis and inflammation, and regulate cell cycle pathways.
SUMMARY
Angiogenesis Expertise and Multiple Mechanism Pathways. We are developing product candidates
that act on multiple cellular pathways and biological processes associated with angiogenesis, cell
cycle regulation and inflammation. This approach has resulted in a focused pipeline with numerous
disease applications.
Oncology Focus with a Broad Therapeutic Potential. We are focused on oncology.
PanzemÒ NCD and MKC-1, our two lead oncology product candidates, are currently in
Phase 2 clinical studies in glioblastoma and metastatic breast cancer patients, respectively. We
anticipate that ENMD-1198 will enter a Phase 1 oncology clinical trial in the first half of 2006.
Recognizing the potential benefits of its drug candidates in other therapeutic areas, we are
developing products to treat inflammatory diseases, particularly rheumatoid arthritis. The
clinical development of other non-oncology applications will depend, directly or indirectly, on establishing alliances with selected
pharmaceutical and biotechnology companies.
7
Development Focus. We are continuing development of its pipeline product candidates,
specifically preclinical studies for 2ME2 in rheumatoid arthritis. Additionally, we will be moving
forward with the Panzem® NCD clinical program by continuing the Phase 1b studies to
assess food effects and a modified dosing schedule. Other key preclinical programs, such as aurora
kinase inhibitors, HDAC inhibitors, and tubulin inhibitors will focus on defining lead molecules
and therapeutic indications.
Commercialization Goal. Our goal is to commercialize our pipeline, led by PanzemÒ
NCD and MKC-1. We are committed to maintaining a balanced portfolio of oncology and
antiinflammatory compounds that can be co-developed with pharmaceutical/biotechnology partners, or
commercialized for our own account. We are committed to pursuing value-creating technologies and
products, making sound financial decisions, and building the financial capacity to develop our
clinical portfolio.
EMPLOYEES
As of December 31, 2005, we had 39 full-time employees and 2 part-time employees. 29 employees
work in our research and development department. We may hire additional personnel, in addition to
utilizing part-time or temporary consultants, on an as-needed basis. None of our employees are
represented by a labor union, and we believe our relations with our employees are satisfactory.
RELATIONSHIPS CORPORATE AND NON-PROFIT
Corporate Transactions.
| Celgene. In March 2005, we in-licensed Celgenes tubulin inhibitor program. We have assumed the responsibility for the preclinical and clinical development of tubulin inhibitors for oncology applications under this program. | ||
| Oxford BioMedica. In September 2005, we amended the agreement by which we licensed from Oxford the localized delivery of the endostatin and angiostatin genes for ophthalmological applications. The amendment extended the timelines for our achievement of certain milestones. Oxford granted this extension due to unforeseen technical difficulties that resulted in an increase in the amount of time required to complete several scientific objectives. | ||
| Childrens Medical Center Corporation (CMCC). As part of our three-way agreement with Alchemgen Therapeutics, Inc. executed in February 2004, CMCC holds the licenses for Endostatin and Angiostatin for all markets outside of Asia. | ||
| Affymax, Inc. In August 2005, we extended our research collaboration agreement with Affymax, Inc. to identify peptides and peptidic compounds for the treatment of cancer. A new research plan was executed outlining the next steps for development and both parties are actively engaged in negotiating a Joint Commercialization Agreement. |
Contract Manufacturing. The manufacturing efforts for the production of our clinical trial
materials are performed by contract manufacturing organizations. Established relationships coupled
with supply agreements have secured the necessary resources to ensure adequate supply of clinical
materials to support our clinical development program. We believe that our current strategy of
outsourcing manufacturing is cost-effective and allows for the flexibility we require.
2ME2 is currently bulk manufactured by Akzo Nobel and PanzemÒ NCD is
currently manufactured by Elan Drug Delivery, Inc. We do not anticipate any challenges in securing
contract manufacturing capacity at either of these facilities to produce PanzemÒ
NCD.
8
Sponsored Research Agreements. To complement our in-house research and development efforts,
we have entered into sponsored research agreements with outside scientists to conduct specific
projects as outlined below. Under these agreements, we have secured the rights to intellectual
property and to develop under exclusive license any discoveries resulting from these
collaborations. The funds we provide in accordance with these agreements partially support the
scientists laboratory, research personnel and research supplies.
Dalhousie University (Dr. Andrew Issekutz)
1-year agreement effective June 1, 2005
Investigation of Anti-arthritic Action of 2ME2
1-year agreement effective June 1, 2005
Investigation of Anti-arthritic Action of 2ME2
Purdue University (Dr. Mark Cushman)
2-year agreement effective December 1, 2005
Design and Synthesis of Tubulin Polymerization Inhibitors
2-year agreement effective December 1, 2005
Design and Synthesis of Tubulin Polymerization Inhibitors
University of California at Los Angeles (Dr. Benjamin Bonavida)
1-year agreement effective June 1, 2005
Molecular Mechanism of 2ME2 and 2ME2 Analog Induced Chemo/Immunosensitization of Drug
Resistant Multiple Myeloma Cell
Molecular Characterization of 2ME2 and 2ME2 Analog Mediated Up-Regulation of DR5
Expression and Sensitization to TRIAL-Induced Apoptosis in Prostate Cancer
1-year agreement effective June 1, 2005
Molecular Mechanism of 2ME2 and 2ME2 Analog Induced Chemo/Immunosensitization of Drug
Resistant Multiple Myeloma Cell
Molecular Characterization of 2ME2 and 2ME2 Analog Mediated Up-Regulation of DR5
Expression and Sensitization to TRIAL-Induced Apoptosis in Prostate Cancer
University of Colorado Health Sciences Center (Dr. Daniel Chan)
1-year agreement effective January 1, 2005
In vitro and in vivo Efficacy Studies of 2ME2 and its Analogs in Human Cancer Cells
1-year agreement effective January 1, 2005
In vitro and in vivo Efficacy Studies of 2ME2 and its Analogs in Human Cancer Cells
University of Paisley (United Kingdom)
3-year agreement ($30,000/year) to support post-doctoral fellow
Evaluation of PAR-2 Antagonist
3-year agreement ($30,000/year) to support post-doctoral fellow
Evaluation of PAR-2 Antagonist
Cooperative Research and Development Agreements (CRADAs). EntreMed extended its existing
CRADA with the National Cancer Institute:
| Preclinical and Clinical Development of 2ME2 (PanzemÒ) (Expires April 2007). |
EntreMed also executed a new CRADA #02067 with Dr. Robert Shoemaker of the National Cancer
Institute (NCI-Frederick) entitled:
| 2-Methoxyestradiol (2ME2) and 2ME2 Analogs as Inhibitors of Hypoxia Inducible Factor-1 alpha (HIF-1a). |
Clinical Trial Centers. As of March 10, 2006, we are conducting clinical trials at the
following institutions:
| Dana-Farber Cancer Institute | ||
| Duke University Medical Center | ||
| Indiana University Cancer Center | ||
| Mayo Clinic | ||
| Wisconsin Comprehensive Cancer Center |
PATENTS, LICENSES AND PROPRIETARY RIGHTS
Our success will depend in part on our ability to obtain patent protection for our products,
both in the United States and abroad. The patent position of biotechnology and pharmaceutical
companies, in general, is highly uncertain and involves complex legal and factual questions.
9
Following
the February 2004 transfer of the licenses for endostatin and
angiostatin back to Childrens
Hospital, Boston, we own or have licensed on an exclusive basis a total of 46 patent
applications and issued patents in the United States for our product candidates. We have a total
of 109 pending patent applications and issued patents in the United States and other countries.
We have exclusively licensed technology from Childrens Hospital, Boston, which covers the use
of steroid-derived small molecular weight compounds such as PanzemÒ that are
antimitotic and antiangiogenic agents. A U.S. patent application has been allowed covering purified
PanzemÒ as a composition of matter. There are six pending United States patent
applications and 13 allowed or issued United States patents covering PanzemÒ
technology. Patent applications also cover estrogen-related compounds with anti-fungal activity and
the treatment of localized atherosclerosis. The terms of the licenses for PanzemÒ
extend until the last underlying patent expires.
We own the technology associated with our 2ME2 analogs, PAR-2 inhibitors, TFPI peptides, and
NCEs for oncology and inflammation.
Many patent applications corresponding to the above-described United States patent
applications have been filed in Europe, Japan, Canada, Australia, and other selected countries.
We have registered the trademarks ENTREMED, PANZEM® and THE ANGIOGENESIS COMPANY in
the U.S. Patent and Trademark Office and have applied for registration of the marks in selected
foreign countries.
GOVERNMENT REGULATION
Our development, manufacture, and potential sale of therapeutics are subject to extensive
regulation by United States and foreign governmental authorities.
In the United States, the Food and Drug Administration (FDA) will regulate our product
candidates currently being developed as drugs or biologics. New drugs are subject to regulation
under the Federal Food, Drug, and Cosmetic Act (FFDCA), and biological products, in addition to
being subject to certain provisions of that Act, are regulated under the Public Health Service Act
(PHSA). We believe that the FDA is likely to regulate the products currently being developed by us
or our collaborators as new drugs. Both the FFDCA and PHSA and corresponding regulations govern,
among other things, the testing, manufacturing, safety, efficacy, labeling, storage, recordkeeping,
advertising and other promotion of biologics or new drugs, as the case may be. FDA clearances or
approvals must be obtained before clinical testing, and before manufacturing and marketing of
biologics or drugs.
Preparing drug candidates for approval has historically been a costly and time-consuming
process. Generally, in order to gain FDA permission to test a new agent, a developer first must
conduct preclinical studies in the laboratory and in animal model systems to gain preliminary
information on an agents effectiveness and to identify any safety problems. The results of these
studies are submitted as a part of an Investigational New Drug (IND) application for a drug or
biologic, which the FDA must review before human clinical trials of an investigational drug can
begin. In addition to the known safety and effectiveness data on the drug or biologic, the IND must
include a detailed description of the clinical investigations proposed to be undertaken. Based on
the current FDA organizational structure, PanzemÒ, 2ME2 analogs, and other
compounds in our small molecule programs are expected to be regulated as new drugs by the FDAs
Center for Drug Evaluation and Research (CDER). Generally, as new chemical entities like our small
molecules are discovered, formal IND-directed toxicology studies will be required prior to
initiating human testing. Clinical testing may begin 30 days after submission of an IND to the FDA
unless FDA objects to the initiation of the study, or at such earlier time as FDA expressly
permits.
In order to commercialize any drug or biological products, we or our collaborators must
sponsorand file an IND and conduct clinical studies to demonstrate the safety and effectiveness
necessary to obtain FDA approval of such products. For studies conducted under INDs sponsored by us
or our collaborators, we or our collaborators will be required to select qualified investigators
(usually physicians within medical institutions) to supervise the administration of the products,
test or otherwise assess patient results, and collect and maintain patient data; monitor
10
the investigations to ensure that they are conducted in accordance with applicable requirements,
including the requirements set forth in the general investigational plan and protocols contained
in the IND; and comply with applicable reporting and recordkeeping requirements.
Clinical trials of drugs or biologics are normally done in three phases, although the phases
may overlap. Phase 1 trials for agents to be used to treat cancer patients are concerned primarily
with the safety and preliminary effectiveness of the drug, involve a small group ranging from 15 -
40 subjects, and may take from six months to over one year to complete. Phase 2 trials normally
involve 30 200 patients and are designed primarily to demonstrate effectiveness in treating or
diagnosing the disease or condition for which the drug is intended, although short-term side
effects and risks in people whose health is impaired may also be examined. Phase 3 trials are
expanded clinical trials with larger numbers of patients which are intended to evaluate the overall
benefit-risk relationship of the drug and to gather additional information for proper dosage and
labeling of the drug. Phase III clinical trials generally take two to five years to complete, but
may take longer. The FDA receives reports on the progress of each phase of clinical testing, as
well as reports of unexpected adverse experiences occurring during the trial, and it may require
the modification, suspension, or termination of clinical trials, if it concludes that an
unwarranted risk is presented to patients, or, in Phase 2 and 3, if it concludes that the study
protocols are deficient in design to meet their stated objectives.
If clinical trials of a new product are completed successfully, the sponsor of the product may
seek FDA marketing approval. If the product is classified as a new drug, an applicant must file a
New Drug Application (NDA) with the FDA and receive approval before commercial marketing of the
drug. The NDA must include detailed information about the product and its manufacture and the
results of product development, preclinical studies and clinical trials. If the product is
classified as a biologic, an applicant must file a Biologies License Application (BLA) containing
similar information.
The testing and approval processes require substantial time and effort and there can be no
assurance that any approval will be obtained on a timely basis, if at all. Although it is the
policy of the FDA to complete the review of the initial submission of NDAs within six to twelve
months, the entire FDA review process may take several years to receive approval. Notwithstanding
the submission of relevant data, the FDA may ultimately decide that the NDA does not satisfy its
regulatory criteria and deny the approval. Further, the FDA may require additional clinical
studies before making a decision on approval. In addition, the FDA may condition marketing approval
on the conduct of specific post-marketing studies to further evaluate safety and effectiveness.
Even if FDA regulatory clearances are obtained, a marketed product is subject to continuing
regulatory requirements and review relating to GMP, adverse event reporting, promotion and
advertising, and other matters. Discovery of previously unknown problems or failure to comply with
the applicable regulatory requirements may result in restrictions on the marketing of a product or
withdrawal of the product from the market, as well as possible civil or criminal sanctions.
COMPETITION
Competition in the pharmaceutical, biotechnology and biopharmaceutical industries is intense
and based significantly on scientific and technological factors, the availability of patent and
other protection for technology and products, the ability and length of time required to obtain
governmental approval for testing, manufacturing and marketing and the ability to commercialize
products in a timely fashion. Moreover, the biopharmaceutical industry is characterized by rapidly
evolving technology that could result in the technological obsolescence of any products that we
develop.
We compete with many specialized biopharmaceutical firms, as well as a growing number of large
pharmaceutical companies that are applying biotechnology to their operations. Many
biopharmaceutical companies have focused their development efforts in the human therapeutics area,
and many major pharmaceutical companies have developed or acquired internal biotechnology
capabilities or made
commercial arrangements with other biopharmaceutical companies. These companies, as well as
academic institutions, governmental agencies and private research organizations, also compete with
us in recruiting and retaining highly qualified scientific personnel and consultants.
11
Our competition will be determined in part by the potential indications for which our product
candidates may be developed and ultimately approved by regulatory authorities. We may rely on third
parties to commercialize our products, and accordingly, the success of these products will depend
in significant part on these third parties efforts and ability to compete in these markets. The
success of any collaboration will depend in part upon our collaborative partners own competitive,
marketing and strategic considerations, including the relative advantages of alternative products
being developed and marketed by our collaborative partners and our competitors.
Many of our existing or potential competitors have substantially greater financial, technical
and human resources than we do and may be better equipped to develop, manufacture and market
products. In addition, many of these competitors have extensive experience in preclinical testing
and human clinical trials and in obtaining regulatory approvals. The existence of competitive
products, including products or treatments of which we are not aware, or products or treatments
that may be developed in the future, may adversely affect the marketability of products that we may
develop.
Available Information
Through our website at www.entremed.com, we make available, free of charge, our filings with
the Securities and Exchange Commission (SEC), including our annual proxy statements, annual
reports on Form 10-K, quarterly reports on Form 10-Q and current reports on Form 8-K, and all
amendments thereto, as soon as reasonably practicable after such reports are filed with or
furnished to the Securities and Exchange Commission. Our filings are also available through the
Securities and Exchange Commission via their website,
http://www.sec.gov. You may also read and
copy any materials we file with the SEC at the SECs Public Reference Room at 100 F Street, N.E.,
Washington, D.C. 20549. You may obtain information on the operation of the Public Reference Room
by calling the SEC at 1-800-SEC-0330. The information contained on our website is not incorporated
by reference in this annual report on Form 10-K and should not be considered a part of this report.
ITEM 1A. | RISK FACTORS. |
We Have a History of Losses and Anticipate Future Losses
To date, we have been engaged primarily in research and development activities. Although we
have received license fees and research and development funding from a former collaborator, limited
revenues on royalties from sales of Thalomid® and certain research grants, we have not derived
significant revenues from operations.
At December 31, 2005, we had an accumulated deficit of approximately $261,747,800. Losses
have continued since December 31, 2005. We will also be required to conduct substantial research
and development and clinical testing activities for our proposed products. We expect that these
activities will result in operating losses for the foreseeable future before we commercialize any
products, if ever. In addition, to the extent we rely on others to develop and commercialize our
products, our ability to achieve profitability will depend upon the success of these other parties.
To support our research and development of certain product candidates, we also may rely on
cooperative agreements from governmental and other organizations as a source of support. If our
cooperative agreements were to be reduced to any substantial extent, it may impair our ability to
continue our research and development efforts. Even if we do achieve profitability, we may be
unable to sustain or increase it.
Development of Our Products is at an Early Stage and is Uncertain
Our proposed products and research programs are in the early stage of clinical development and
require significant, time-consuming and costly research and development, testing and regulatory
clearances. In developing our products, we are subject to risks of failure that are inherent
in the development of products and therapeutic procedures. For example, it is possible that any or
all of our proposed products will be ineffective or toxic, or otherwise will fail to receive
necessary FDA clearances. There is a risk that the proposed products will be uneconomical to
manufacture or market or will not achieve market acceptance. There is also a risk that third
parties may hold proprietary rights that preclude us from marketing our proposed products or that
others will market a superior or equivalent product. Further, our research and development
activities might never result in commercially viable products.
12
Our product candidates are at the clinical and preclinical stages of development. Although
several of our product candidates have demonstrated some promising results in early clinical
(human) trials and preclinical (animal) studies, they may not prove to be effective in humans. For
example, testing on animals may occur under different conditions than testing in people and
therefore the results of animal studies may not accurately predict human experience. Likewise,
early clinical studies may not be predictive of eventual safety or effectiveness results in
larger-scale pivotal clinical trials.
There are many regulatory steps that must be taken before any of these product candidates will
be eligible for FDA approval and subsequent sale, including the completion of preclinical and
clinical trials. We do not expect that these product candidates will be commercially available for
several years, if ever.
We Are Uncertain Whether Additional Funding Will Be Available For Our Future Capital Needs and
Commitments
We will require substantial funds in addition to our existing working capital to develop our
product candidates and otherwise to meet our business objectives. We have never generated
sufficient revenue during any period since our inception to cover our expenses and have spent, and
expect to continue to spend, substantial funds to continue our research and development and
clinical programs. Any one of the following factors, among others, could cause us to require
additional funds or otherwise cause our cash requirements in the future to increase materially:
| results of research and development activities; | ||
| progress of our preclinical studies or clinical trials; | ||
| results of clinical trials; | ||
| changes in or terminations of our relationships with strategic partners; | ||
| changes in the focus, direction, or costs of our research and development programs; | ||
| competitive and technological advances; | ||
| establishment of marketing and sales capabilities; | ||
| manufacturing; | ||
| the regulatory approval process; or | ||
| product launch. |
We Must Show the Safety and Efficacy of Our Product Candidates Through Clinical Trials, the Results
of Which Are Uncertain
Before obtaining regulatory approvals for the commercial sale of our products, we must
demonstrate, through preclinical studies (animal testing) and clinical trials (human testing), that
our proposed products are safe and effective for use in each target indication. Testing of our
product candidates will be required, and failure can occur at any stage of testing. Clinical trials
may not demonstrate sufficient
safety and efficacy to obtain the required regulatory approvals or result in marketable
products. The failure to adequately demonstrate the safety and efficacy of a product under
development could delay or prevent regulatory approval of the potential product.
Clinical trials for the product candidates we are developing may be delayed by many factors,
including that potential patients for testing are limited in number. The failure of any clinical
trials to meet applicable regulatory standards could cause such trials to be delayed or terminated,
which could further delay the commercialization of
13
any of our product candidates. Newly emerging
safety risks observed in animal or human studies also can result in delays of ongoing or proposed
clinical trials. Any such delays will increase our product development costs. If such delays are
significant, they could negatively affect our financial results and the commercial prospects for
our products.
PanzemÒ NCD May Not be Successful
We have reformulated our lead product candidate, PanzemÒ, in order to
increase its concentration in the blood stream. Through the use of NanoCrystal® Colloidal
Dispersion (NCD), a proprietary technology of Elan Drug Delivery, Inc. (Elan), we have
reformulated PanzemÒ as an orally-administered liquid suspension. In February
2006, we commenced a Phase 2 study using PanzemÒ NCD in patients with glioblastoma
multiforme (GBM). Although PanzemÒ NCD showed increased levels in the blood in
Phase 1b clinical trials, it may not work as well in upcoming trials as it has in earlier testing.
