Statera Biopharma, Inc. - Annual Report: 2007 (Form 10-K)
United
States Securities and Exchange Commission
WASHINGTON,
D.C. 20549
FORM
10-K
(Mark
One)
x Annual
Report Pursuant to Section 13 or 15(d) of the Securities Exchange Act of 1934
For
the fiscal year ended December 31, 2007
or
¨ Transition
Report pursuant to Section 13 or 15(d) of the Securities Exchange Act of
1934
For
the transition period from ___________to ___________
Commission
file number 001-32954
CLEVELAND
BIOLABS, INC.
(Exact
name of registrant as specified in its charter)
DELAWARE
|
20-0077155
|
|
(State
or other jurisdiction of incorporation
or
organization)
|
(I.R.S.
Employer Identification No.)
|
73
High Street, Buffalo, NY 14203
|
(716)
849-6810
|
|
(Address
of principal executive offices)
|
Telephone
No.
|
Securities
Registered Pursuant to Section 12(b) of the Act:
Title
of each class
|
Name
of each exchange which registered
|
|
Common
Stock, par value $0.005 per share
|
NASDAQ
Global Market
|
Securities
Registered Pursuant to Section 12(g) of the Act:
None
Indicate
by check mark if the registrant is a well-known seasoned issuer, as defined
in
Rule 405 of the Securities Act. Yes ¨
No x
Indicate
by check mark if the registrant is not required to file reports pursuant to
Section 13 or Section 15(d) of the Act. Yes ¨
No x
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 x
No ¨
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 registrant’s 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. x
Indicate
by check mark whether the registrant is a large accelerated filer, an
accelerated filer, a non-accelerated filer, or a smaller reporting company.
See
definition of “large accelerated filer,” “accelerated filer” and “smaller
reporting company” in Rule 