We May Need New Collaborative Partners to Further Develop and Commercialize Products, and if We
Enter Into Such Arrangements, We May Give Up Control Over the Development and Approval Process and
Decrease Our Potential Revenue
We plan to develop and commercialize our product candidates with or without corporate
alliances and partners. Nonetheless, we intend to explore opportunities for new corporate alliances
and partners to help us develop, commercialize and market our product candidates. We expect to
grant to our partners certain rights to commercialize any products developed under these
agreements, and we may rely on our partners to conduct research and development efforts and
clinical trials on, obtain regulatory approvals for, and manufacture and market any products
licensed to them. Each individual partner will seek to control the amount and timing of resources
devoted to these activities generally. We anticipate obtaining revenues from our strategic partners
under such relationships in the form of research and development payments and payments upon
achievement of certain milestones. Since we generally expect to obtain a royalty for sales or a
percentage of profits of products licensed to third parties, our revenues may be less than if we
retained all commercialization rights and marketed products directly. In addition, there is a risk
that our corporate partners will pursue alternative technologies or develop competitive products as
a means for developing treatments for the diseases targeted by our programs.
We may not be successful in establishing any collaborative arrangements. Even if we do
establish such collaborations, we may not successfully commercialize any products under or derive
any revenues from these arrangements. Our strategy also involves entering into multiple, concurrent
strategic alliances to pursue commercialization of our core technologies. There is a risk that we
will be unable to manage simultaneous programs successfully. With respect to existing and potential
future strategic alliances and collaborative arrangements, we will depend on the expertise and
dedication of sufficient resources by these outside parties to develop, manufacture, or market
products. If a strategic alliance or collaborative partner fails to develop or commercialize a
product to which it has rights, we may not recognize any revenues on that particular product.
We Have No Current Manufacturing or Marketing Capacity and Rely on Only One Supplier For Some of
Our Products
We do not expect to manufacture or market products in the near term, but we may try to do so
in certain cases. We do not currently have the capacity to manufacture or market products and we
have limited experience in these activities. If we elect to perform these functions, we will be
required to either develop these capacities, or contract with others to perform some or all of
these tasks. We may be
dependent to a significant extent on corporate partners, licensees, or other entities for
manufacturing and marketing of products. If we engage directly in manufacturing or marketing, we
will require substantial additional funds and personnel and will be required to comply with
extensive regulations. We may be unable to develop or contract for these capacities when required
to do so in connection with our business.
We are currently manufacturing products for clinical trials on a contract basis.
PanzemÒ NCD, our lead small molecule clinical drug candidate, is currently
manufactured by Elan. We do not have arrangements in place with alternative suppliers if our
current supplier Elan was unable to deliver the product in necessary quantities.
14
We depend on our third-party manufacturers to perform their obligations effectively and on a
timely basis. These third parties may not meet their obligations and any such non-performance may
delay clinical development or submission of products for regulatory approval, or otherwise impair
our competitive position. Any significant problem experienced by one of our suppliers could result
in a delay or interruption in the supply of materials to us until such supplier resolves the
problem or an alternative source of supply is located. Any delay or interruption would likely lead
to a delay or interruption of manufacturing operations, which could negatively affect our
operations. Although we have identified alternative suppliers for our product candidates, we have
not entered into contractual or other arrangements with them. If we needed to use an alternate
supplier for any product, we would experience delays while we negotiated an agreement with them for
the manufacture of such product. In addition, we may be unable to negotiate manufacturing terms
with a new supplier that are as favorable as the terms we have with our current suppliers.
Problems with any manufacturing processes could result in product defects, which could require
us to delay shipment of products or recall products previously shipped. In addition, any prolonged
interruption in the operations of the manufacturing facilities of one of our sole-source suppliers
could result in the cancellation of shipments. A number of factors could cause interruptions,
including equipment malfunctions or failures, or damage to a facility due to natural disasters or
otherwise. Because our manufacturing processes are or are expected to be highly complex and subject
to a lengthy FDA approval process, alternative qualified production capacity may not be available
on a timely basis or at all. Difficulties or delays in our manufacturing could increase our costs
and damage our reputation.
The manufacture of pharmaceutical products can be an expensive, time consuming, and complex
process. Manufacturers often encounter difficulties in scaling-up production of new products,
including quality control and assurance and shortages of personnel. Delays in formulation and
scale-up to commercial quantities could result in additional expense and delays in our clinical
trials, regulatory submissions, and commercialization.
Failure of Manufacturing Facilities Producing Our Product Candidates to Maintain Regulatory
Approval Could Delay or Otherwise Hinder Our Ability to Market Our Product Candidates
Any manufacturer of our product candidates will be subject to applicable Good Manufacturing
Practices (GMP) prescribed by the FDA or other rules and regulations prescribed by foreign
regulatory authorities. We and any of our collaborators may be unable to enter into or maintain
relationships either domestically or abroad with manufacturers whose facilities and procedures
comply or will continue to comply with GMP and who are able to produce our small molecules in
accordance with applicable regulatory standards. Failure by a manufacturer of our products to
comply with GMP could result in significant time delays or our inability to obtain marketing
approval or, should we have market approval, for such approval to continue. Changes in our
manufacturers could require new product testing and facility compliance inspections. In the United
States, failure to comply with GMP or other applicable legal requirements can lead to federal
seizure of violated products, injunctive actions brought by the federal government, inability to
export product, and potential criminal and civil liability on the part of a company and its
officers and employees.
Manufacturing Our Product Candidates May Not Be Commercially Feasible
The manufacturing processes for several of the small molecules we are developing have not yet
been tested at commercial levels, and it may not be possible to manufacture these materials in a
cost-effective manner.
We Depend on Patents and Other Proprietary Rights, Some of Which are Uncertain
Our success will depend in part on our ability to obtain patents for our products, both in the
United States and abroad. The patent position of biotechnology and pharmaceutical companies in
general is highly uncertain and involves complex legal and factual questions. Risks that relate to
patenting our products include the following:
15
| our failure to obtain additional patents; | ||
| challenge, invalidation, or circumvention of patents already issued to us; | ||
| failure of the rights granted under our patents to provide sufficient protection; | ||
| independent development of similar products by third parties; or | ||
| ability of third parties to design around patents issued to our collaborators or us. |
For several of the products that we are developing, including Panzem®, composition of matter
patents are not available because the compounds are in the public domain. In these cases, only
patents covering the use of the product are available. In general, patents covering a new use for
a known compound can be more difficult to enforce against infringers of the use claims in the
patent.
Our potential products may conflict with patents that have been or may be granted to
competitors, universities or others. As the biotechnology industry expands and more patents are
issued, the risk increases that our potential products may give rise to claims that may infringe
the patents of others. Such other persons could bring legal actions against us claiming damages and
seeking to enjoin clinical testing, manufacturing and marketing of the affected products. Any such
litigation could result in substantial cost to us and diversion of effort by our management and
technical personnel. If any of these actions are successful, in addition to any potential liability
for damages, we could be required to obtain a license in order to continue to manufacture or market
the affected products. We may not prevail in any action and any license required under any needed
patent might not be made available on acceptable terms, if at all.
We are a party to sponsored research agreements and license agreements that require us to make
milestone payments upon attainment of certain regulatory milestones. Failure to meet such
milestones could result in the loss of certain rights to compounds covered under such license
agreements.
We also rely on trade secret protection for our confidential and proprietary information.
However, trade secrets are difficult to protect and others may independently develop substantially
equivalent proprietary information and techniques and gain access to our trade secrets and disclose
our technology. We may be unable to meaningfully protect our rights to unpatented trade secrets. We
require our employees to complete confidentiality training that specifically addresses trade
secrets. All employees, consultants, and advisors are required to execute a confidentiality
agreement when beginning an employment or a consulting relationship with us. The agreements
generally provide that all trade secrets and inventions conceived by the individual and all
confidential information developed or made known to the individual during the term of the
relationship automatically become our exclusive property. Employees and consultants must keep such
information confidential and may not disclose such information to third parties except in specified
circumstances. However, these agreements may not provide meaningful protection for our proprietary
information in the event of unauthorized use or disclosure of such information.
To the extent that consultants, key employees, or other third parties apply technological
information independently developed by them or by others to our proposed projects, disputes may
arise as
to the proprietary rights to such information. Any such disputes may not be resolved in our
favor. Certain of our consultants are employed by or have consulting agreements with other
companies and any inventions discovered by them generally will not become our property.
Our Potential Products Are Subject to Government Regulatory Requirements and an Extensive Approval
Process
Our research, development, preclinical and clinical trials, manufacturing, and marketing of
most of our product candidates are subject to an extensive regulatory approval process by the FDA
and other regulatory agencies in the United States and abroad. The process of obtaining FDA and
other required regulatory approvals for drug and biologic products, including required preclinical
and clinical testing, is time consuming and expensive. Even after
16
spending time and money, we may
not receive regulatory approvals for clinical testing or for the manufacturing or marketing of any
products. Our collaborators or we may encounter significant delays or costs in the effort to secure
necessary approvals or licenses. Even if we obtain regulatory clearance for a product, that product
will be subject to continuing review. Later discovery of previously unknown defects or failure to
comply with the applicable regulatory requirements may result in restrictions on a products
marketing or withdrawal of the product from the market, as well as possible civil or criminal
penalties.
Potential Products May Subject Us to Product Liability for Which Insurance May Not Be Available
The use of our potential products in clinical trials and the marketing of any pharmaceutical
products may expose us to product liability claims. We have obtained a level of liability insurance
coverage that we believe is adequate in scope and coverage for our current stage of development.
However, our present insurance coverage may not be adequate to protect us from liabilities we might
incur. In addition, our existing coverage will not be adequate as we further develop products and,
in the future, adequate insurance coverage and indemnification by collaborative partners may not be
available in sufficient amounts or at a reasonable cost. If a product liability claim or series of
claims are brought against us for uninsured liabilities, or in excess of our insurance coverage,
the payment of such liabilities could have a negative effect on our business and financial
condition.
ITEM 1B. | UNRESOLVED STAFF COMMENTS. |
None.
ITEM 2. | PROPERTIES. |
We currently lease approximately 46,000 square feet of space (approximately 32,000 square feet
of which is laboratory space) in Rockville, Maryland. The lease expires in February 2009. We
believe that our existing facilities will be adequate to accommodate the implementation of our
current business plan.
ITEM 3. | LEGAL PROCEEDINGS. |
EntreMed is subject in the normal course of business to various legal proceedings in which
claims for monetary or other damages may be asserted. Management does not believe such legal
proceedings, except as otherwise disclosed herein, are material.
ITEM 4. | SUBMISSION OF MATTERS TO A VOTE OF SECURITY HOLDERS. |
None.
17
\
PART II
ITEM 5. | MARKET FOR REGISTRANTS COMMON EQUITY RELATED STOCKHOLDER MATTERS AND ISSUER PURCHASES OF EQUITY SECURITIES. |
Our common stock began trading publicly on the Nasdaq National Market under the symbol ENMD
on June 12, 1996. The following table sets forth the high and low closing price for our common
stock by quarter, as reported by the Nasdaq National Market, for the periods indicated:
HIGH | LOW | |||||||
2004: |
||||||||
First Quarter |
$ | 4.37 | $ | 3.09 | ||||
Second Quarter |
4.01 | 2.01 | ||||||
Third Quarter |
2.08 | 1.28 | ||||||
Fourth Quarter |
3.43 | 1.75 | ||||||
2005: |
||||||||
First Quarter |
$ | 4.64 | $ | 2.10 | ||||
Second Quarter |
3.11 | 2.19 | ||||||
Third Quarter |
3.31 | 2.30 | ||||||
Fourth Quarter |
2.52 | 1.88 | ||||||
2006: |
||||||||
First Quarter (through March 7, 2006) |
$ | 2.52 | $ | 1.99 |
On March 7, 2006, the closing price of our common stock, as reported by the Nasdaq
National Market, was $2.35 per share. As of March 7, 2006 there
were approximately 935 holders of
record of our common stock.
Since our initial public offering in 1996, we have not paid cash dividends on our common
stock. We currently anticipate that any earnings will be retained for the continued development of
our business and we do not anticipate paying any cash dividends on our common stock in the
foreseeable future.
18
ITEM 6. | SELECTED FINANCIAL DATA. |
The following selected consolidated financial data set forth below has been derived from our
audited consolidated financial statements. The data should be read in conjunction with the
consolidated financial statements and related notes, Managements Discussion and Analysis of
Financial Condition and Results of Operations in Item 7 and other financial information included
elsewhere in this annual report on
Form 10-K.
Year Ended December 31, | ||||||||||||||||||||
STATEMENTS OF OPERATIONS DATA: | 2005 | 2004 | 2003 | 2002 | 2001 | |||||||||||||||
Revenues |
||||||||||||||||||||
Collaborative research
and development |
$ | | $ | | $ | 667,796 | $ | 835,493 | $ | | ||||||||||
License fees |
590,992 | 495,496 | 310,496 | 115,496 | | |||||||||||||||
Grant revenues |
| | 508,243 | 131,681 | 358,427 | |||||||||||||||
Royalty revenues |
5,310,439 | 5,918 | 2,705 | 38,790 | 1,440,070 | |||||||||||||||
Other |
16,624 | 12,581 | 86,306 | 55,030 | 63,444 | |||||||||||||||
Total revenues |
5,918,055 | 513,995 | 1,575,546 | 1,176,490 | 1,861,941 | |||||||||||||||
Expenses: |
||||||||||||||||||||
Research and development |
17,325,048 | 10,523,252 | 14,252,196 | 31,308,427 | 54,201,179 | |||||||||||||||
General and administrative |
5,920,455 | 6,570,664 | 7,022,986 | 13,932,133 | 14,473,012 | |||||||||||||||
Interest expense |
| | | 390,941 | 344,969 | |||||||||||||||
Investment income |
(1,010,771 | ) | (313,940 | ) | (205,580 | ) | (317,910 | ) | (1,437,966 | ) | ||||||||||
Gain on discharge of liabilities |
| | | (2,174,765 | ) | | ||||||||||||||
Gain on sale of asset |
(3,420 | ) | (124,083 | ) | | (2,940,184 | ) | | ||||||||||||
Gain on sale of securities |
| (520,000 | ) | | | | ||||||||||||||
Gain on sale of royalty interest |
| (3,000,000 | ) | | | (22,410,182 | ) | |||||||||||||
Net loss |
$ | (16,313,257 | ) | $ | (12,621,898 | ) | $ | (19,494,056 | ) | $ | (39,022,152 | ) | $ | (43,309,071 | ) | |||||
Dividends on Series A convertible preferred
stock |
( 1,005,000 | ) | ( 1,005,000 | ) | ( 1,005,000 | ) | | | ||||||||||||
Net loss attributable to common shareholders |
$ | (17,318,257 | ) | $ | (13,626,898 | ) | $ | (20,499,056 | ) | $ | (39,022,152 | ) | $ | (43,309,071 | ) | |||||
Net loss per share |
$ | (0.36 | ) | $ | (0.37 | ) | $ | (0.68 | ) | $ | (1.78 | ) | $ | (2.39 | ) | |||||
Weighted average number of
shares outstanding |
48,176,914 | 37,170,544 | 29,943,161 | 21,892,520 | 18,093,174 |
As of December 31, | ||||||||||||||||||||
2005 | 2004 | 2003 | 2002 | 2001 | ||||||||||||||||
BALANCE SHEET DATA: |
||||||||||||||||||||
Cash and cash equivalents
and short-term investments |
$ | 30,082,388 | $ | 34,539,516 | $ | 36,941,430 | $ | 24,067,045 | $ | 41,386,300 | ||||||||||
Working capital |
28,510,176 | 34,979,936 | 33,405,818 | 7,716,002 | 21,257,950 | |||||||||||||||
Total assets |
36,431,885 | 39,404,002 | 40,153,764 | 27,810,212 | 46,218,450 | |||||||||||||||
Deferred revenue, less
current portion |
| 95,496 | 192,993 | 286,488 | | |||||||||||||||
Accumulated deficit |
(261,747,829 | ) | (245,434,572 | ) | (232,812,674 | ) | (213,318,618 | ) | (174,296,466 | ) | ||||||||||
Total stockholders equity |
29,369,190 | 35,704,754 | 34,858,883 | 10,493,646 | 23,194,898 |
19
ITEM 7. | MANAGEMENTS DISCUSSION AND ANALYSIS OF FINANCIAL CONDITION AND RESULTS OF OPERATIONS. |
The following discussion should be read in conjunction with the Consolidated Financial
Statements and Notes thereto appearing elsewhere in this report. See Risk Factors.
OVERVIEW
Since our inception in September 1991, we have devoted substantially all of our efforts and
resources to sponsoring and conducting research and development on our own behalf and through
collaborations. Through December 31, 2005, all of our revenues have been generated from license
fees, research and development funding, royalty payments, the sale of royalty rights and certain
research grants; we have not generated any revenue from direct product sales. We anticipate our
primary revenue sources for the next few years will include research grants, royalties and
collaboration payments under current or future arrangements. The timing and amounts of such
revenues, if any, will likely fluctuate and depend upon the achievement of specified research and
development milestones. Results of operations for any period may be unrelated to the results of
operations for any other period.
Historically our research and development efforts have been focused on the identification and
development of new compounds for the treatment of certain diseases utilizing our understanding of
the interrelationships of angiogenesis, cell cycle regulation, and inflammation processes vital
to the treatment of multiple diseases, including cancer. In 2005, our main focus was on advancing
the reformulated 2ME2, our lead drug candidate Panzem®NCD, through Phase 1b clinical
trials and also moving ENMD-1198, an internally discovered novel tubulin binding agent, to an IND
filing. The goal of our reformulation effort was to increase the level of Panzem®in
the patients bloodstream. Based on the results of two Phase 1b clinical studies conducted in
2005, we have achieved the initial goal and have selected a dose and schedule for Phase 2 trials
with Panzem®NCD which began in January 2006. We are also evaluating Panzem®in preclinical development for rheumatoid arthritis.
In addition to our work with Panzem®, we evaluated various analogs of 2ME2 in
preclinical studies. One of these compounds, ENMD-1198, progressed into IND-directed development
and an IND was filed with and accepted by the Food and Drug Administration. Clinical trials in
oncology patients with ENMD-1198 are expected to be initiated during the first quarter of 2006.
In January 2006, we announced the acquisition of Miikana Therapeutics Inc., a clinical-stage
biopharmaceutical company. Through the transaction, we enhanced our clinical and preclinical
pipeline with the acquisition of MKC-1, a Phase 2 cell cycle inhibitor and two preclinical
programs, one in aurora kinase inhibition and one in HDAC inhibition.
We have three additional programs with compounds in various stages of discovery and
preclinical research. Our expenses will exceed our revenues as we continue the development of
Panzem® and bring our other drug candidates through preclinical research to clinical
trials.
With Panzem®NCD and MKC-1 in Phase 2 clinical trials and ENMD-1198 heading into
the clinic, we are continuing our progression from a fundamentally research organization to that of
a product development and commercialization organization. We have de-emphasized early discovery
activities and we continue to devote resources to key preclinical development programs to advance
these programs towards IND. We may also continue to seek product acquisitions, co-development
alliances and in-licensing opportunities to attempt to build a broader portfolio of late
preclinical and clinical product candidates.
CRITICAL ACCOUNTING POLICIES AND THE USE OF ESTIMATES
The preparation of our financial statements in conformity with accounting principles generally
accepted in the U.S. requires management to make estimates and assumptions that affect the amounts
reported in our
consolidated financial statements and accompanying notes. Actual results could differ
materially from those estimates. Our critical accounting policies, including the items in our
financial statements requiring significant estimates and judgments, are as follows:
20
- | Revenue Recognition The Company recognizes revenue in accordance with the provisions of Staff Accounting Bulletin No. 104, Revenue Recognition, whereby revenue is not recognized until it is realized or realizable and earned. Revenue is recognized when all of the following criteria are met: persuasive evidence of an arrangement exists, delivery has occurred or services have been rendered, the price to the buyer is fixed and determinable and collectibility is reasonably assured. |
- | Royalty Revenue Royalties from licenses are based on third-party sales and recorded as earned in accordance with contract terms, when third-party results are reliably measured and collectibility is reasonably assured. The majority of our 2005 revenues were from royalties on the sale of Thalomid®, which we began to recognize in the third quarter. In 2004, certain provisions of a purchase agreement dated June 14, 2001 by and between Bioventure Investments kft (Bioventure) and the Company were satisfied and, as a result, beginning in 2005, we are entitled to share in the royalty payments received by Royalty Pharma Finance Trust, successor to Bioventure, on annual Thalomid® sales above a certain threshold. Based on the licensing agreement royalty formula, annual royalty sharing commences with Thalomid® annual sales of approximately $225 million. The Company is also eligible to receive royalty payments under a February 2004 agreement with Childrens Medical Center Corporation (CMCC) and Alchemgen Therapeutics. Under the agreement, Alchemgen received rights to market endostatin and angiostatin in Asia. Future royalty payments, if any, will be recorded as revenue when received and/or when collectibility is reasonably assured. No royalties under this agreement have been earned to date. | ||
- | Licensing Revenue The Company has recognized licensing revenues resulting from the January 2002 five-year strategic alliance with Allergan to develop and commercialize small molecule angiogenic inhibitors for treatment and prevention of diseases and conditions of the eye. The initial net fee was being amortized to revenue over the five-year license term. In April 2005, Allergan terminated the license in accordance with its terms, which resulted in the acceleration of deferred revenue in the three-month period ended June 30, 2005. In February 2004, the Company transferred rights to the proteins, endostatin and angiostatin pursuant to an agreement with CMCC and Alchemgen Therapeutics. Under the agreement, the Company received an upfront and a second cash payment. The upfront licensing cash payment was fully amortized in 2004, as the Company had completed its obligations to transfer data and material. Due to rights negotiations between the licensee and CMCC, the second and final licensing cash payment in the amount of $400,000 was received in May 2005. Management concluded collectibility was not reasonably assured until the funds were received. In September 2003, the Company entered into a licensing agreement with Oxford Biomedica, PLC and Oxford Biomedica (UK) Limited for the use of endostatin and angiostatin genes in the development of locally delivered gene therapy for ophthalmologic applications. Under the agreement, the Company had no continuing obligations. As such, we recorded as revenue the value of the initial net cash and shares of common stock received under the agreement. | ||
- | Collaborative Research and Development Revenue In 2003 we received revenues for performance under commercial research and development contracts. These contracts required us to provide services directed toward specific objectives and include developmental milestones and deliverables. These revenues were recognized at the time that research and development activities were performed. | ||
- | Grant Revenue In 2003 we received a government grant to support financially our Phase 2 endostatin clinical trial in patients with neuroendocrine tumors. Grants are funded in specific amounts based on funding requests submitted to the grantor. Grant revenues are recognized and realized at the time that research and development activities are performed. |
21
- | Research and Development Research and development expenses consist primarily of compensation and other expenses related to research and development personnel, research collaborations, costs associated with pre-clinical testing and clinical trials of our product candidates, including the costs of manufacturing the product candidates, and facilities expenses. Research and development costs are expensed as incurred. | ||
- | Stock-Based Compensation We have stock option plans under which options to purchase shares of our common stock may be granted to employees, consultants and directors at a price no less than the fair market value on the date of grant. We account for our stock-based compensation in accordance with the provisions of APB No. 25, Accounting for Stock Issued to Employees (APB No. 25). Under APB No. 25, compensation expense is based on the difference, if any, on the date of the grant between the fair value of the Companys stock and the exercise price of the option and is recognized ratably over the vesting period of the option. Because our options must be granted at no less than fair market value, we recognize no compensation expense in accordance with APB No. 25. If we were to adopt SFAS No. 123, Accounting for Stock-Based Compensation (SFAS No. 123), we would recognize compensation expense based upon the fair value at the grant date for awards under the plans using the fair value method. The Company currently expects to adopt SFAS No. 123R in the quarter ended March 31, 2006, using the modified prospective method, although the Company continues to review its options for adoption under this new pronouncement. Please refer to footnote 1 of the footnotes to our audited financial statements included elsewhere herein for additional information. We expense equity instruments issued to nonemployees in accordance with EITF 96-18, Accounting for Equity Instruments that are issued to other than employees for acquiring, or in conjunction with selling goods or services. |
RESULTS OF OPERATIONS
Years Ended December 31, 2005, 2004 and 2003.
Revenues. Revenues increased 1,051% in 2005 to $5,918,000 from $514,000 in 2004 after
decreasing 67% in 2004 from $1,576,000 in 2003. The fluctuation between periods results from
changes in the revenue components and also from the manner and timing of when certain revenues are
recorded. The three years presented reflect the following revenue types: royalty revenues,
licensing revenues, collaborative research and development revenues and grant revenues. The
significant increase in revenues in 2005 results from the recognition of royalty revenue earned on
sales of Thalomid®. As discussed above, beginning in 2005, we are entitled to share in
the royalty payments received by Royalty Pharma Finance Trust on annual Thalomid® sales
above approximately $225 million. Thalomid® sales in 2005 surpassed the sharing point
in the third quarter and we recorded estimated royalty revenues of $5,310,000. No royalty revenues
were recorded on Thalomid® sales in 2004 or 2003.
Licensing revenues increased to $591,000 in 2005 from $495,000 in 2004. The 2004 amount
was an increase from $310,000 in 2003. The 2005 amount
reflects the accelerated
recognition of deferred licensing revenues from the January 2002 agreement with Allergan,
which was terminated in April 2005 by Allergan in accordance with the terms of the agreement,
and the recognition of a $400,000 licensing payment from Alchemgen in May 2005. This amount was
recorded as revenue when collectibility was deemed to be reasonably assured. The
increase in 2004 from 2003 was attributable to the recognition of amortized licensing revenues from
the February 2004 agreement with Alchemgen in addition to the revenues from the January 2002
five-year strategic alliance with Allergan. The 2003 licensing revenue was comprised of the
amortization of the upfront licensing fee from Allergan coupled with recognition of the value of
cash and stock received from Oxford Biomedica as revenue.
Collaborative research and development revenues of $668,000 received in 2003 represent work
under an NIH sponsored contract, which was completed in 2003. We did not receive collaborative
research and development revenues in 2005 or 2004. We also did not recognize grant revenue in 2005
or 2004. We recognized grant revenues of $508,000 in 2003, which represented a grant sponsored by
the FDA that provided financial support for our Phase 2 endostatin clinical trial in patients with
neuroendocrine tumors.
22
Research and Development Expenses. At December 31, 2005, accumulated direct project expenses
for Panzem®, our lead drug candidate, totaled $35,295,000. Reflected in our 2005 R&D
expenses totaling $17,325,000 are direct project expenses for Panzem® of $7,594,000 and
$3,237,000 related to our 2ME2 analog program. The balance of our R&D expenditures includes
facilities costs and other departmental overhead, and expenditures related to the advancement of
our pre-clinical pipeline. The 2005 R&D expenditures reflect an increase in clinical and
regulatory activity along with associated contract manufacturing activities. Panzem®
NCD completed two Phase 1B oncology trials and moved into Phase 2 oncology trials in January 2006.
IND-directed toxicity studies for ENMD-1198 were conducted and an IND was submitted and accepted in
late 2005. ENMD-1198, a tubulin binding agent, will commence clinical development with the
initiation of clinical trials in oncology in 2006. In 2005, we also recorded increased contract
manufacturing expenses. These expenses included material to support 2005 activities and also the
acquisition of API to support Panzem® and ENMD-1198 clinical programs in 2006. In 2005,
we also incurred formulation costs for finished drug product for both candidates
including certain Panzem® NCD Contract Manufacturing milestones triggered by clinical
events.
Research and Development expenses for 2004 totaled $10,523,000, including direct project costs
for Panzem® of $3,558,000 and for our 2ME2 analog program of $2,135,000. The higher
costs recorded for Panzem® reflect reformulation activities including material
acquisition, animal testing and sample analysis. The significant decrease in overall research and
development spending in 2004 and 2003 reflect the Companys shift in focus to small molecules and
the resulting dramatic decrease in costs associated with endostatin and angiostatin. Research and
Development expenses were $14,252,000 in 2003. The 2003 amount includes project costs for
Panzem®, endostatin and angiostatin of $6,328,000, $1,107,000 and $808,000,
respectively.
The expenditures that will be necessary to execute our business plan are subject to numerous
uncertainties, which may adversely affect our liquidity and capital resources. As of December 31,
2005, our proprietary product candidate, Panzem®, is in Phase 1 and Phase 2 clinical
trials in a capsule formulation and additionally in Phase 1b clinical trials in a liquid
formulation. Early in 2006, we announced that the liquid formulation, Panzem® NCD, had
advanced into Phase 2 trials. We expect our R&D expenses to trend higher reflecting the costs of
supporting multiple Phase 2 trials including the costs of securing clinical drug supply.
Completion of clinical trials may take several years or more, but the length of time generally
varies substantially according to the type, complexity, novelty and intended use of a product
candidate.
We estimate that clinical trials of the type we generally conduct are typically completed over
the following timelines:
ESTIMATED | ||
COMPLETION | ||
CLINICAL PHASE | PERIOD | |
Phase I
|
1 Year | |
Phase II
|
1-2 Years | |
Phase III
|
2-4 Years |
The duration and the cost of clinical trials may vary significantly over the life of a project
as a result of differences arising during the clinical trial protocol, including, among others, the
following:
- | the number of patients that ultimately participate in the trial; | ||
- | the duration of patient follow-up that seems appropriate in view of the results; | ||
- | the number of clinical sites included in the trials; and | ||
- | the length of time required to enroll suitable patient subjects. |
We test our potential product candidates in numerous pre-clinical studies to identify
indications for which they may be product candidates. We may conduct multiple clinical trials to
cover a variety of indications for each product candidate. As we obtain results from trials, we
may elect to discontinue clinical trials for certain product
candidates or for certain indications in order to focus our resources on more promising
product candidates or indications.
23
Our proprietary product candidates also have not yet achieved FDA regulatory approval, which
is required before we can market them as therapeutic products. In order to proceed to subsequent
clinical trial stages and to ultimately achieve regulatory approval, the FDA must conclude that our
clinical data establish safety and efficacy. Historically, the results from preclinical testing and
early clinical trials have often not been predictive of results obtained in later clinical trials.
A number of new drugs and biologics have shown promising results in clinical trials, but
subsequently failed to establish sufficient safety and efficacy data to obtain necessary regulatory
approvals.
An important element of our business strategy is to pursue the research and development of a
range of product candidates for a variety of oncology and non-oncology indications. This allows us
to diversify the risks associated with our research and development expenditures. As a result, we
intend to pursue development of our existing product candidates internally or through development
partnerships, as well as through the acquisition and subsequent development of promising
candidates. The goal is to align our future capital requirements with multiple product candidates
and to increase the likelihood that our future financial success is not substantially dependent on
any one product candidate. To the extent we are unable to maintain a broad range of product
candidates, our dependence on the success of one or a few product candidates would increase.
Furthermore, our business strategy includes the option of entering into collaborative
arrangements with third parties to complete the development and commercialization of our products.
In the event that third parties take over the clinical trial process for one of our product
candidates, the estimated completion date would largely be under the control of that third party
rather than us. We cannot forecast with any degree of certainty which proprietary products or
indications, if any, will be subject to future collaborative arrangements, in whole or in part, and
how such arrangements would affect our capital requirements.
As a result of the uncertainties discussed above, among others, we are unable to estimate the
duration and completion costs of our research and development projects. Our inability to complete
our research and development projects in a timely manner or our failure to enter into collaborative
agreements, when appropriate, could significantly increase our capital requirements and could
adversely impact our liquidity. These uncertainties could force us to seek additional, external
sources of financing from time to time in order to continue with our business strategy. There can
be no assurance that we would be able to raise additional capital if needed. Our inability to
raise additional capital, or to do so on terms reasonably acceptable to us, would jeopardize the
future success of our business.
Research and development expenses consist primarily of compensation and other expenses related
to research and development personnel, research collaborations, costs associated with internal and
contract pre-clinical testing and clinical trials of our product candidates, including the costs of
manufacturing the product candidates, and facilities expenses. Overall research and development
expenses increased to approximately $17,325,000 in 2005 from $10,523,000 and decreased to
$10,523,000 in 2004 from $14,252,000 in 2003. The increase in overall research and development
spending in 2005 is due to increased clinical and regulatory activity along with associated
contract manufacturing activities. Panzem® NCD completed enrollment in two Phase 1B
oncology trials and moved into Phase 2 oncology trials in January 2006. IND-directed toxicity
studies for ENMD-1198 were conducted and an IND was submitted and accepted in late 2005. ENMD-1198,
a tubulin binding agent, will commence clinical development with the initiation of clinical trials
in oncology in 2006. In 2005 we also recorded increased contract manufacturing expenses. These
expenses include material to support 2005 activities and also the acquisition of API to support
Panzem® and ENMD-1198 clinical programs in 2006. In 2005 we also incurred formulation
costs for finished drug product for both candidates including certain Panzem® NCD
contract manufacturing milestones triggered by clinical events. The 2005 amount also reflects
increased costs associated with further development of various drug candidates, including a $1
million upfront fee associated with the license of Celgenes small molecule tubulin inhibitor
compounds for the treatment of cancer and a $200,000 success fee to Ferghana Partners, Inc., a
related party, in connection with the license agreement. The increase in R&D expenses
reflected in 2005 and the decrease in 2004 was specifically impacted by the following:
24
- | Outside Services We utilize outsourcing to conduct our product development activities. Larger-scale small molecule synthesis, in vivo testing and data analysis are examples of the services that we outsource. We expended $2,399,000 in 2005, $1,741,000 in 2004 and $2,357,000 in 2003 on these activities. The 2005 increase resulted from continued development work on our preclinical pipeline programs, including IND-directed toxicity studies for ENMD-1198. The higher 2003 expenses, as compared to 2004, reflect our focus on reformulating Panzem® while our 2004 efforts had been directed towards optimizing Panzem® NCD and the selection of our lead 2ME2 analog candidate ENMD-1198. | ||
- | Collaborative Research Agreements We made payments to our collaborators of $673,000, $622,000 and $170,000 in years 2005, 2004 and 2003, respectively. Sponsored research payments to academic collaborators include payments to Childrens Hospital of $225,000 in 2005, $300,000 in 2004, and $75,000 in 2003. Our collaborative efforts are primarily directed towards further exploration of 2ME2 mechanism-of-action (MOA) in-vivo testing of therapeutic combination studies, and PanzemÒ non-oncology applications. | ||
- | Clinical Trial Costs Clinical costs increased from $1,008,000 in 2004 to $1,090,000 in 2005 versus the decrease in clinical costs from $1,432,000 in 2003 to $1,008,000 in 2004. The 2005 amount reflects two Phase 1b clinical trials for the reformulated PanzemÒ NCD. The prior year amount reflects a Phase Ia clinical trial to test various dosing approaches for reformulated PanzemÒ along with continuing clinical trials of the capsule Panzem® formulation. Costs of such trials include the clinical site fees, monitoring costs and data management costs. In addition, we contribute PanzemÒ clinical material in capsule form under our Collaborative Research and Development Agreement (CRADA) to the NCI, which is conducting trials with EntreMed. The 2004 decrease reflects that we were no longer responsible for supporting the endostatin and angiostatin trials as a result of the February 2004 licensing agreement with Alchemgen. | ||
- | Contract Manufacturing Costs The costs of manufacturing the material used in clinical trials for our product candidates is reflected in contract manufacturing. These costs include bulk manufacturing, formulation, encapsulation and fill finish services, product release costs and also payments to contract manufacturers for technology access or licensing fees. Contract manufacturing costs increased significantly in 2005 to $5,606,000. The 2005 costs include material to support 2005 activities and also the acquisition of API to support PanzemÒ and ENMD-1198 clinical programs in 2006. In 2005, we also incurred formulation costs for finished drug product for both candidates including certain PanzemÒ NCD contract manufacturing milestones triggered by clinical events. Product manufacturing costs were $1,392,000 and $3,012,000 in 2004 and 2003, respectively. The 2004 costs reflect expenditures for the preparation of Panzem® NCD for preclinical and clinical use and encapsulation runs for the capsule form to support our Phase 2 and NCI clinical trials. The 2003 costs include the acquisition of bulk material and multiple encapsulation runs for the capsule form of Panzem®. | ||
- | Personnel Costs The 2005 and 2004 personnel costs were approximately $3,000,000, down slightly from $3,212,000 in 2003. |
Also reflected in our 2005 research and development expenses are patent costs of $654,000,
facility and related expenses of $1,351,000, laboratory supplies and animal costs of $772,000,
consulting fees of $296,000 and travel expenses of $200,000. In 2004, these expenses totaled
$493,000, $1,551,000, $531,000, $174,000 and $69,000, respectively, and in 2003, these expenses
totaled $906,000, $1,916,000, $374,000, $211,000 and $96,000, respectively. The 2005 increase in
patent costs relates primarily to our preclinical pipeline. The 2004 decrease in patent expense
reflects the transfer of responsibility for endostatin and angiostatin .
General and Administrative Expenses. General and administrative expenses include compensation
and other expenses related to finance, business development and administrative personnel,
professional services and facilities.
25
General and administrative expenses decreased to approximately $5,920,000 in 2005 from
$6,571,000 in 2004 and $7,023,000 in 2003. The overall 2005 decrease primarily reflects the
reduction in legal and professional services related to compliance with provisions of the
Sarbanes-Oxley Act of 2002 relating to documentation, review and audit of internal control over
financial reporting, as compared to the amount incurred in 2004, the first year we were required to
comply with these provisions. General and administrative costs were lower in 2004 than in the
preceding year due to reduced personnel costs resulting from executive management changes, lower
severance obligations and the recording in 2003 of costs associated with settling certain disputes.
These lower expenditures in 2004 were partially offset by increased expenditures for professional
services related to compliance with the internal control over financial reporting provisions of the
Sarbanes-Oxley Act of 2002.
Interest expense. The Company had no interest-bearing debt during 2005, 2004 or 2003.
Investment income. Investment income increased by 222% in 2005 to $1,011,000 as a result of
higher yields on interest bearing cash accounts and investments. Investment income was $314,000 in
2004, an increase of 52% from $206,000 in 2003.
Dividends on Series A convertible preferred stock. The Consolidated Statements of Operations
for the years ended December 31, 2005, 2004 and 2003 reflect dividends of $1,005,000 relating to
Series A Convertible Preferred Stock held by Celgene pursuant to a Securities Purchase Agreement
dated December 31, 2002. The holders of Series A Preferred Stock will accumulate dividends at a
rate of 6% and will participate in dividends declared and paid on the common stock, if any. All
accumulated dividends must be paid before any dividends may be declared or paid on the Common
Stock. The Company has no plans to pay any dividends in the foreseeable future.
Gain on sale of securities. The Consolidated Statement of Operations for the year ended
December 31, 2004 reflects a gain of $520,000 resulting from the sale of certain securities of an
independent private biotechnology company. The securities were acquired in 1996 through 1999 and
accounted for using the equity method. The cost of these securities was written off in prior
periods and we had no residual cost basis in the securities when sold. As such, we have recorded a
gain on the sale equal to the sale proceeds.
Gain on sale of royalty interest. The Consolidated Statement of Operations for the year
ended December 31, 2004 reflects a gain of $3,000,000, which represents a one-time sale price
adjustment pursuant to a purchase agreement dated August 6, 2001 by and between Bioventure and the
Company. The adjustment was triggered by $800,000,000 in cumulative Thalomid® sales
through December 31, 2004.
LIQUIDITY AND CAPITAL RESOURCES
To date, we have been engaged primarily in research and development activities. As a result,
we have incurred and expect to continue to incur operating losses for 2006 and the foreseeable
future before we commercialize any products. Under the terms of the license agreement for 2ME2, we
must be diligent in bringing potential products to market and may be required to make future
milestone payments of up to $850,000. In addition, in March 2005, we in-licensed Celgenes small
molecule tubulin inhibitor compounds for the treatment of cancer. Under the terms of the agreement,
Celgene received an upfront licensing fee and may receive additional payments up to approximately
$25.25 million based upon the attainment of certain milestones. If we fail to comply with the
milestones or fail to make any required sponsored research or milestone payment, we could face the
termination of the relevant license agreement. As of December 31, 2005, none of these milestones
have been reached and, as such, there are no payments due.
At December 31, 2005, we had cash and short-term investments of approximately $30,082,388 with
working capital of approximately $28,510,176. In addition, we announced in January 2006 the
acquisition of Miikana Therapeutics in an all-stock transaction, and in February 2006, the
completion of the private placement of 12.9 million shares and 6.5 million warrants to purchase
common stock to institutional healthcare investors. In conjunction with the private placement we
received net proceeds of approximately $28 million.
26
We invest our capital resources with the primary objective of capital preservation. As a
result of trends in interest rates in 2005, we have invested in some securities with maturity dates
of more than 90 days to enhance our investment yields. As such, some of our invested balances are
classified as short-term investments rather than cash equivalents in our consolidated financial
statements at December 31, 2005.
To accomplish our business plans, we will be required to continue to conduct substantial
development activities for some or all of our proposed products. The acquisition of Miikana
Therapeutics in January 2006 provides an additional product pipeline, including MKC-1, a cell cycle
regulator, which entered Phase 2 oncology trials in January 2006. Under our current operating
plans, which include supporting two Phase 2 and one Phase 1 clinical trials for oncology compounds,
we expect our 2006 results of operations to reflect a net loss of approximately $30,000,000 before
non-cash charges associated with the Miikana acquisition and the adoption of SFAS 123R. We expect
that the majority of our 2006 revenues will continue to be from royalties on the sale of
Thalomid®. Based on historical trend and analyst consensus for Thalomid®
sales in 2006, we expect to record royalty sharing revenues in excess of $3.0 million in 2006;
however, there can be no assurance in this regard. In addition, under our licensing agreement
with Oxford Biomedica, PLC and Oxford Biomedica (UK) Limited Oxford, we are entitled to receive
payments upon the achievement of certain milestones. However, we do not control the drug
development efforts of Oxford and have no control over when or whether such milestones will be
reached. We do not believe that we will receive any developmental milestone payments under these
agreements in 2006.
Based on our assessment of our current capital resources coupled with anticipated inflows, in
the absence of additional financing, we believe that we will have adequate resources to fund
planned operations for more than twelve months. Our estimate may change, however, based on our
decisions with respect to future clinical trials related to Panzem®, the timing of
receipt of milestone payments, developments in our business including the acquisition of additional
intellectual property, other investments in new or complimentary technology, and our success in
executing our current business plan.
To address our long-term capital needs, we intend to continue to pursue strategic
relationships that would provide resources for the further development of our product candidates.
There can be no assurance, however, that these discussions will result in relationships or
additional funding. In addition, we may continue to seek capital through the public or private
sale of securities, if market conditions are favorable for doing so. If we are successful in
raising additional funds through the issuance of equity securities, stockholders will likely
experience substantial dilution, or the equity securities may have rights, preferences or
privileges senior to those of the holders of our common stock. If we raise funds through the
issuance of debt securities, those securities would have rights, preferences and privileges senior
to those of our common stock. There can be no assurance that we will be successful in seeking
additional capital.
INFLATION AND INTEREST RATE CHANGES
Management does not believe that our working capital needs are sensitive to inflation and
changes in interest rates.
CONTRACTUAL OBLIGATIONS
The table below sets forth our contractual obligations at December 31, 2005.
CONTRACTUAL OBLIGATIONS | PAYMENTS DUE BY PERIOD | |||||||||||||||||||
Total | Less than 1 | 1-3 years | 3 - 5 years | More than | ||||||||||||||||
year | 5 years | |||||||||||||||||||
Operating Leases Obligations |
$ | 3,159,000 | $ | 975,000 | $ | 2,013,000 | $ | 171,000 | $ | | ||||||||||
Purchase Obligations |
||||||||||||||||||||
Clinical Trial Contracts |
888,000 | 533,000 | 355,000 | | | |||||||||||||||
Collaborative Research
Contracts |
123,000 | 123,000 | | | | |||||||||||||||
Contract Manufacturing |
302,000 | 302,000 | | | | |||||||||||||||
Outside Service Contracts |
90,000 | 90,000 | | | | |||||||||||||||
Total Contractual Obligations |
$ | 4,562,000 | $ | 2,023,000 | $ | 2,368,000 | $ | 171,000 | $ | |
27
ITEM 7A QUANTITATIVE AND QUALITATIVE DISCLOSURES ABOUT MARKET RISK.
The primary objective of our investment activities is to preserve our capital until it is
required to fund operations while at the same time maximizing the income we receive from our
investments without incurring investment market volatility risk. Our investment income is sensitive
to the general level of U.S. interest rates. In this regard, changes in the U.S. interest rates
affect the interest earned on our cash and cash equivalents. Due to the short-term nature of our
cash and cash equivalent holdings, a 10% movement in market interest rates would not materially
impact on the total fair market value of our portfolio as of December 31, 2005.
ITEM 8. FINANCIAL STATEMENTS AND SUPPLEMENTARY DATA.
The response to this item is submitted in a separate section of this report. See Index to
Consolidated Financial Statements on Page F-1.
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
Under the supervision and with the participation of the Companys President and Chief
Executive Officer and its Chief Financial Officer (its principal executive officer and principal
financial officer), management has reviewed and evaluated the effectiveness of the design and
operation of the Companys disclosure controls and procedures. Based on that evaluation, the
President and Chief Executive Officer and the Chief Financial Officer have concluded that these
disclosure controls and procedures are effective as of December 31, 2005.
Managements Report on Internal Control Over Financial Reporting
The management of EntreMed, Inc. is responsible for establishing and maintaining adequate
internal control over financial reporting (as defined in Rules 13a-15(f) and 15d-15(f) under the
Securities Exchange Act of 1934, as amended). EntreMeds internal control over financial reporting
was designed to provide reasonable assurance to EntreMeds management and board of directors
regarding the reliability of financial reporting and the preparation of financial statements for
external purposes in accordance with generally accepted accounting principles. Any internal
control over financial reporting, no matter how well designed, has inherent limitations. As a
result of these inherent limitations, internal control over financial reporting may not prevent or
detect misstatements. Therefore, even those internal controls determined to be effective can
provide only reasonable assurance with respect to reliability of financial reporting and the
preparation of financial statements for external purposes in accordance with generally accepted
accounting principles.
EntreMeds management assessed the effectiveness of EntreMeds internal control over financial
reporting as of December 31, 2005 based on the criteria set forth by the Committee of Sponsoring
Organizations of the Treadway Commission (COSO) in Internal Control Integrated Framework.
Managements assessment included an evaluation of the design of EntreMeds internal control over
financial reporting and testing of the operational effectiveness of EntreMeds internal control
over financial reporting. Based on this assessment, EntreMeds management concluded that, as of
December 31, 2005, EntreMeds internal control over financial reporting was effective.
Ernst & Young LLP, an independent registered public accounting firm, has issued an attestation
report on managements assessment of EntreMeds internal control over financial reporting, as
stated in their report included herein.
28
Report of Ernst & Young LLP, Independent Registered Public Accounting Firm,
Regarding Internal Control over Financial Reporting
Regarding Internal Control over Financial Reporting
Board of Directors
EntreMed, Inc.
EntreMed, Inc.
We have audited managements assessment, included in the accompanying Managements Report on
Internal Control Over Financial Reporting, that EntreMed, Inc. (EntreMed) 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). EntreMeds 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 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 EntreMed 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, EntreMed 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 EntreMed as of December 31, 2005 and
2004, and the related consolidated statements of operations, stockholders equity, and cash flows
for each of the three years in the period ended December 31, 2005 and our report dated March 8,
2006 expressed an unqualified opinion thereon.
/s/ Ernst & Young LLP
McLean, Virginia
March 8, 2006
McLean, Virginia
March 8, 2006
29
Changes in Internal Control Over Financial Reporting
There were no changes in the Companys internal control over financial reporting during the
quarter ended December 31, 2005 that have materially affected, or are reasonably likely to
materially affect, the internal control over financial reporting.
PART III
ITEM 10. DIRECTORS AND EXECUTIVE OFFICERS OF THE REGISTRANT.
The following table sets forth the names, ages and positions of our executive officers and
directors as of March 2, 2006:
Name | Age | Position | ||||
Michael M. Tarnow
|
61 | Chairman of the Board of Directors | ||||
Ronald Cape (2) (3)
|
73 | Director | ||||
Donald S. Brooks
|
70 | Director | ||||
Dwight L. Bush (1) (6)
|
49 | Director | ||||
Jennie C. Hunter-Cevera
|
58 | Director | ||||
Peter S. Knight (2) (4)
|
55 | Director | ||||
Mark C. M. Randall (1) (5)
|
43 | Director | ||||
James S. Burns
|
59 | Director/President and CEO | ||||
Dane R. Saglio
|
48 | Chief Financial Officer | ||||
Carolyn F. Sidor, M.D.
|
58 | Corporate Vice President and Chief Medical Officer | ||||
Marc G. Corrado
|
41 | Vice President, Corporate Development | ||||
James D. Johnson, Ph.D., J.D.
|
58 | Senior Vice President and General Counsel |
(1) | Member of Audit Committee | |
(2) | Member of Compensation Committee | |
(3) | Member of the Nominating and Corporate Governance Committee | |
(4) | Chairman of Audit Committee | |
(5) | Chairman of the Compensation Committee | |
(6) | Chairman of the Nominating and Corporate Governance Committee |
Michael M. Tarnow was appointed chairman of our Board in February 2003. Since 1995, Mr.
Tarnow has been an advisor to and member of the boards of directors of several healthcare-related
organizations in the U.S., Canada and Europe, including publicly traded companies Axcan Pharma,
Inc. and MediGene AG. He also serves on the Boards of the University of Illinois College of Law,
Massachusetts College of Art Foundation and the Food Drug Law Institute. From 1995-2000, he was
President and CEO of Boston-based Creative BioMolecules, Inc. Prior to 1995, he spent 22 years at
Merck & Co., Inc., where he served in a wide variety of positions including heading corporate
development, President and CEO of Merck Frosst Canada and Executive Vice President of Merck-Medco.
Mr. Tarnow received his J.D. from the University of Illinois and his bachelors degree from Wayne
State University.
30
Ronald Cape, Ph.D. has been a director of the Company since 2003. He is the founder of Ronald
Cape Investment Management, LLC, a consulting firm and was the co-founder of Cetus Corporation, a
genetic engineering company, where he was Chairman of the Board of Directors for 20 years until the
company merged with Chiron Corporation in 1991. He was also a founding member of the Industrial
Biotechnology Association (now the Biotechnology Industry Organization BIO) and served as its
President for three years. Since 199l Dr. Cape has been an investor in the field of biotechnology
and a board member of many companies. He was the founding Chairman of Darwin Molecular
Corporation, which was later sold to Chiroscience plc., and is Chairman and a Director of Caprion,
Inc. and Neugenesis Corporation. He is also a Director of Neurobiological Technologies, Inc.,
Cellicon Biotechnologies, Inc., Ionian Technologies, Inc., Planet Biotechnology, Inc. and PureTech
Ventures, LLC.
Donald S. Brooks has been one of EntreMeds directors since April 1996 and was Vice President,
Legal Affairs from 1998 until August 2001. Between 1993 and 1998, Mr. Brooks was a practicing
attorney with the law firm of Carella Byrne Bain Gilfillan Cecchi Stewart and Olstein, Roseland,
New Jersey. Prior thereto, Mr. Brooks was employed by Merck and Co., Inc. for 27 years, most
recently, from 1986 to 1993, as Senior Counsel. From 1980 to 1985, Mr. Brooks served as a U.S.
employer delegate to the Chemical Industries Committee, International Labor Organization in Geneva,
Switzerland. He currently serves as a member of the Board of Directors of BioDiem, Ltd., an
Australian biotechnology company. In addition, he recently served as a Board member of Xenon
Pharmaceuticals, Inc., a Canadian biotechnology firm, and currently acts as a consultant to that
company.
Dwight L. Bush has been a director of the Company since June 2004. Mr. Bush has been a
principal of Stuart Mill Capital, LLC, an Arlington, Virginia-based investment firm, since 1997.
Since 2004, Mr. Bush has served as Vice Chairman of Enhanced Capital partners, LLC. From 1999
until 2002, Mr. Bush also served as Vice President and Chief Financial Officer of Sato Travel
Holdings, Inc. Prior to that, from 1994 through 1997, Mr. Bush was Vice President-Corporate
Development of Sallie Mae Corporation, a $60 billion financial service corporation and the nations
leading provider of education credit. Mr. Bush joined Sallie Mae after a successful 15-year career
at the Chase Manhattan Bank, where he began his professional career in 1979. His tenure at Chase
included international corporate banking assignments in Latin America, Asia and the Middle East,
and corporate finance and project finance in New York and Washington, D.C. Mr. Bush serves on the
governing boards of several organizations involved in industry, education and the arts, including
Cornell University, The Vaccine Fund, ICBC Broadcast Holdings, Inc, and The National Symphony
Orchestra. Mr. Bush earned his bachelors degree from Cornell University, College of Arts and
Sciences.
Jennie C. Hunter-Cevera has been a director of the Company since June 2001. Dr. Hunter-Cevera
is the President of the University of Maryland Biotechnology Institute. Prior to joining the
University of Maryland in October 1999, Dr. Hunter-Cevera had been the head of the Center for
Environmental Biotechnology at Lawrence Berkeley National Laboratory between November 1994 and
October 1999, Director of Fermentation, Research and Development at Cetus Corporation and a
scientist at E.R. Squibb and Company. She was co-founder of The Biotic Network and Blue Sky
Laboratories in Sonora, CA. Dr. Hunter-Cevera was elected to the American Academy of Microbiology
in 1995, the recipient of the 1996 SIM Charles Porter Award, elected as a SIM Fellow in 1997 and
the 1999 Nath Lecturer at West Virginia University. She is the 2004 recipient of the ASM Porter
Award for achievement in biodiversity research.
Peter S. Knight has been a Director of the Company since June 2000. Mr. Knight has been
President of Generation Investment Management US, a London-based asset management company, since
August 2004. From 2001 2003 he was a Managing Director of MetWest Financial. From 1977 to 1989,
Mr. Knight served as Chief of Staff to Al Gore when Mr. Gore was a member of the U.S. House of
Representatives and later the U.S. Senate. Mr. Knight also served as the General Counsel of
Medicis Pharmaceutical from 1989 to 1991. In 1991 Mr. Knight helped established the law firm of
Wunder, Knight, which represented numerous Fortune 500 companies. He practiced with this firm as a
partner until 1999. Mr. Knight has held senior positions on the last four presidential campaigns,
including serving as the campaign manager for the successful 1996 re-election of President Clinton.
Mr. Knight currently serves as a director of Medicis Pharmaceutical Corp. and Pharmaceutical
Resources, Inc. He is also a director of Schroders mutual fund and hedge fund family, a member of
the Cornell University Council and
the Cornell University College of Arts and Sciences Advisory Council. He holds a B.A. degree from
Cornell University and a J.D. degree from Georgetown University Law Center.
31
Mark C. M. Randall has been a director of the Company since April 1996. He has been CEO of
Commander Asset Management Ltd. since May 2002. Prior to this appointment he was associated with
Sarasin International Securities Limited, London, England, a wholly owned subsidiary of Bank
Sarasin and Cie, a private bank based in Switzerland, where he was a Director since 1994 and
Managing Director since 1999. Mr. Randall also serves as Chairman of Acorn Alternative Strategies
(Overseas) Ltd., an investment fund company.
James S. Burns has been President and Chief Executive Officer of EntreMed, Inc. since June
2004 and a director since September 2004. From 2001-2003, Mr. Burns was a co-founder and served as
President and as Executive Vice President of MedPointe, Inc., a specialty pharmaceutical company
that develops, markets and sells branded prescription pharmaceuticals. From 2000-2001, he served
as a founder and Managing Director of MedPointe Capital Partners, a private equity firm that led a
leveraged buyout to form MedPointe Pharmaceuticals. Previously, Mr. Burns was a founder, Chairman,
President and CEO of Osiris Therapeutics, Inc., a biotech company developing therapeutic stem cell
products for the regeneration of damaged or diseased tissue. He has also been Vice Chairman of
HealthCare Investment Corporation and a founding General Partner of Healthcare Ventures L.P., a
venture capital partnership specializing in forming companies built around new pharmaceutical and
biotechnology products; Group President at Becton Dickinson and Company, a multidivisional
biomedical products company; and was Vice President and Partner at Booz Allen & Hamilton, Inc., a
multinational consulting firm. Mr. Burns is Chairman of the Executive Committee the American Type
Culture Collection (ATCC), and a Director of Ciphergen Biosystems, Inc. He earned his BS and MS
degrees in biological sciences from the University of Illinois and an MBA degree from DePaul
University.
Dane R. Saglio joined EntreMed in April of 2000 as our Controller. He was appointed our Chief
Financial Officer in February 2003. Prior to joining EntreMed, Mr. Saglio had been Director of
Finance at Public Communications Associates (Cominex), a private telecommunications company, since
1996. Mr. Saglio has 20 years of experience, holding positions in accounting and general
management in the telecommunications, real estate, and government contracting industries prior to
joining EntreMed. Mr. Saglio received his bachelors degree in Business Administration from the
University of Maryland and is a certified public accountant licensed in Maryland.
Carolyn F. Sidor joined EntreMed in 2001 as Vice President, Clinical & Regulatory Affairs.
She was appointed Chief Medical Officer in September 2004. Dr. Sidor is a board-certified
hematologist/medical oncologist. From 1993 until 2001, Dr. Sidor held various leadership positions
at Cato Research Ltd., a prominent Clinical Research Organization (CRO), including Vice President,
Scientific and Medical Development; Medical Director; Senior Clinical Research Physician; and
Project Director. In those roles, Dr. Sidor served as medical monitor for clinical studies in
several therapeutic areas. Her experience includes participating in FDA meetings, leading
investigator meetings, recruiting and evaluating potential sites for clinical studies, and
evaluating new technologies. Since 1995, Dr. Sidor has held a staff appointment in the Department
of Medicine at the University of North Carolina at Chapel Hill and she retains that appointment.
From 1985 to 1993, Dr. Sidor was Associate Professor and Physician at Philadelphias Lankenau
Hospital, Department of Medicine. Prior to earning her MD degree from Thomas Jefferson University
School of Medicine in 1982 and completing her residency and medical training in 1988, Dr. Sidor
earned her MBA degree from the University of Delaware while working for E.I. DuPont in Central
Research and Development.
Marc Corrado joined
EntreMed in March 2005 as Vice President, Corporate Development.
From 2003 to 2004, Mr. Corrado served as Senior Director, Mergers and
Acquisitions for Aventis in Bridgewater, New Jersey. Prior to 2003,
Mr. Corrado held various positions at Aventis including Director,
Oncology New Product Commercialization from 2002 to 2003 and
Director, Global Business Development from 2000 to 2002. From 1995 to
2000, Mr. Corrado held positions at Rhône-Poulenc Rorer in Paris,
France, including Senior Manager, Worldwide Business Development from
1998 to 2000 and Manager, Worldwide Business Development &
Licensing from 1995 to 1997. Mr. Corrado holds a J.D. from the
American University in Washington, D.C., and a M.B.A. from INSEAD
in Fontainebleau, France.
32
James D. Johnson, Ph.D., J.D., has been our Senior Vice President and General Counsel since
May 2001. Dr. Johnson has been a partner with the law firm of Kilpatrick, Stockton since 2000.
Before that he had been a partner with the law firm of Jones and Askew, which merged with
Kilpatrick, Stockton in 2000, since 1993. He graduated from the University of the South with a
B.A. in Chemistry and received his Ph.D. in Biochemistry from the Emory University School of
Medicine. He graduated from the Emory University School of Law.
Section 16(a) Beneficial Ownership Reporting Compliance
Section 16(a) of the 1934 Securities and Exchange Act requires the Companys executive
officers, directors and persons who beneficially own more than 10% of a registered class of the
Companys equity securities to file with the SEC initial reports of ownership and reports of
changes in ownership of Common Stock and other equity securities of the Company. Such executive
officers, directors, and greater than 10% beneficial owners are required by SEC regulation to
furnish the Company with copies of all Section 16(a) reports filed by such reporting persons.
Based solely on our review of such forms furnished to the Company and written representations
from certain reporting persons, we believe that all filing requirements applicable to our executive
officers, directors and greater than 10% beneficial owners were complied with during fiscal 2005.
Audit Committee
As indicated above, the Companys Board of Directors has a designated audit committee. The
current members of the audit committee are Mr. Knight, Mr. Bush and Mr. Randall. The Board of
Directors has determined that Mr. Bush is an audit committee financial expert as defined in rules
and regulations of the SEC. Mr. Bush is an independent director as that term is defined in
Nasdaq listing standards.
Code of Ethics
The Company has adopted a Code of Ethics, as defined in applicable SEC and NASD rules, that
applies to the Companys directors, officers and employees, including the Companys principal
executive officer and principal financial and accounting officer. The Code of Ethics is available
on the Companys website at www.entremed.com.
33
ITEM 11. EXECUTIVE COMPENSATION.
Executive Compensation and Other Matters
The following summary compensation table sets forth the aggregate compensation paid or accrued
by us to the Chief Executive Officer and to executive officers whose annual compensation exceeded
$100,000 for fiscal 2005 (collectively, the named executive officers) for services during the
fiscal years ended December 31, 2005, 2004 and 2003.
SUMMARY COMPENSATION TABLE
LONG TERM | ||||||||||||||||||||
COMPENSATION | ||||||||||||||||||||
AWARDS | ||||||||||||||||||||
SECURITIES | ||||||||||||||||||||
UNDERLYING | ||||||||||||||||||||
ANNUAL | BONUS | OPTIONS/SARS | ALL OTHER | |||||||||||||||||
NAME AND PRINCIPAL POSITION | YEAR | SALARY ($) | ($) | (#) | COMPENSATION | |||||||||||||||
James S. Burns |
2005 | 372,000 | 160,000 | 0 | 10,579 | (1) | ||||||||||||||
President and Chief Executive Officer |
2004 | 195,000 | (4) | 80,000 | 600,000 | 4,125 | ||||||||||||||
Dane R. Saglio |
2005 | 220,000 | 55,000 | 40,000 | 13,525 | (1) | ||||||||||||||
Chief Financial Officer |
2004 | 200,000 | 50,000 | 75,000 | 12,748 | |||||||||||||||
2003 | 190,212 | 50,000 | 75,000 | 9,681 | ||||||||||||||||
Carolyn F. Sidor, M.D. |
2005 | 240,000 | 65,000 | 40,000 | 10,279 | (1) | ||||||||||||||
Corporate Vice President and |
2004 | 234,500 | 50,000 | 100,000 | 1,365 | |||||||||||||||
Chief Medical Officer |
2003 | 226,600 | 22,600 | 50,000 | 288 | |||||||||||||||
James D. Johnson, Ph.D., J.D. |
2005 | 10,800 | (5) | 0 | 0 | 40 | (2) | |||||||||||||
Senior Vice President and General Counsel |
2004 | 10,800 | 0 | 0 | 40 | |||||||||||||||
2003 | 10,800 | 0 | 40,000 | 0 | ||||||||||||||||
Marc Corrado |
2005 | 153,000 | (6) | 40,375 | 25,000 | 47,513 | (3) | |||||||||||||
Vice President, Corporate Development |
(1) | Consists of group health and life insurance premiums paid on behalf of such officer. | |
(2) | Consists of life insurance premiums paid on behalf of such officer. | |
(3) | Consists of group health and life insurance premiums in the amount of $7,889 and relocation costs of $39,624 paid on behalf of such officer. | |
(4) | Represents salary from June 15, 2004 date of hire. | |
(5) | In addition, the Company pays legal fees to a law firm of which Dr. Johnson is a partner for his services as Senior Vice President and General Counsel. Such firm also provides additional legal services to the Company. | |
(6) | Represents salary from March 1, 2005, date of hire. |
34
The following table sets forth certain information with respect to individual grants of
stock options made during the fiscal year ended December 31, 2005 to each of the named executive
officers.
Option/SAR Grants in Last Fiscal Year
Individual Grants | ||||||||||||||||||||||||
Number of | % of Total | Potential Realizable | ||||||||||||||||||||||
Securities | Options/SARS | Value at Assumed | ||||||||||||||||||||||
Underlying | Granted | Exercise | Annual Rates of Stock | |||||||||||||||||||||
Options/SAR | to Employees | or Base | Price Appreciation for | |||||||||||||||||||||
Granted | in Fiscal | Price | Expiration | Option Term (1) | ||||||||||||||||||||
Name | (#) (2) | Year | ($/sh) | Date | 5% ($) | 10% ($) | ||||||||||||||||||
Dane R. Saglio |
40,000 | 10.53 | % | 1.94 | 12/23/2015 | 48,802 | 123,674 | |||||||||||||||||
Carolyn F. Sidor,
M.D. |
40,000 | 10.53 | % | 1.94 | 12/23/2015 | 48,802 | 123,674 | |||||||||||||||||
Marc Corrado |
100,000 | 26.34 | % | 3.70 | 2/16/2015 | 232,691 | 589,685 | |||||||||||||||||
30,000 | 7.90 | % | 1.94 | 12/23/2015 | 36,602 | 92,756 |
(1) | Calculated by multiplying the exercise price by the annual appreciation rate shown (as prescribed by SEC rules and compounded for the term of the options), subtracting the exercise price per share and multiplying the gain per share by the number of shares covered by the options. These amounts are not intended to forecast possible future appreciation, if any, of the price of our shares. The actual value realized upon exercise of the options to purchase our shares will depend on the fair market value of our shares on the date of exercise. | |
(2) | The options listed were granted under our 2001 Plan. 25% of such options were exercisable on the date of grant. 25% of the balance becomes exercisable on each anniversary date of the grant over the next three years. In the event of certain transactions, including those that may result in a change in control, as defined under the Companys stock plans, unvested installments of options to purchase our shares may become immediately exercisable. |
The following table sets forth information concerning all option holdings for the fiscal year
ended December 31, 2005 for each of the named executive officers.
Aggregated Option/SAR Exercises in Last Fiscal Year and Fiscal Year End Option/SAR Values
Number of Securities | Value of Unexercised | |||||||||||||||
Underlying Options/SARs at | In-the-Money Options/SARs | |||||||||||||||
Fiscal Year-End (#) | at Fiscal Year-End ($) (1) | |||||||||||||||
Name | Exercisable | Unexercisable | Exercisable | Unexercisable | ||||||||||||
James S. Burns |
300,000 | 300,000 | 0 | 0 | ||||||||||||
Dane R. Saglio |
164,450 | 86,250 | 21,250 | 0 | ||||||||||||
Carolyn F. Sidor,
M.D. |
184,733 | 86,250 | 43,949 | 6,250 | ||||||||||||
James D. Johnson,
Ph.D., J.D. |
385,250 | 10,000 | 85,000 | 0 | ||||||||||||
Marc Corrado |
57,500 | 72,500 | 0 | 0 |
(1) | Calculated by multiplying the number of unexercised options outstanding at December 31, 2005 by the difference between the fair market value of our shares at December 31, 2005 ($1.94) and the option exercise price. |
35
Compensation of Directors
Upon joining the Board of Directors, each new non-employee director is granted an option,
vesting over three years, to purchase 50,000 shares of Common Stock. Continuing directors receive
an option to purchase 30,000 shares of Common Stock as of the date of the annual meeting of
stockholders, 25% of which is exercisable immediately and 25% of which become exercisable each year
over the next three years. Chairpersons of Board committees receive an option to purchase an
additional 5,000 shares of Common Stock as of the date of the annual meeting, 25% of which is
exercisable immediately and 25% of which become exercisable each year over the next three years.
Non-employee directors also receive an annual retainer fee of $25,000 that is payable solely in
restricted stock.
Pursuant to a Board Service Agreement between the Companys Chairman, Michael M. Tarnow, and
the Company dated February 5, 2003, Mr. Tarnow is paid $5,000 per month for his services as
Chairman. He also received an option to purchase 250,000 shares of Common Stock, 25% of which were
exercisable immediately and 25% of which become exercisable each year over the next three years.
Mr. Tarnow is also reimbursed for expenses in connection with his service as Chairman, including
travel to and from Board meetings, and participates in all other Board compensation programs,
including the receipt of the annual option grants granted to all directors of the Company.
Employment Contracts and Termination of Employment and Change-In-Control Arrangements
On June 15, 2004, the Company entered into an employment agreement with James S. Burns, its
President and Chief Executive Officer. The term of the employment agreement is for three years,
subject to automatic one-year extensions unless either party gives at least sixty days prior
written notice not to extend.
The Agreement provides for an annualized minimum base salary of $360,000, with incentive
compensation at a minimum of 40% of base salary. The base salary will be reviewed at least annually
in accordance with the Companys customary practices for executives. In addition, the Company
granted Mr. Burns stock options covering 500,000 shares, vested as to 25% on the date of grant and
vesting in 25% annual cumulative installments thereafter.
During the first eighteen months of the term of the agreement, if the Company terminates Mr.
Burns without cause, Mr. Burns will receive a severance benefit equal to 18 months of salary,
payable in accordance with the Companys customary pay practices, a pro-rata portion of any
incentive compensation he would have been entitled to for that year, and continued insurance
coverage for up to 12 months. After the first 18 months of the Agreement, if the Company
terminates Mr. Burns without cause or fails to extend the employment agreement, Mr. Burns will be
entitled to the same severance benefits except that he will be entitled to a base salary equal to
12 months instead of 18 months. In addition, if Mr. Burns is terminated without cause anytime
after the first year of the term of the agreement, all of his unexpired and unvested stock options
will become vested on the effective date of such termination. Mr. Burns also may resign at any
time for good reason, as defined in the employment agreement, by providing at least sixty days
prior written notice. Resignation for good reason will be deemed a termination without cause. In
addition, if Mr. Burns employment is terminated upon disability or death, Mr. Burns or his estate
will be entitled to receive a payment equal to 12 months salary plus a pro-rated amount of any
incentive compensation he would have been entitled to for that year.
The employment agreement imposes non-compete and confidentiality obligations on Mr. Burns
following termination of employment.
On July 1, 2004, the Company entered into an employment agreement with Dane Saglio, its Chief
Financial Officer. The term of the employment agreement is for one year, and the agreement may be
renewed for successive one-year periods upon mutual agreement by both parties.
On May 20, 2005, the Company entered into a one-year extension of its employment agreement
with Dane Saglio, our Chief Financial Officer. The one-year extension is for the period beginning
July 1, 2005 through June 30, 2006. There were no other changes to the terms of Mr. Saglios employment agreement
pursuant to the extension.
36
The Agreement provides for an annualized minimum base salary of $200,000. The base salary will
be reviewed at least annually in accordance with the Companys customary practices for executives.
Mr. Saglio is also entitled to incentive compensation as determined by the Companys Board of
Directors or a committee thereof.
If the Company terminates Mr. Saglio without cause, Mr. Saglio will receive a severance
benefit equal to 12 months of salary, payable in accordance with the Companys customary pay
practices, a pro-rata portion of any incentive compensation he would have been entitled to for that
year, and continued insurance coverage for up to 12 months. In addition, if Mr. Saglio is
terminated without cause, all of his unexpired and unvested stock options will become vested on the
effective date of such termination. Mr. Saglio also may resign at any time for good reason, as
defined in the employment agreement, by providing at least sixty days prior written notice.
Resignation for good reason will be deemed a termination without cause. In addition, if Mr.
Saglios employment is terminated upon disability or death, Mr. Saglio or his estate will be
entitled to receive a payment equal to 12 months salary plus a pro-rated amount of any incentive
compensation he would have been entitled to for that year.
The employment agreement imposes non-compete and confidentiality obligations on Mr. Saglio
following termination of employment.
On December 1, 2004, the Company entered into an employment agreement with Dr. Carolyn F.
Sidor, its Vice President and Chief Medical Officer. The term of the employment agreement is for
one year, subject to automatic one-year extensions unless either party gives at least sixty days
prior written notice not to extend.
The Agreement provides for an annualized minimum base salary of $240,000, with incentive
compensation targeted at 25% of base salary. The base salary will be reviewed at least annually in
accordance with the Companys customary practices for executives. In addition, the Company granted
Dr. Sidor stock options covering 50,000 shares, vested as to 25% on the date of grant and vesting
in 25% annual cumulative installments thereafter.
If the Company terminates Dr. Sidor without cause or fails to extend the employment
agreement, Dr. Sidor will be entitled to the same severance benefits except that she will be
entitled to a base salary equal to six months instead of 12 months. Dr. Sidor also may resign at
any time for good reason, as defined in the employment agreement, by providing at least sixty
days prior written notice. Resignation for good reason will be deemed a termination without
cause. In addition, if Dr. Sidors employment is terminated upon disability or death, Dr. Sidor or
her estate will be entitled to receive a payment equal to six months salary plus a pro-rated amount
of any incentive compensation she would have been entitled to for that year.
The employment agreement imposes non-compete and confidentiality obligations on Dr. Sidor
following termination of employment.
In the event of certain transactions, including those that may result in a change in control,
as defined under our Incentive Stock Option Plans, unvested installments of options to purchase our
shares may become immediately exercisable.
On May 20, 2005, the Company entered into an employment agreement with Marc G. Corrado, its
Vice President, Corporate Development. The term of the employment agreement commenced on March 1,
2005 and continues for one year, subject to automatic one-year extensions unless either party gives
at least thirty days prior written notice not to extend.
The Agreement provides for an annualized minimum base salary of $204,000, with incentive
compensation targeted at 25% of base salary. The base salary will be reviewed at least annually in
accordance with the Companys customary practices for executives. In addition, the Company granted
Mr. Corrado stock options covering 100,000 shares, vested as to 25% on the date of grant and
vesting in 25% annual cumulative installments thereafter.
The Agreement provides for reimbursement of certain relocation costs, including two
house-hunting trips, lease termination penalties (estimated at $5,000), short-term living expenses
for up to three months in the Washington, D.C. area, and up to $25,000 for other relocation
expenses such as moving and storage of household goods and belongings.
37
If the Company terminates Mr. Corrado without cause, Mr. Corrado will receive a severance
benefit equal to six months of salary, payable in accordance with the Companys customary pay
practices, a pro-rata portion of any incentive compensation he would have been entitled to for that
year, and continued insurance coverage for up to six months. Mr. Corrado also may resign at any
time for good reason, as defined in the employment agreement, by providing at least thirty days
prior written notice. Resignation for good reason will be deemed a termination without cause.
In addition, if Mr. Corrados employment is terminated upon disability or death, Mr. Corrado or his
estate will be entitled to receive a payment equal to six months salary plus a pro-rated amount of
any incentive compensation he would have been entitled to for that year.
The employment agreement imposes non-compete and confidentiality obligations on Mr. Corrado
following termination of employment.
In the event of certain transactions, including those that may result in a change in control,
as defined under our Incentive Stock Option Plans, unvested installments of options to purchase our
shares may become immediately exercisable.
ITEM 12. SECURITY OWNERSHIP OF CERTAIN BENEFICIAL OWNERS AND MANAGEMENT AND RELATED
STOCKHOLDER MATTERS.
The following table sets forth, as of March 2, 2006, certain information concerning stock
ownership of all persons known by us to own beneficially more that 5% of our Common Stock, $.01 par
value per share, as well as each director or director nominee, each executive officer named under
Executive Compensation and Other Matters and all of our directors and executive officers as a
group.
Amount and Nature of | ||||||||
Name of Beneficial Owner (1) | Beneficial Ownership(1) | Percentage of Class | ||||||
Michael M. Tarnow |
349,191 | (2) | * | |||||
Donald S. Brooks |
204,334 | (2) | * | |||||
James S. Burns |
365,000 | (2) | * | |||||
Dwight L. Bush |
83,388 | (2) | * | |||||
Ronald Cape |
84,191 | (2) | * | |||||
Marc Corrado |
57,500 | (2) | * | |||||
Jennie C. Hunter-Cevera, Ph.D. |
144,191 | (2) | * | |||||
James D.
Johnson Ph.D., J.D. |
385,250 | (2) | * | |||||
Peter S. Knight |
186,691 | (2) | * | |||||
Mark C. M. Randall |
253,025 | (2) | * | |||||
Dane R. Saglio |
166,750 | (2) | * | |||||
Carolyn F. Sidor |
184,733 | (2) | * | |||||
All executive officers and |
2,464,244 | (2) | 3.23 | % | ||||
directors as a group (13 persons) |
||||||||
More Than 5% Beneficial Owners |
||||||||
Celgene Corporation |
24,614,864 | (3) | 24.95 | % | ||||
7 Powder Horn Drive |
||||||||
Warren, N.J. 07059 |
* | Less than 1% | |
(1) | Beneficial ownership is defined in accordance with the rules of the Securities and Exchange Commission (SEC) and generally means the power to vote and/or to dispose of the securities regardless of any economic interest therein. | |
(2) | Includes shares issuable upon exercise of options and warrants which are exercisable within 60 days in the following amounts: Mr. Tarnow, 325,000; Mr. Brooks, 204,334; Mr. Burns, 300,000; Mr. Bush, 63,750; Dr. Cape, 60,000; Mr. Corrado, 57,500; Dr. Hunter-Cevera, 120,000; Dr. Johnson, 385,250; Mr. Knight, 162,500; Mr. Randall, 228,834; Mr. Saglio, 164,450; Dr. Sidor, 184,733; and all officers and directors as a group, 2,256,351. |
38
(3) | Includes 3,350,000 shares of the Companys Series A convertible preferred stock convertible into 16,750,000 shares of common stock. |
Options under Employee Benefit Plans
The following table discloses certain information about the options issued and available for
issuance under all outstanding Company option plans, as of December 31, 2005.
(a) | (b) | (c) | ||||||||||
Number of securities | ||||||||||||
remaining available for | ||||||||||||
future issuance under | ||||||||||||
Number of securities to | Weighted-average | equity compensation | ||||||||||
be issued upon exercise | exercise price of | plans (excluding | ||||||||||
of outstanding options, | outstanding options, | securities reflecting in | ||||||||||
Plan category | warrants and rights | warrants and rights | column (a)) | |||||||||
Equity compensation
plans approved by
security holders |
7,962,017 | $ | 9.04 | 631,176 | ||||||||
Equity compensation
plans not approved
by security holders |
147,295 | $ | 4.20 | 0 | ||||||||
Total |
8,109,312 | $ | 8.95 | 631,176 | ||||||||
Warrants issued under the unauthorized plans represent compensation for consulting services
rendered by the holders.
ITEM 13. CERTAIN RELATIONSHIPS AND RELATED TRANSACTIONS.
James D. Johnson, our Senior Vice President, is a partner at Kilpatrick, Stockton, which
provides certain patent prosecution and certain other legal services to us. We paid approximately
$1,180,000 to Kilpatrick, Stockton for these services in 2005. This amount represents less than 5%
of Kilpatrick, Stocktons total revenues for 2005.
In addition, during 2005 the Company received financial advisory services from Ferghana
Partners, Inc., a provider of corporate financial advice to firms in the Life Sciences field. The
Companys chairman also serves as non-executive chairman of Ferghana Partners, Inc. The Companys
chairman and CEO both hold a de minimis ownership interest in Ferghana Partners, Inc. The Company
paid approximately $785,000 in fees to Ferghana Partners, Inc. in 2005. This amount represents less
than 5% of Ferghana Partners total revenues for 2005.
ITEM 14. PRINCIPAL ACCOUNTING FEES AND SERVICES.
The following information discloses the fees we paid to our auditors during fiscal years 2005
and 2004.
Audit Fees
The Company incurred from Ernst & Young audit fees of $175,000 in fiscal 2005 and
$172,000 in fiscal 2004 for audit fees, covering professional services rendered for (1) the audit
of the Companys annual financial statements included in the Companys Annual Report on Form 10-K
for the fiscal year ended December 31, 2004 and 2003 and (2) the reviews of the financial
statements included in the Companys quarterly reports on Form 10-Q for the first three quarters of
2005 and 2004.
39
The Company incurred from Ernst & Young audit fees of $130,000 in fiscal 2005 and
$128,500 in fiscal 2004 for fees related to the auditors review of the internal control over
financial reporting, including their attestation report.
The Company incurred from Ernst & Young audit fees of $25,000 in fiscal 2005 and 2004 for fees
related to SEC filings and issuances of consents.
Audit-Related Fees
The Company did not incur any audit-related fees in fiscal 2005 or in fiscal 2004.
Tax Fees
The Company incurred from Ernst & Young tax fees of $17,500 in fiscal 2005 and $14,500 in
fiscal 2004, for tax compliance services, including preparation of tax returns.
All Other Fees
The Company did not incur any other fees from Ernst & Young in fiscal 2005 or in fiscal 2004.
PART IV
ITEM 15. EXHIBITS, FINANCIAL STATEMENT SCHEDULES.
(a) 1. FINANCIAL STATEMENTS See index to Consolidated Financial Statements.
2. Schedules
All financial statement schedules are omitted because they are not applicable, not required
under the instructions or all the information required is set forth in the financial statements or
notes thereto.
3. Exhibits
2.1(20) | Agreement and Plan of Merger, dated as of December 22, 2005 among EntreMed, Inc., E.M.K. Sub, Inc., Miikana Therapeutics, Inc., and Andrew Schwab |
3.1 | Amended and Restated Certificate of Incorporation of EntreMed, Inc. (incorporated by reference from our Form 10-K for the year ended December 31, 2002 previously filed with the Securities and Exchange Commission) | |
3.2(1) | By-laws of EntreMed, Inc. | |
4.1 | Certificate of Designations of the Series A Convertible Preferred Stock (incorporated by reference to Exhibit 99.4 of our Form 8-K dated December 31, 2002, and filed with the Commission on January 15, 2003) | |
4.2 | Warrant to Purchase Common Stock, dated January 13, 2003, issued by EntreMed, Inc. in favor of Celgene Corporation (incorporated by reference to Exhibit 99.5 of our Form 8-K dated December 31, 2002, and filed with the Commission on January 15, 2003) | |
10.1(1) | 1992 Stock Incentive Plan* |
40
10.2(1) | Amended and Restated 1996 Stock Option Plan* | |
10.3(1) | Form of Stock Option Agreement* | |
10.4(2) | License Agreement between Childrens Hospital Medical Center Corporation and EntreMed, Inc. signed December 20, 1996 regarding Estrogenic Compounds as Anti-Mitotic Agents | |
10.5(3) | Amendment to the 1996 Stock Option Plan* | |
10.6(4) | License Agreement between Celgene Corporation and EntreMed, Inc. signed December 9, 1998 regarding thalidomide intellectual property | |
10.7(4) | Lease Agreement between EntreMed, Inc. and Red Gate III Limited Partnership, dated June 10, 1998 | |
10.8(5) | 1999 Long-Term Incentive Plan* | |
10.9(6) | EntreMed, Inc. 2001 Long-Term Incentive Plan* | |
10.10.1(7) | Purchase Agreement between Bioventure Investments kft and EntreMed, Inc., dated June 15, 2001+ | |
10.10.2(7) | Amendment 1 to Purchase Agreement between Bioventure Investments kft and EntreMed, Inc., dated July 13, 2001 | |
10.10.3(7) | Amendment 2 to Purchase Agreement between Bioventure Investments kft and EntreMed, Inc., dated July 30, 2001 | |
10.10.4(7) | Amendment 3 to Purchase Agreement between Bioventure Investments kft and EntreMed, Inc., dated August 3, 2001 | |
10.11(8) | Board Service Agreement, dated February 5, 2003, between Michael M. Tarnow and EntreMed, Inc. * | |
10.12(9) | Asset Purchase Agreement by and Between Celgene Corporation and EntreMed, Inc., dated as of December 31, 2002 | |
10.13(9) | Securities Purchase Agreement by and among EntreMed, Inc., and Celgene Corporation, dated as of December 31, 2002 | |
10.14(9) | Investor and Registration Rights Agreement by and between EntreMed, Inc. and Celgene Corporation, dated as of December 31, 2002 | |
10.15(10) | Private Placement of Common Stock and Warrants to Certain Institutional Investors, dated as of November 3, 2003 | |
10.16(11) | Employment Agreement between EntreMed and James S. Burns effective June 15, 2004* | |
10.17(12) | Employment Agreement between EntreMed and Dane Saglio effective July 1, 2004* | |
10.18(17) | Letter Agreement between EntreMed and Dane Saglio dated May 20, 2005* | |
10.19(13) | Employment Agreement between EntreMed and Carolyn F. Sidor, M.D. effective December 1, 2004* | |
10.20(14) | Securities Purchase Agreement by and among EntreMed and Certain Institutional Investors, dated as of December 23, 2004 |
41
10.21(15) | EntreMed, Inc. 2001 Long Term Incentive Plan Non-Qualified Stock Option Grant Agreement (Director)* | |
10.22(15) | EntreMed, Inc. 2001 Long Term Incentive Plan Non-Qualified Stock Option Grant Agreement (Non-Director Employee)* | |
10.23(16) | Form of Letter Agreement between EntreMed and James S. Burns* | |
10.24(16) | Form of Restricted Stock Award under EntreMed, Inc. 2001 Long Term Incentive Plan* | |
10.25(16) | Employment Agreement by and between EntreMed and Marc Corrado, dated as of May 20, 2005* | |
10.26(18) | License Agreement between EntreMed and Celgene Corporation signed March 24, 2005 regarding the development and commercialization of Celgenes small molecule tubulin inhibitor compounds for the treatment of cancer+ | |
10.27(19) | Description of Compensation of Directors* | |
23.1 | Consent of Independent Registered Public Accounting Firm | |
31.1 | Rule 13a-14(a) Certification of President and CEO | |
31.2 | Rule 13a-14(a) Certification of Chief Financial Officer | |
32.1 | Rule 13a-14(b) Certification by President and CEO | |
32.2 | Rule 13a-14(b) Certification by Chief Financial Officer | |
* | Compensatory Plan, Contract or Arrangement. | |
+ | Certain portions of this exhibit have been omitted based upon a request for confidential treatment. The omitted portions have been filed with the Commission pursuant to our application for confidential treatment. | |
(1) | Incorporated by reference from our Registration Statement on Form S-1 (File No. 333-3536) declared effective by the Securities and Exchange Commission on June 11, 1996. | |
(2) | Incorporated by reference from our Form 10-K for the year ended December 31, 1996 previously filed with the Securities and Exchange Commission. | |
(3) | Incorporated by reference from our Form 10-K for the year ended December 31, 1997 previously filed with the Securities and Exchange Commission. | |
(4) | Incorporated by reference from our Form 10-K for the year ended December 31, 1998 previously filed with the Securities and Exchange Commission. | |
(5) | Incorporated by reference from our Form 10-Q for the quarter ended June 30, 1999 previously filed with the Securities and Exchange Commission. | |
(6) | Incorporated by reference from Exhibit A to our definitive proxy statement filed with the Securities and Exchange Commission on June 27, 2005. | |
(7) | Incorporated by reference from our Form 10-Q for the quarter ended June 30, 2001 previously filed with the Securities and Exchange Commission. |
42
(8) | Incorporated by reference from our Form 10-K/A for the year ended December 31, 2002 previously filed with the Securities and Exchange Commission. | |
(9) | Incorporated by reference from our Form 8-K dated December 31, 2002 filed with the Securities and Exchange Commission on January 15, 2003. | |
(10) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on November 4, 2003. | |
(11) | Incorporated by reference from our Form 10-Q for the quarter ended June 30, 2004 previously filed with the Securities and Exchange Commission. | |
(12) | Incorporated by reference from our Form 10-Q for the quarter ended September 30, 2004 previously filed with the Securities and Exchange Commission. | |
(13) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on December 6, 2004. | |
(14) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on December 29, 2004. | |
(15) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on February 23, 2005. | |
(16) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on March 11, 2005. | |
(17) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on May 24, 2005. | |
(18) | Incorporated by reference from our Form 10-Q for the quarter ended March 31, 2005 previously filed with the Securities and Exchange Commission. | |
(19) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on August 2, 2005. | |
(20) | Incorporated by reference from our Form 8-K filed with the Securities and Exchange Commission on December 29, 2005. |
43
SIGNATURES
Pursuant to the requirements of Section 13 or 15(d) of the Securities Exchange Act of 1934,
the registrant has duly caused this report to be signed on its behalf by the undersigned, thereunto
duly authorized.
ENTREMED, INC. | ||||||
By: | /s/ | James S. Burns. | ||||
James S. Burns | ||||||
President and Chief Executive Officer | ||||||
March 13, 2006 |
Pursuant to the requirements of the Securities Act of 1934, this report has been signed below
by the following persons in the capacities and on the dates indicated.
SIGNATURE | TITLE | DATE | ||
/s/ Michael M. Tarnow
|
Chairman of the Board | March 9, 2006 | ||
Michael M. Tarnow |
||||
/s/ James S. Burns
|
President and Chief | |||
James S. Burns
|
Executive Officer | March 13, 2006 | ||
/s/ Dane R. Saglio
|
Chief Financial Officer | |||
Dane R. Saglio
|
(Principal Financial and | |||
Accounting Officer) | March 9, 2006 | |||
/s/ Donald S. Brooks
|
Director | March 9, 2006 | ||
Donald S. Brooks |
||||
/s/ Dwight L. Bush
|
Director | March 10, 2006 | ||
Dwight L. Bush |
||||
/s/ Jennie C. Hunter-Cevera
|
Director | March 11, 2006 | ||
Jennie C. Hunter-Cevera |
||||
/s/ Mark C. M. Randall
|
Director | March 9, 2006 | ||
Mark C. M. Randall |
||||
/s/ Ronald Cape
|
Director | March 13, 2006 | ||
Ronald Cape |
||||
/s/ Peter S. Knight
|
Director | March 9, 2006 | ||
Peter S. Knight |
44
The following consolidated financial statements of EntreMed, Inc. are included in Item 8:
Report of Independent Registered Public Accounting Firm.
|
F-2 | |
Consolidated Balance Sheets as of December 31, 2005 and 2004.
|
F-3 | |
Consolidated Statements of Operations for the years ended December 31, 2005, 2004 and 2003.
|
F-4 | |
Consolidated Statements of Stockholders Equity for the years ended December 31, 2005, 2004 and 2003
|
F-5 | |
Consolidated Statements of Cash Flows for the years ended December 31, 2005, 2004 and 2003
|
F-6 | |
Notes to Consolidated Financial Statements.
|
F-7 |
F-1
Report of Ernst & Young LLP, Independent Registered Public Accounting Firm,
on the Audited Consolidated Financial Statements
on the Audited Consolidated Financial Statements
Board of Directors
EntreMed, Inc.
EntreMed, Inc.
We have audited the accompanying consolidated balance sheets of EntreMed, Inc. as of December 31,
2005 and 2004, and the related consolidated statements of operations, stockholders 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 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 EntreMed, Inc. 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 EntreMed, Inc.s 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 8, 2006 expressed an unqualified opinion thereon.
/s/ Ernst & Young LLP
McLean, Virginia
March 8, 2006
McLean, Virginia
March 8, 2006
F-2
EntreMed, Inc.
Consolidated Balance Sheets
Consolidated Balance Sheets
DECEMBER 31, | ||||||||
2005 | 2004 | |||||||
ASSETS |
||||||||
Current assets: |
||||||||
Cash and cash equivalents |
$ | 11,407,652 | $ | 20,425,495 | ||||
Short-term investments |
18,674,736 | 14,114,021 | ||||||
Accounts receivable |
3,723,433 | 3,250,783 | ||||||
Note receivable |
1,000,000 | | ||||||
Interest receivable |
181,231 | 85,089 | ||||||
Prepaid expenses and other |
338,462 | 367,222 | ||||||
Total current assets |
35,325,514 | 38,242,610 | ||||||
Property and equipment, net |
915,337 | 1,150,087 | ||||||
Other assets |
191,034 | 11,305 | ||||||
Total assets |
$ | 36,431,885 | $ | 39,404,002 | ||||
LIABILITIES AND STOCKHOLDERS EQUITY |
||||||||
Current liabilities: |
||||||||
Accounts payable |
$ | 5,487,014 | $ | 1,550,413 | ||||
Payable to related parties |
228,380 | 200,321 | ||||||
Accrued liabilities |
1,038,975 | 1,416,444 | ||||||
Current portion of deferred rent |
60,969 | 32,931 | ||||||
Current portion of deferred revenue |
| 95,496 | ||||||
Total current liabilities |
6,815,338 | 3,295,605 | ||||||
Deferred rent, less current portion |
230,206 | 291,175 | ||||||
Deferred revenue, less current portion |
| 95,496 | ||||||
Minority interest |
17,151 | 16,972 | ||||||
Stockholders equity: |
||||||||
Convertible preferred stock, $1.00 par value;
5,000,000 shares authorized and 3,350,000 shares issued and
outstanding at December 31, 2005 and 2004 (liquidation
value $33,500,000 and $54,270,000 , respectively) |
3,350,000 | 3,350,000 | ||||||
Common stock, $.01 par value: 120,000,000
and 90,000,000 shares authorized;
51,106,857 and 43,628,173 shares issued and outstanding
at December 31, 2005 and 2004, respectively |
511,069 | 436,282 | ||||||
Additional paid-in capital |
295,392,194 | 285,387,288 | ||||||
Deferred stock-based compensation |
(102,000 | ) | | |||||
Treasury stock, at cost: 874,999 shares held at
December 31, 2005 and 2004 |
(8,034,244 | ) | (8,034,244 | ) | ||||
Accumulated deficit |
(261,747,829 | ) | (245,434,572 | ) | ||||
Total stockholders equity |
29,369,190 | 35,704,754 | ||||||
Total liabilities and stockholders equity |
$ | 36,431,885 | $ | 39,404,002 | ||||
See accompanying notes.
F-3
EntreMed, Inc.
Consolidated Statements of Operations
Consolidated Statements of Operations
YEAR ENDED DECEMBER 31, | ||||||||||||
2005 | 2004 | 2003 | ||||||||||
Revenues: |
||||||||||||
Collaborative research and development |
$ | | $ | | $ | 667,796 | ||||||
Licensing |
590,992 | 495,496 | 310,496 | |||||||||
Grants |
| | 508,243 | |||||||||
Royalties |
5,310,439 | 5,918 | 2,705 | |||||||||
Other |
16,624 | 12,581 | 86,306 | |||||||||
5,918,055 | 513,995 | 1,575,546 | ||||||||||
Costs and expenses: |
||||||||||||
Research and development |
17,325,048 | 10,523,252 | 14,252,196 | |||||||||
General and administrative |
5,920,455 | 6,570,664 | 7,022,986 | |||||||||
23,245,503 | 17,093,916 | 21,275,182 | ||||||||||
Investment income |
1,010,771 | 313,940 | 205,580 | |||||||||
Gain on sale of assets |
3,420 | 124,083 | | |||||||||
Gain on sale of securities ( Note 2) |
| 520,000 | | |||||||||
Gain on sale of royalty interest (Note 4) |
| 3,000,000 | | |||||||||
Net loss |
(16,313,257 | ) | (12,621,898 | ) | (19,494,056 | ) | ||||||
Dividends on Series A convertible preferred stock |
(1,005,000 | ) | (1,005,000 | ) | (1,005,000 | ) | ||||||
Net loss attributable to common shareholders |
$ | (17,318,257 | ) | $ | (13,626,898 | ) | $ | (20,499,056 | ) | |||
Net loss per share (basic and diluted) |
$ | (0.36 | ) | $ | (0.37 | ) | $ | (0.68 | ) | |||
Weighted average number of shares outstanding (basic and
diluted) |
48,176,914 | 37,170,544 | 29,943,161 | |||||||||
See accompanying notes.
F-4
ENTREMED, INC.
CONSOLIDATED STATEMENT OF STOCKHOLDERS EQUITY
Periods Ended December 31, 2005, 2004 and 2003
Periods Ended December 31, 2005, 2004 and 2003
Additional | Deferred | |||||||||||||||||||||||||||||||||||
Preferred Stock | Common Stock | Treasury | Paid-in | Stock-Based | Accumulated | |||||||||||||||||||||||||||||||
Shares | Amount | Shares | Amount | Stock | Capital | Compensation | Deficit | Total | ||||||||||||||||||||||||||||
Balance at December 31, 2002 |
3,350,000 | $ | 3,350,000 | 23,270,694 | $ | 241,457 | $ | (8,034,244 | ) | $ | 228,316,897 | $ | (61,846 | ) | $ | (213,318,618 | ) | $ | 10,493,646 | |||||||||||||||||
Issuance of common stock for
options
and warrants exercised |
| | 172,575 | 1,725 | | 189,092 | | | 190,817 | |||||||||||||||||||||||||||
Sale of common stock
at $2.50 per share |
| | 4,100,000 | 41,000 | | 7,125,344 | | | 7,166,344 | |||||||||||||||||||||||||||
Sale of common stock
at $3.00 per share |
| | 3,000,000 | 30,000 | | 8,016,609 | | | 8,046,609 | |||||||||||||||||||||||||||
Sale of common stock
at $4.25 per share |
| | 5,250,000 | 52,500 | | 18,020,631 | | | 18,073,131 | |||||||||||||||||||||||||||
Issuance of common stock and
warrants pursuant
to debt settlement agreements |
| | 1,147,872 | 11,479 | | 1,102,521 | | | 1,114,000 | |||||||||||||||||||||||||||
Recognition of non cash
stock compensation |
| | 31,871 | 319 | | 174,681 | | | 175,000 | |||||||||||||||||||||||||||
Fair value of warrants issued |
| | | | | 4,452,117 | | | 4,452,117 | |||||||||||||||||||||||||||
Change in basis in MaxCyte |
| | | | | 4,579,429 | 61,846 | | 4,641,275 | |||||||||||||||||||||||||||
Net loss |
| | | | | | | (19,494,056 | ) | (19,494,056 | ) | |||||||||||||||||||||||||
Balance at December 31, 2003 |
3,350,000 | $ | 3,350,000 | 36,973,012 | $ | 378,480 | $ | (8,034,244 | ) | $ | 271,977,321 | $ | | $ | (232,812,674 | ) | $ | 34,858,883 | ||||||||||||||||||
Issuance of common stock for
options
and warrants exercised |
| | 208,946 | 2,090 | | 5,677 | | | 7,767 | |||||||||||||||||||||||||||
Sale of common stock
at $2.55 per share |
| | 5,490,198 | 54,902 | | 10,781,472 | | | 10,836,374 | |||||||||||||||||||||||||||
Recognition of non cash
stock compensation |
| | 81,018 | 810 | | 174,189 | | | 174,999 | |||||||||||||||||||||||||||
Fair value of warrants issued |
| | | | | 2,448,629 | | | 2,448,629 | |||||||||||||||||||||||||||
Net loss |
| | | | | | | (12,621,898 | ) | (12,621,898 | ) | |||||||||||||||||||||||||
Balance at December 31, 2004 |
3,350,000 | $ | 3,350,000 | 42,753,174 | $ | 436,282 | $ | (8,034,244 | ) | $ | 285,387,288 | $ | | $ | (245,434,572 | ) | $ | 35,704,754 | ||||||||||||||||||
Issuance of common stock for
options
exercised |
| | 67,070 | 671 | | 80,685 | | | 81,356 | |||||||||||||||||||||||||||
Issuance of common stock for
warrants
exercised |
| | 7,355,166 | 73,552 | | 9,826,448 | | | 9,900,000 | |||||||||||||||||||||||||||
Stock issuance costs |
| | | | | (76,651 | ) | | | (76,651 | ) | |||||||||||||||||||||||||
Deferred stock-based
compensation |
| | 56,448 | 564 | | 174,424 | (174,988 | ) | | | ||||||||||||||||||||||||||
Amortization of deferred
stock-based compensation |
| | | | | | 72,988 | | 72,988 | |||||||||||||||||||||||||||
Net loss |
| | | | | | | (16,313,257 | ) | (16,313,257 | ) | |||||||||||||||||||||||||
Balance at December 31, 2005 |
3,350,000 | $ | 3,350,000 | 50,231,858 | $ | 511,069 | $ | (8,034,244 | ) | $ | 295,392,194 | $ | (102,000 | ) | $ | (261,747,829 | ) | $ | 29,369,190 | |||||||||||||||||
See accompanying notes.
F-5
EntreMed, Inc.
Consolidated Statements of Cash Flows
Consolidated Statements of Cash Flows
YEAR ENDED DECEMBER 31, | ||||||||||||
2005 | 2004 | 2003 | ||||||||||
CASH FLOWS FROM OPERATING ACTIVITIES |
||||||||||||
Net loss |
$ | (16,313,257 | ) | $ | (12,621,898 | ) | $ | (19,494,056 | ) | |||
Adjustments to reconcile net loss to net cash used by operating
activities: |
||||||||||||
Depreciation and amortization |
475,848 | 773,775 | 930,772 | |||||||||
Loss on disposal of equipment |
| | 9,679 | |||||||||
Gain on sale of assets |
(3,420 | ) | (124,083 | ) | | |||||||
Gain on sale of securities |
| (520,000 | ) | | ||||||||
Gain on sale of royalty interest |
| (3,000,000 | ) | | ||||||||
Amortization of deferred stock-based compensation |
72,988 | 174,999 | 175,000 | |||||||||
Amortization of (discount) premium on short term investments |
(393,815 | ) | (15,447 | ) | 205,982 | |||||||
Minority interest |
178 | (127 | ) | (124 | ) | |||||||
Changes in operating assets and liabilities: |
||||||||||||
Accounts receivable |
(472,650 | ) | 178,196 | (209,693 | ) | |||||||
Note receivable |
(1,000,000 | ) | | | ||||||||
Interest receivable |
(96,142 | ) | 177,103 | (262,097 | ) | |||||||
Prepaid expenses and other |
(150,969 | ) | 151,120 | (247,939 | ) | |||||||
Accounts payable |
3,936,601 | (2,248,891 | ) | (4,683,579 | ) | |||||||
Payable to related parties |
28,059 | 47,108 | 153,213 | |||||||||
Accrued liabilities |
(377,469 | ) | 709,483 | (1,119,462 | ) | |||||||
Deferred rent |
(32,931 | ) | (5,709 | ) | 329,815 | |||||||
Deferred revenue |
(190,992 | ) | (97,496 | ) | (93,496 | ) | ||||||
Net cash used in operating activities |
(14,517,971 | ) | (16,421,867 | ) | (24,305,985 | ) | ||||||
CASH FLOWS FROM INVESTING ACTIVITIES |
||||||||||||
Proceeds from sale of property and equipment, net |
11,000 | 355,275 | | |||||||||
Proceeds from sale of securities |
| 520,000 | | |||||||||
Reduction in ownership of subsidiarys cash |
| | (418,108 | ) | ||||||||
Purchases of short term investments |
(51,491,900 | ) | (37,718,991 | ) | (8,585,565 | ) | ||||||
Maturities of short term investments |
47,325,000 | 25,750,000 | 6,250,000 | |||||||||
Purchases of furniture and equipment |
(248,677 | ) | (163,539 | ) | (32,715 | ) | ||||||
Net cash used in investing activities |
(4,404,577 | ) | (11,257,255 | ) | (2,786,388 | ) | ||||||
CASH FLOWS FROM FINANCING ACTIVITIES |
||||||||||||
Net proceeds from sale of common stock |
9,904,705 | 13,292,770 | 33,476,901 | |||||||||
Net proceeds from sale of warrants |
| | 4,452,117 | |||||||||
Payment of principal on note payable |
| | (91,843 | ) | ||||||||
Net cash provided by financing activities |
9,904,705 | 13,292,770 | 37,837,175 | |||||||||
Net increase (decrease) in cash and cash equivalents |
(9,017,843 | ) | (14,386,352 | ) | 10,744,802 | |||||||
Cash and cash equivalents at beginning of year |
20,425,495 | 34,811,847 | 24,067,045 | |||||||||
Cash and cash equivalents at end of year |
$ | 11,407,652 | $ | 20,425,495 | $ | 34,811,847 | ||||||
SUPPLEMENTAL DISCLOSURE OF CASH FLOW INFORMATION |
||||||||||||
Interest paid |
$ | | $ | | $ | 2,451 | ||||||
F-6
EntreMed, Inc.
Notes to Consolidated Financial Statements
1. SUMMARY OF SIGNIFICANT ACCOUNTING POLICIES
DESCRIPTION OF BUSINESS AND BASIS OF PRESENTATION
EntreMed, Inc. (EntreMed) is a clinical-stage pharmaceutical company focused on developing
next generation multi-mechanism oncology and antiinflammatory drugs that target disease cells
directly and the blood vessels that nourish them. EntreMed is focused on developing drugs that are
safe and convenient, and provide the potential for improved patient outcomes. Panzem®
(2-methoxyestradiol or 2ME2), one of the Companys lead drug candidates, is currently in
clinical trials for cancer, as well as in preclinical development for rheumatoid arthritis. MKC-1,
a novel cell cycle inhibitor acquired through the recent acquisition of Miikana Therapeutics, is
also in Phase 2 clinical trials for cancer. ENMD-1198, a novel tubulin binding agent discovered by
EntreMed, has an active Investigational New Drug (IND) application on file with the Food and Drug
Administration (FDA), and will commence a Phase 1 clinical trial in 1H 2006.
In January 2006, EntreMed acquired Miikana Therapeutics, Inc., a clinical-stage
biopharmaceutical company headquartered in Fremont, California with research laboratories in
Toronto, Canada. As a result of the transaction, EntreMed enhanced its pipeline with the addition
of a Phase 2 drug candidate, MKC-1, and two preclinical programs, one in aurora kinase inhibition
and one in HDAC inhibition.
EntreMeds goal is to develop and commercialize therapeutics based on the Companys scientific
expertise in angiogenesis, cell cycle regulation and inflammation processes vital to the
progression of cancer and other diseases. The Companys three clinical product candidates are
based on these mechanisms. The Companys expertise has also led to the identification of new
molecules, including new chemical entities derived from 2ME2, modulators of fibroblast growth
factor-2 (FGF-2) activity, proteinase activated receptor-2 (PAR-2) antagonists, and tissue factor
pathway inhibitor (TFPI) peptides.
In order to further advance its commercial objectives, EntreMed may seek strategic alliances,
licensing relationships and co-development partnerships with other companies to develop compounds
for both oncology and non-oncology therapeutic areas.
The accompanying consolidated financial statements include the accounts of the Companys
controlled subsidiary, Cytokine Sciences, Inc. All intercompany balances and transactions have
been eliminated in consolidation.
To date, the Company has been engaged primarily in research and development activities. As a
result the Company has incurred operating losses through 2005 and expects to continue to incur
operating losses for 2006 and the foreseeable future before commercialization of any products.
To accomplish the Companys business plans, EntreMed will be required to continue to conduct
substantial development activities for all proposed products. The Company intends to continue to
pursue strategic relationships to provide resources for the further development of our product
candidates. There can be no assurance, however, that these discussions will result in relationships
or additional funding. In addition, the Company will continue to seek capital through the public
or private sale of securities. There can be no assurance that EntreMed will be
successful in seeking such additional capital.
SEGMENT INFORMATION
The Company currently operates in one business segment, which is the development of
therapeutics primarily for the treatment of cancer. The Company is managed and operated as one
business. A single management team that reports to the Companys President and Chief Executive
Officer comprehensively manages the entire business. The Company does not operate separate lines of
business with respect to its products or product
candidates. Accordingly, the Company does not have separately reportable segments as defined
by FASB Statement No. 131, Disclosures about Segments of an Enterprise and Related Information.
F-7
RESEARCH AND DEVELOPMENT
Research and development expenses consist primarily of compensation and other expenses related
to research and development personnel, research collaborations, costs associated with pre-clinical
testing and clinical trials of our product candidates, including the costs of manufacturing the
product candidates, and facilities expenses. Research and development costs are expensed as
incurred, including costs incurred in filing, defending and maintaining patents.
PROPERTY AND EQUIPMENT
Furniture and equipment and leasehold improvements are stated at cost and are depreciated over
their estimated useful lives of 5 to 10 years. Depreciation is determined on a straight-line basis.
Depreciation expense was $475,848, $773,775 and $930,772 in 2005, 2004 and 2003, respectively.
Property and equipment consists of the following:
DECEMBER 31 | ||||||||
2005 | 2004 | |||||||
Furniture and equipment |
$ | 4,321,634 | $ | 5,575,772 | ||||
Leasehold improvements |
1,288,791 | 1,284,991 | ||||||
5,610,425 | 6,860,763 | |||||||
Less: accumulated depreciation |
(4,695,088 | ) | (5,710,676 | ) | ||||
$ | 915,337 | $ | 1,150,087 | |||||
CASH AND CASH EQUIVALENTS
Cash and cash equivalents include cash and highly liquid investments with original maturities
of less than 90 days. Substantially all of the Companys cash equivalents are held in short-term
money market accounts of banks and brokerage houses.
SHORT-TERM INVESTMENTS
Short-term investments consist primarily of corporate debt securities, all of which mature
within one year. The Company has classified these investments as available for sale. Such
securities are carried at cost which approximates market. The cost of securities sold is calculated
using the specific identification method. Unrealized gains and losses on these securities, if any,
are reported as accumulated other comprehensive income (loss), which is a separate component of
stockholders equity. There were no unrealized gains or losses as of December 31, 2005 and 2004.
Realized gains and losses and declines in value judged to be other than temporary on securities
available for sale, if any, are included in operations. Short-term investments are principally
uninsured and subject to normal credit risk. (See Note 2 for Gain on Sale of Securities)
ACCOUNTS RECEIVABLE
Accounts receivable are stated net of allowances for doubtful accounts. Allowances for
doubtful accounts are determined on a specific item basis. Management reviews the credit
worthiness of individual customers and past payment history to determine the allowance for doubtful
accounts. There is no allowance for doubtful accounts at December 31, 2005 and 2004.
As of December 31, 2005 and 2004, one individual customer represented 98% and 92%,
respectively, of the total accounts receivable.
F-8
NOTE RECEIVABLE
In December 2005, the Company loaned Miikana Therapeutics $1 million to repay certain
obligations. This is reflected as a note receivable in the accompanying December 31, 2005
consolidated balance sheet. Upon consummation of the acquisition of
Miikana Therapeutics (See Note 11), the note receivable was used to
pay a portion of the purchase price obligation.
INCOME TAXES
Income taxes have been provided using the liability method in accordance with Statement of
Financial Accounting Standards No. 109, Accounting for Income Taxes.
REVENUE RECOGNITION
The Company recognizes revenue in accordance with the provisions of Staff Accounting Bulletin
No. 104, Revenue Recognition, whereby revenue is not recognized until it is realized or realizable
and earned. Revenue is recognized when all of the following criteria are met: persuasive evidence
of an arrangement exists, delivery has occurred or services have been rendered, the price to the
buyer is fixed and determinable and collectibility is reasonably assured.
Royalty Revenue Royalties from licenses are based on third-party sales and recorded as
earned in accordance with contract terms, when third-party results are reliably measured and
collectibility is reasonably assured. The majority of the Companys 2005 revenues were from
royalties on the sale of Thalomid®, which the Company began to recognize in the third
quarter. In 2004 certain provisions of a purchase agreement dated June 14, 2001 by and between
Bioventure Investments kft (Bioventure) and the Company were satisfied, and, as a result, in 2005
the Company is entitled to share in the royalty payments received by Royalty Pharma Finance Trust,
successor to Bioventure, on annual Thalomid® sales above a certain threshold. Based on
the licensing agreement royalty formula, annual royalty sharing commences with Thalomid®
annual sales of approximately $225 million. The Company is eligible to receive royalty payments
under a February 2004 agreement with Childrens Medical Center Corporation (CMCC) and Alchemgen
Therapeutics. Under the agreement, Alchemgen received rights to market endostatin and angiostatin
in Asia. Future royalty payments, if any, will be recorded as revenue when received and/or when
collectibility is reasonably assured.
Licensing Revenue The Company has recognized licensing revenues resulting from the January
2002 five-year strategic alliance with Allergan to develop and commercialize small molecule
angiogenic inhibitors for treatment and prevention of diseases and conditions of the eye. The
initial net fee was being amortized to revenue over the five-year license term. In April 2005,
Allergan terminated the license in accordance with its terms, which resulted in the accelerated
recognition of deferred revenue. In February 2004 the Company transferred rights to the proteins,
endostatin and angiostatin, in an agreement with Childrens Medical Center Corporation (CMCC) and
Alchemgen Therapeutics. Under the agreement, the Company received an upfront and a second cash
payment. The upfront licensing cash payment was fully amortized in 2004, as the Company had
completed its obligations to transfer data and material. Due to rights negotiations between the
licensee and CMCC, the second and final licensing cash payment in the amount of $400,000 was
received in May 2005. Management concluded collectibility was not reasonably assured until the
funds were received. In September 2003 the Company entered into a licensing agreement with Oxford
Biomedica, PLC and Oxford Biomedica (UK) Limited for the use of endostatin and angiostatin genes in
the development of locally delivered gene therapy for ophthalmologic applications. Under the
agreement, the Company had no continuing obligations. As such, the Company recorded as revenue the
value of the initial net cash and shares of common stock received under the agreement.
Collaborative Research and Development Revenue In 2003 the Company received revenues for
performance under commercial research and development contracts. These contracts required that the
Company provide services directed toward specific objectives and include developmental milestones
and deliverables. These revenues were recognized at the time that research and development
activities were performed.
F-9
Grant Revenue In 2003 the Company received a government grant to financially support our
Phase II Endostatin clinical trial in patients with neuroendocrine tumors. Grants are funded in
specific amounts based on funding requests submitted to the grantor. Grant revenues are recognized
and realized at the time that research and development activities are performed.
NET LOSS PER SHARE
Net loss per share (basic and diluted) was computed by dividing net loss available to common
stock by the weighted average number of shares of common stock outstanding. Common stock
equivalents, including Preferred Series A common stock equivalents, totaling 27,781,813 were
anti-dilutive and, therefore, were not included in the computation of weighted average shares used
in computing diluted loss per share.
COMPREHENSIVE LOSS
Under Financial Accounting Standard No. 130 (SFAS 130), Reporting Comprehensive Income, the
Company is required to display comprehensive loss and its components as part of the consolidated
financial statements. Comprehensive loss is comprised of the net loss and other comprehensive
income (loss), which includes certain changes in equity that are excluded from net loss.
Comprehensive loss for the Company was the same as net loss for all years presented.
STOCK-BASED COMPENSATION
The Company recognizes expense for stock-based compensation arrangements in accordance with
the provisions of Accounting Principles Board Opinion No. 25, Accounting for Stock Issued to
Employees, and related Interpretations. Accordingly, compensation cost is recognized for the excess
of the estimated fair value of the stock at the grant date over the exercise price, if any. The
Company accounts for equity instruments issued to non-employees in accordance with EITF 96-18,
Accounting for Equity Instruments that are Issued to Other Than Employees for Acquiring, or in
Conjunction with Selling, Goods, or Services.
F-10
Disclosures regarding alternative fair values of measurement and recognition methods
prescribed by Statement of Accounting Standards No. 148, Accounting for Stock-Based
Compensation-Transition and Disclosure and Statement of Financial Accounting Standards No. 123,
Accounting for Stock-Based Compensation (SFAS No. 123) are presented in the table below. The
following table illustrates the effect on net loss if the Company had applied the fair value
recognition provisions of SFAS No. 123, to stock-based compensation:
Year ended December 31 | ||||||||||||
2005 | 2004 | 2003 | ||||||||||
Actual net loss |
$ | (16,313,257 | ) | $ | (12,621,898 | ) | $ | (19,494,056 | ) | |||
Add: Stock-based employee
compensation included in
reported net loss |
72,988 | 174,999 | 175,000 | |||||||||
Deduct: Stock-based employee
compensation expense if SFAS
No. 123 had been applied to all
awards |
(1,923,575 | ) | (6,664,579 | ) | (10,691,564 | ) | ||||||
Proforma net loss |
$ | (18,163,844 | ) | $ | (19,111,478 | ) | $ | (30,010,620 | ) | |||
Dividend on Series A convertible
preferred stock |
$ | (1,005,000 | ) | $ | (1,005,000 | ) | $ | (1,005,000 | ) | |||
Proforma net loss per share
available to common
shareholders |
$ | (19,168,844 | ) | $ | (20,116,478 | ) | $ | (31,015,620 | ) | |||
Net loss per share: |
||||||||||||
Basic and diluted as reported |
$ | (0.36 | ) | $ | (0.37 | ) | $ | (0.68 | ) | |||
Basic and diluted pro forma |
$ | (0.40 | ) | $ | (0.54 | ) | $ | (1.03 | ) |
The effect of applying SFAS No. 123 on a pro forma net loss as stated above is not necessarily
representative of the effect on reported net loss for future years due to, among other things, the
vesting period of the stock options and the fair value of additional options to be granted in
future years.
F-11
Significant assumptions used in the Black-Scholes option pricing model are as follows:
Year ended December 31, | ||||||||||||
2005 | 2004 | 2003 | ||||||||||
Risk free rate |
4.25 | % | 3.73 | % | 4.0 | % | ||||||
Expected life |
5 yrs | 5 yrs | 6 yrs | |||||||||
Volatility |
1.08 | 1.20 | 1.20 | |||||||||
Expected dividend yield |
| | |
FINANCIAL INSTRUMENTS AND CONCENTRATIONS OF RISK
Financial instruments that potentially subject the Company to concentrations of credit risk
consist principally of cash and cash equivalents, short-term investments, accounts receivable and
note receivable. The Company maintains its cash and cash equivalents in bank deposit accounts,
which, at times, may exceed federally insured amounts. The Company believes it is not exposed to
any significant credit risk on cash and cash equivalents or short-term investments. The carrying
amount of current assets and liabilities approximates their fair values due to their short-term
maturities.
RECENT ACCOUNTING STANDARDS
In October 2004, the FASB concluded that SFAS No. 123 (revised 2004),
Share-Based Payment (SFAS 123R), which requires all companies to measure compensation cost for
all share-based payments (including employee stock options) at fair value, originally effective for
interim or annual periods beginning after June 15, 2005. SFAS 123R provides two tentative adoption
methods. The first method is a modified prospective transition method whereby a company would
recognize share-based employee costs from the beginning of the fiscal period in which the
recognition provisions are first applied as if the fair-value-based accounting method had been used
to account for all employee awards granted, modified, or settled after the effective date and to
any awards that were not fully vested as of the effective date. Measurement and attribution of
compensation cost for awards that are unvested as of the effective date of SFAS 123R would be based
on the same estimate of the grant-date fair value and the same attribution method used previously
under SFAS 123. The second adoption method is a modified retrospective transition method whereby a
company would recognize employee compensation cost for periods presented prior to the adoption of
SFAS 123R in accordance with the original provisions of SFAS 123; that is, an entity would
recognize employee compensation costs in the amounts reported in the pro forma disclosures provided
in accordance with SFAS 123. A company would not be permitted to make any changes to those amounts
upon adoption of SFAS 123R unless those changes represent a correction of an error. For periods
after the date of adoption of SFAS 123R, the modified prospective transition method described above
would be applied.
In April 2005, the SEC announced that it would provide for a phased-in implementation process
for SFAS 123R; therefore, the Company currently expects to adopt SFAS 123R in the quarter ended
March 31, 2006, using the modified prospective method, although the Company continues to review its
options for adoption under this new pronouncement. The Company currently anticipates that the
impact of adopting SFAS 123R will result in recording approximately $2 million of employee
compensation expense in 2006, but could fluctuate depending on levels of share-based payments
granted.
In November 2005, the Financial Accounting Standards Board issued FASB Staff Position (FSP)
FAS 115-1, The Meaning of Other-Than-Temporary Impairment and Its Application to Certain
Investments (FSP FAS 115-1). FSP FAS 115-1, which is effective for reporting periods beginning
after December 15, 2005, provides guidance on other-than-temporary impairment models for marketable
debt and equity securities accounted for under
F-12
SFAS No. 115 and non-marketable equity securities accounted for under the cost method. FSP FAS
115-1 provides a basic three-step model to evaluate whether an investment is other-than-temporarily
impaired. Under FSP FAS 115-1, other than temporary losses on short-term investments would be
expensed rather than included in stockholders equity. The Company currently does not have any
investments scheduled to mature greater than one year. The Company does not believe that the
adoption of FSP FAS 115-1 will have a material effect on the Companys results of operations,
financial condition or liquidity.
USE OF ESTIMATES
The preparation of consolidated financial statements in conformity with accounting principles
generally accepted in the United States requires management to make estimates and assumptions that
affect the amounts reported in the financial statements and accompanying notes. Actual results may
differ from those estimates, and such differences may be material to the financial statements.
RECLASSIFICATIONS
Certain prior-year amounts have been reclassified to conform to the current year presentation.
2. SALE OF SECURITIES
In September 2004, the Company sold certain securities of an independent private biotechnology
company. The securities were acquired in 1996 through 1999 and accounted for using the equity
method. Consistent with this approach, the cost of these securities was written down to $0 in
prior periods as the Company recorded its share of losses, and the Company had no residual cost
basis in the securities when sold. As such, the Company recorded a gain on the sale in 2004 equal
to the sale proceeds of $520,000.
3. SPONSORED RESEARCH PROGRAM AGREEMENTS
Prior to 2003, the Company entered into several agreements to sponsor external research
programs. The Companys primary external research program agreement was entered into with the
Childrens Hospital, in Boston, Massachusetts, an entity affiliated with Harvard Medical School
(Childrens Hospital, Boston).
Under this sponsored research agreement, the Company agreed to pay Childrens Hospital, Boston
to continue the research on the role of angiogenesis in pathological conditions. In accordance
with the terms of this sponsored research agreement, the Company agreed to pay $1,500,000 each year
to Childrens Hospital, Boston. As of December 31, 2003, all amounts were paid and there was no
remaining commitment.
In November 2003, the Company reached an agreement to extend one of the sponsored research
agreements with Childrens Hospital, Boston. Under the extended agreement the Company paid
$300,000 each year for the period of November 1, 2003 through November 1, 2005. Payments under the
agreement were made quarterly in advance and were expensed as paid. The Company has made all
sponsored research payments due Childrens Hospital, Boston as of December 31, 2005.
4. LICENSE AGREEMENTS
On January 18, 2002 the Company entered into a five-year strategic alliance with Allergan, an
ophthalmic research and development and pharmaceutical company, to develop and commercialize small
molecule angiogenic inhibitors for treatment and prevention of diseases and conditions of the eye
(the Agreement). In April 2005, Allergan terminated the license in accordance with its terms,
which resulted in the accelerated recognition of deferred revenue.
On September 8, 2003 the Company entered into a licensing agreement with Oxford Biomedica, PLC
and Oxford Biomedica (UK) Limited. Under the terms of the agreement, Oxford BioMedica receives
exclusive worldwide rights to use EntreMeds endostatin and angiostatin genes in the development of
locally delivered gene
F-13
therapy for ophthalmologic applications. Oxford BioMedica plans to utilize EntreMeds genes
in its proprietary therapeutic RetinoStatTM program for the treatment of age-related
macular degeneration and diabetic retinopathy. In return, EntreMed received an initial cash
payment of $125,000 and 301,748 shares of Oxford BioMedica common stock valued at $129,000.
Additionally, EntreMed may collect up to $10 million on the achievement of regulatory and clinical
milestones. The Company may receive royalties on future worldwide sales of products resulting from
the agreement. There can be no assurance that the Company will receive any additional amounts
under this agreement.
In February 2004, the Company transferred rights for its protein-based drug candidate
programs, endostatin and angiostatin, in an agreement with Childrens Medical Center Corporation in
Boston (CMCC) and Alchemgen Therapeutics, Inc. (Alchemgen). Under the agreement, CMCC and
Alchemgen are continuing the development of endostatin and angiostatin and bear all expenses
associated with the programs, including costs that the Company may incur in transferring these
compounds. In exchange, the Company receives upfront and future cash and royalty payments. Under
the terms of the three-party agreement, the Houston-based, privately-held company Alchemgen
received exclusive rights to market endostatin and angiostatin in Asia. CMCC holds the
endostatin and angiostatin license for the rest of the world therefore the Company has no future
milestone payment obligations related to endostatin and angiostatin. The Company would receive 20%
of all future proceeds (e.g. upfront, milestone and royalty payments) resulting from any subsequent
CMCC license outside of Asia; however, there can be no assurance that the Company will receive
future additional payments under this arrangement.
Pursuant to a purchase agreement dated June 14, 2001 by and between Bioventure Investments kft
(Bioventure) and the Company, as amended July 13, 2001, July 30, 2001 and August 3, 2001 (the
Purchase Agreement), Bioventure purchased all of the Companys right, title and interest to the
net royalty payments payable by Celgene Corporation (Celgene) to the Company under the agreement
dated as of December 9, 1998 by and between the Company and Celgene (the Celgene Sublicense).
A provision of the Bioventure purchase agreement provided the potential for an adjustment in
the purchase price if cumulative sales of Thalomid® exceeded $800 million by December 31, 2004.
Based on Thalomid® sales reported publicly by Celgene, the Company concluded that cumulative
Thalomid® sales had reached this milestone by December 31, 2004. As such, the Company recorded a
gain on the sale of our royalty rights and a corresponding receivable of $3.0 million. The Company
received payment of this amount in March 2005. In addition to triggering this one-time adjustment
in the purchase price, exceeding the $800 million cumulative sales amount also triggers a royalty
sharing provision. Beginning the year after cumulative sales reach $800 million, EntreMed is
entitled to share in the royalty payments received by Royalty Pharma Finance Trust, successor to
Bioventure, on annual Thalomid® sales above a certain threshold. In 2005 Thalomid®
sales surpassed the royalty-sharing point and we recorded estimated royalty revenues of $5,310,000.
There can be no assurance that the Company will receive additional material royalties under the
royalty sharing provision in the future.
In March 2005, we entered into an exclusive worldwide license agreement with Celgene
Corporation for the development and commercialization of Celgenes small molecule tubulin inhibitor
compounds for the treatment of cancer. Under the terms of the agreement, Celgene received an
upfront licensing fee and may receive additional payments upon successful completion of certain
clinical, regulatory and sales milestones. No such milestones have been reached through December
31, 2005. EntreMed will assume responsibility for preclinical and clinical development of the
tubulin inhibitors for oncology applications. The upfront license fee of $1,000,000 was recorded as
a component of research and development expense in the Consolidated Statement of Operations for the
year ended December 31, 2005.
5. RELATED PARTY TRANSACTIONS
The Company receives
legal services from a law firm with which one of the Companys officers is associated.
Total expenses for service from this law firm were $1,180,000,
$1,015,000 and $1,192,000 in 2005, 2004, and 2003, respectively. The
amounts reflected as research and development, of $779,000, $628,000
and $916,000 in 2005, 2004 and 2003, respectively, in
the table below primarily represent patent work. The amounts
reflected as general and administrative, of $351,000, $387,000 and
$276,000 in 2005, 2004 and 2003, respectively, represent legal services.
F-14
In addition to legal services, the Company also received financial advisory services from
Ferghana Partners, Inc., a provider of corporate financial advice to firms in the Life Sciences
field. The Companys chairman also serves as non-executive chairman of Ferghana Partners, Inc.
The Companys chairman and CEO both hold a de minimis ownership interest in Ferghana Partners, Inc.
Pursuant to a series of business transactions the Company paid
$785,000, $100,000 and $3,205,000 in fees to
Ferghana Partners, Inc. in 2005, 2004 and 2003, respectively. The 2005 amount includes financial
advisory fees of $60,000, a $200,000 fee associated with the March 2005 Celgene license agreement
and a $525,000 fee resulting from Celgenes March 2005 exercise of a warrant, issued as part of the
2002 transaction, for 7,000,000 shares of common stock. The $60,000
paid in advisory fees was
recorded as general and administrative costs and the $200,000 fee was recorded as research and
development expense. The balance of the 2005 fees paid to Ferghana and all of the fees
previously paid to Ferghana in 2004 and 2003 were recorded as offsets against gross
equity transaction proceeds and, as such, are not reflected as expenses in the current period.
The 2005 research and development amount also includes a $1 million upfront licensing fee paid
to Celgene pursuant to our license of Celgenes tubulin inhibitor program. Expenses from related
parties are included in the following accounts within the consolidated financial statements,
$228,380 and $200,321 of which are included in accounts payable at December 31, 2005 and 2004,
respectively:
2005 | 2004 | 2003 | ||||||||||
Research and development |
$ | 1,979,000 | $ | 628,000 | $ | 916,000 | ||||||
General and administrative |
411,000 | 387,000 | 276,000 | |||||||||
Additional paid in capital |
525,000 | 100,000 | 3,205,000 | |||||||||
Capitalized acquisition costs |
50,000 | | | |||||||||
$ | 2,965,000 | $ | 1,115,000 | $ | 4,397,000 | |||||||
6. INCOME TAXES
The Company has net operating loss carryforwards for income tax purposes of approximately
$261,541,000 at December 31, 2005 ($249,988,000 at December 31, 2004) that expires in years 2006
through 2025. The Company also has research and development tax credit carryforwards of
approximately $10,616,000 as of December 31, 2005 that expire in years 2007 through 2025. These net
operating loss carryforwards include approximately $19,800,000, related to exercises of stock
options for which the income tax benefit, if realized, would increase additional paid-in capital.
The utilization of the net operating loss and research and development carryforwards may be limited
in future years due to changes in ownership of the Company pursuant to Internal Revenue Code
Section 382. For financial reporting purposes, a valuation allowance has been recognized to reduce
the net deferred tax assets to zero due to uncertainties with respect to the Companys ability to
generate taxable income in the future sufficient to realize the benefit of deferred income tax
assets.
Deferred income taxes reflect the net effect of temporary differences between the carrying
amounts of assets and liabilities for financial reporting purposes and the amounts used for income
tax purposes. Significant components of the Companys deferred income tax assets and liabilities
as of December 31, 2005 and 2004 are as follows:
DECEMBER 31, | ||||||||
2005 | 2004 | |||||||
Deferred income tax assets (liabilities): |
||||||||
Net operating loss carryforwards |
$ | 101,005,000 | $ | 94,995,000 | ||||
Research and development credit carryforward |
10,616,000 | 10,889,000 | ||||||
Deferred revenues |
| 88,000 | ||||||
Equity investment |
70,000 | 69,000 | ||||||
Other |
735,000 | 721,000 | ||||||
Depreciation |
161,000 | 263,000 | ||||||
Valuation allowance for deferred income tax assets |
(112,587,000 | ) | (107,025,000 | ) | ||||
Net deferred income tax assets |
$ | | $ | | ||||
F-15
A reconciliation of the provision for income taxes to the federal statutory rate is as
follows:
2005 | 2004 | 2003 | ||||||||||
Tax benefit at statutory rate |
$ | (5,547,000 | ) | $ | (4,291,000 | ) | $ | (7,034,000 | ) | |||
State taxes |
(754,000 | ) | (505,000 | ) | (826,000 | ) | ||||||
R & D credits |
273,000 | 383,000 | (853,000 | ) | ||||||||
Permanent M-1s |
29,000 | 6,000 | 9,000 | |||||||||
Change in valuation allowance |
7,565,000 | 4,407,000 | 8,704,000 | |||||||||
Change in estimated effective rate |
(1,566,000 | ) | | | ||||||||
$ | | $ | | $ | | |||||||
7. STOCKHOLDERS EQUITY
In 2002, the Company issued 3,350,000 shares of Series A Preferred Stock to Celgene.
The Series A Preferred Stock is convertible, at the option of Celgene, at any time, into common
stock at an initial per share conversion price of $5.00 (1 share of preferred converts into 5
shares of common). The conversion price is subject to change for certain dilutive events, as
defined. At any time after December 31, 2005, the Company may cause the Series A Preferred Stock
to convert automatically provided all of the following conditions are met:
(i) | As of the conversion date, the common stock is traded and was traded during the 60 trading days preceding the conversion date, on a national securities exchange; | ||
(ii) | The average per share closing price of the common stock is greater than $5.00 over a 60-trading day period ending on the conversion date, and | ||
(iii) | A registration statement with respect to resale of the common stock issuable in the conversion to the holders of the Series A Preferred Stock has been filed with the SEC, such registration statement is effective and the Company has agreed to maintain the effectiveness of the registration statement for at least 180 consecutive days beginning with the conversion date. |
The Series A Preferred Stock accrues and accumulates dividends at a rate of 6% and will
participate in dividends declared and paid on the common stock, if any. At December 31, 2005,
cumulative unpaid preferred stock dividends totaled $3,015,000 or $.90 per share. All unpaid
preferred stock dividends must be paid before any dividends may be declared or paid on the Common
Stock, and will be added to the liquidation preference of the Series A Preferred Stock payable upon
the liquidation, dissolution or winding up of the Company. The liquidation preference is equal to
the greater of:
(i) | Two times the original per share purchase price plus accrued and unpaid dividends or | ||
(ii) | The amount per share that would be payable to a holder of shares of the Series A Preferred Stock had all of the shares been converted to common stock immediately prior to a liquidation event. |
The liquidation preference of the Series A Preferred Stock on a converted basis at December
31, 2005 totaled approximately $33,500,000, excluding cumulative unpaid preferred stock dividends
as discussed above. This value is calculated based on the contractual liquidation preference
articulated in the Series A Preferred Stock agreement. There can be no assurance what impact the
conversion of the Series A Preferred to common stock would have on the trading value of the
Companys common stock.
Holders of the Series A Preferred Stock generally vote together with the holders of common
stock, with each share of Series A Preferred Stock representing the number of votes equal to that
number of shares of common stock into which it is then convertible.
In April 2003, the Company completed a private placement of 4,100,000 shares of its common
stock and warrants to purchase a total of 1,025,000 shares of common stock at an exercise price of
$3.375, resulting in gross proceeds, prior to the deduction of fees and commissions of
approximately $10.25 million (net proceeds of $9 million).
In May 2003, the Company completed a private placement of 3,000,000 shares of its common stock
resulting in gross proceeds, prior to the deduction of fees and commissions of $9 million (net
proceeds of $8.1 million).
F-16
In November 2003, the Company completed a private placement of 5,250,000 shares of its
common stock and warrants to purchase a total of 787,500 shares of common stock at an exercise
price of $5.00, resulting in gross proceeds, prior to the deduction of fees and commissions of
approximately $22.3 million (net proceeds of $20.7 million).
In conjunction with the three 2003 transactions described above, we issued to Ferghana
Securities, Inc., warrants to purchase 123,500 shares of our common stock at exercise prices
ranging from $2.75 to $4.67 for services as financial advisors. The fair value of the warrants
issued to Ferghana Securities, Inc. in 2003 ranged from $1.72 to $3.08.
In December 2004, the Company completed a private placement of 5,490,198 shares of its common
stock and warrants to purchase a total of 1,098,040 shares of common stock at an exercise price of
$3.67, resulting in gross proceeds, prior to the deduction of fees and commissions of approximately
$14.0 million (net proceeds of $13.3 million).
In March 2005, the Company issued 7,000,000 shares of its common stock pursuant to the
exercise of a warrant held by Celgene Corporation. The warrant, exercisable at $1.50 per share was
issued to Celgene as part of the 2002 transaction and resulted in gross proceeds, prior to the
deduction of fees and commissions of $10.5 million (net proceeds of $9.9 million).
8. STOCK OPTIONS AND WARRANTS
The Company has adopted incentive and nonqualified stock option plans whereby 10,983,333
shares of the Companys common stock were reserved for grants to various executive, scientific and
administrative personnel of the Company as well as outside directors and consultants, of which
631,176 shares remain available for grant under the Companys 2001 Long-term Incentive Plan as of
December 31, 2005. These options vest over periods varying from immediately to four years and
generally expire 10 years from the date of grant.
The Company recorded non-cash compensation charges of $73,000, $175,000 and $175,000 in 2005,
2004 and 2003 related to the issuance of restricted stock to members of our Board of Directors, as
each Non-employee director receives as an annual retainer fee of $25,000 that is payable in
restricted stock. As of December 31, 2005 and 2004, $102,000 and $0, is reflected as deferred
compensation related to the vesting of this stock.
The Black-Scholes option valuation model was developed for use in estimating the fair value of
traded options which have no vesting restrictions and are fully transferable. In addition, option
valuation models require the input of highly subjective assumptions including the expected stock
price volatility. Because the Companys employee stock options have characteristics significantly
different from those of traded options, and because changes in the subjective input assumptions can
materially affect the fair value estimate, in managements opinion, the existing models do not
necessarily provide a reliable single measure of the fair value of its employee stock options.
For purposes of pro forma disclosures, the estimated fair values of the options and warrants
are amortized to expense over the vesting period. The estimated weighted average fair value per
option granted in 2005, 2004 and 2003 was $2.13, $2.00 and $2.43, respectively.
F-17
A summary of the Companys stock options and warrants granted to employees and directors and
related information for the years ended December 31 follows:
Weighted Average | ||||||||
Number of Options | Exercise Price | |||||||
Outstanding at January 1, 2003 |
6,609,265 | $ | 12.04 | |||||
Exercised |
(172,575 | ) | $ | 1.42 | ||||
Granted |
1,487,505 | $ | 2.75 | |||||
Canceled |
(416,918 | ) | $ | 11.32 | ||||
Outstanding at December 31, 2003 |
7,507,277 | $ | 10.48 | |||||
Exercised |
(7,125 | ) | $ | 1.09 | ||||
Granted |
1,176,728 | $ | 2.39 | |||||
Canceled |
(308,902 | ) | $ | 6.29 | ||||
Outstanding at December 31, 2004 |
8,367,978 | $ | 9.33 | |||||
Exercised |
(67,070 | ) | $ | 1.21 | ||||
Granted |
604,692 | $ | 2.68 | |||||
Canceled |
(943,582 | ) | $ | 12.66 | ||||
Outstanding at December 31, 2005 |
7,962,017 | $ | 9.04 | |||||
Exercisable at December 31, 2005 |
6,736,260 | $ | 10.23 | |||||
The following summarizes information about stock options granted to employees and
directors outstanding at December 31, 2005:
Options Outstanding | Options Exercisable | |||||||||||||||||||||
Weighted | ||||||||||||||||||||||
Average | Weighted | Weighted | ||||||||||||||||||||
Number | Remaining | Average | Number | Average | ||||||||||||||||||
Range of | Outstanding | Contractual | Exercise | Exercisable | Exercise | |||||||||||||||||
Exercise Prices | at 12/31/05 | Life in Years | Price | at 12/31/05 | Price | |||||||||||||||||
$0.00-$1.50 |
1,200,119 | 6.9 | $ | 1.10 | 1,110,203 | $ | 1.10 | |||||||||||||||
$1.51-$3.00 |
1,453,566 | 8.5 | $ | 2.17 | 770,035 | $ | 2.21 | |||||||||||||||
$3.01-$4.50 |
994,205 | 8.2 | $ | 3.54 | 565,643 | $ | 3.76 | |||||||||||||||
$4.51-$6.00 |
256,272 | 6.9 | $ | 5.04 | 232,524 | $ | 5.01 | |||||||||||||||
$6.01-$10.00 |
1,392,650 | 4.6 | $ | 9.12 | 1,392,650 | $ | 9.12 | |||||||||||||||
$10.01-$15.00 |
945,684 | 3.1 | $ | 12.77 | 945,684 | $ | 12.77 | |||||||||||||||
$15.01-$25.00 |
1,139,450 | 4.0 | $ | 18.78 | 1,139,450 | $ | 18.78 | |||||||||||||||
$25.01-$35.00 |
557,948 | 4.1 | $ | 27.60 | 557,948 | $ | 27.60 | |||||||||||||||
$35.01-$50.00 |
3,611 | 4.6 | $ | 44.82 | 3,611 | $ | 44.82 | |||||||||||||||
$50.01-$65.00 |
18,512 | 4.3 | $ | 53.20 | 18,512 | $ | 53.20 | |||||||||||||||
7,962,017 | 5.9 | $ | 9.04 | 6,736,260 | $ | 10.23 | ||||||||||||||||
Warrants granted generally expire after 5 years from the date of grant. Stock warrant
activity to non-employees is as follows:
Weighted Average | ||||||||
Number of Shares | Exercise Price | |||||||
Outstanding at January 1, 2003 |
10,111,580 | $ | 4.13 | |||||
Granted |
1,936,001 | $ | 4.06 | |||||
Outstanding at December 31, 2003 |
12,047,581 | $ | 4.12 | |||||
Granted |
1,098,040 | $ | 3.67 | |||||
Exercised |
(337,500 | ) | $ | .94 | ||||
Expired |
(566,071 | ) | $ | 32.55 | ||||
Outstanding at December 31, 2004 |
12,242,050 | $ | 2.85 | |||||
Granted |
| | ||||||
Exercised |
(7,562,500 | ) | $ | 1.46 | ||||
Expired |
(74,672 | ) | $ | 6.60 | ||||
Outstanding at December 31, 2005 |
4,604,878 | $ | 5.06 | |||||
Exercisable at December 31, 2005 |
4,604,878 | $ | 5.06 | |||||
F-18
9. COMMITMENTS AND CONTINGENCIES
Commitments
The Company entered into two license agreements with Childrens Hospital, Boston for the
exclusive, worldwide, royalty-bearing licenses to make, use and sell Endostatin and
2-methoxyestradiol (2ME2), both inhibitors of angiogenesis. In February 2004, the Company
transferred rights to Endostatin in an agreement with Childrens Medical Center Corporation and
Alchemgen Therapeutics. Therefore, the Company has no future milestone payment obligations related
to endostatin. In consideration for retaining the 2ME2 rights, the Company must pay a royalty on
any sublicensing fees, as defined in the agreement, to Childrens Hospital, Boston. The agreement
obligates the Company to pay up to $1,000,000 upon the attainment of certain milestones. As of
December 31, 2005, the Company has paid $150,000 under this agreement for the milestones that have
been achieved to date.
In March 2005, the Company entered into an exclusive worldwide license agreement with Celgene
Corporation for the development and commercialization of Celgenes small molecule tubulin inhibitor
compounds for the treatment of cancer. Under the terms of the agreement, Celgene received an
upfront licensing fee of $1,000,000 and may receive additional payments up to approximately $25.25
million based upon the attainment of certain milestones. No such milestones have been reached
through December 31, 2005.
The Company leases its primary facilities through February 2009. The lease agreement provides
for escalation of the lease payments over the term of the lease; however, rent expense is
recognized under the straight-line method. Additionally, the Company leases office equipment under
operating leases. The future minimum payments under its facilities and equipment leases as of
December 31, 2005 are as follows:
2006 |
974,954 | |||
2007 |
991,534 | |||
2008 |
1,021280 | |||
2009 |
171,644 | |||
Thereafter |
| |||
Total minimum payments |
$ | 3,159,412 | ||
Rental expense for the years ended December 31, 2005, 2004 and 2003 was $929,000,
$926,000, and $1,296,000, respectively.
Contingencies
EntreMed is subject in the normal course of business to various legal proceedings in which
claims for monetary or other damages may be asserted. Management does not believe such legal
proceedings, unless otherwise disclosed herein, are material.
10. EMPLOYEE RETIREMENT PLAN
The Company sponsors the EntreMed, Inc. 401(k) and Trust. The plan covers substantially all
employees and enables participants to contribute a portion of salary and wages on a tax-deferred
basis. Contributions to the plan by the Company are discretionary. Contributions by the Company
totaled $77,000, $87,000 and $89,000 in 2005, 2004 and 2003, respectively.
11. SUBSEQUENT EVENTS
In January 2006 the Company acquired Miikana Therapeutics, a private biotechnology company.
Pursuant to the Merger Agreement EntreMed acquired all of the outstanding capital stock of Miikana
Therapeutics, Inc. in exchange for 9.96 million shares of EntreMed common stock valued at
approximately $21.2 million, based on the closing price of EntreMeds common stock the day before
the merger was announced. In addition, based on the success of the acquired pre-clinical programs,
EntreMed may pay up to an additional $18 million upon the achievement of certain clinical and
regulatory milestones. Such additional payments will be made in cash or shares of stock at
EntreMeds option.
F-19
In February 2006 the Company completed the private placement of units consisting of shares of
common stock and warrants at a purchase price of $2.3125 per unit with institutional healthcare
investors. In connection with the placement, the Company issued approximately 13 million shares of
common stock and warrants to purchase up to 6.5 million additional shares of common stock at an
exercise price of $2.50 per share. The warrants will not become exercisable until six months after
the closing. In conjunction with the sale we received gross proceeds of approximately $30 million,
net proceeds of approximately $28 million.
12. QUARTERLY FINANCIAL INFORMATION (UNAUDITED)
Summarized unaudited quarterly financial information for the years ended December 31, 2005 and
2004 is as follows:
QUARTER ENDED | ||||||||||||||||
MARCH 31, | JUNE 30, | SEPTEMBER 30, | DECEMBER 31, | |||||||||||||
2005 |
||||||||||||||||
Revenues |
$ | 25,249 | $ | 579,461 | $ | 1,249,600 | $ | 4,063,745 | ||||||||
Research and development costs |
4,379,356 | 3,812,353 | 4,128,756 | 5,004,583 | ||||||||||||
General and administrative expenses |
1,260,822 | 1,339,150 | 1,786,868 | 1,533,615 | ||||||||||||
Investment income |
158,461 | 271,832 | 284,195 | 296,283 | ||||||||||||
Gain on sale of assets |
2,000 | 1,420 | ||||||||||||||
Net loss |
(5,456,468 | ) | (4,300,210 | ) | (4,379,829 | ) | (2,176,750 | ) | ||||||||
Dividends on Series A convertible
preferred stock |
(251,250 | ) | (251,250 | ) | (251,250 | ) | (251,250 | ) | ||||||||
Net loss attributable to common
Shareholders |
(5,707,718 | ) | (4,551,460 | ) | (4,631,079 | ) | (2,428,000 | ) | ||||||||
Net loss per share (basic and diluted) |
$ | (0.13 | ) | $ | (0.09 | ) | $ | (0.09 | ) | $ | (0.05 | ) | ||||
2004 |
||||||||||||||||
Revenues |
$ | 97,967 | $ | 139,380 | $ | 142,738 | $ | 133,910 | ||||||||
Research and development costs |
3,056,970 | 2,484,426 | 2,553,494 | 2,428,362 | ||||||||||||
General and administrative expenses |
2,092,939 | 1,963,410 | 1,337,922 | 1,176,393 | ||||||||||||
Investment income |
84,594 | 60,733 | 75,495 | 93,118 | ||||||||||||
Gain on sale-royalty interest |
3,000,000 | |||||||||||||||
Gain on sale of securities |
520,000 | |||||||||||||||
Gain on sale of assets |
6,335 | 117,748 | ||||||||||||||
Net loss |
(4,967,348 | ) | (4,247,723 | ) | (3,146,848 | ) | (259,979 | ) | ||||||||
Dividends on Series A convertible
preferred stock |
(251,250 | ) | (251,250 | ) | (251,250 | ) | (251,250 | ) | ||||||||
Net loss attributable to common
Shareholders |
(5,218,598 | ) | (4,498,973 | ) | (3,398,098 | ) | (511,229 | ) | ||||||||
Net loss per share (basic and diluted) |
$ | (0.14 | ) | $ | (0.12 | ) | $ | (0.09 | ) | $ | (0.02 | ) |
F-20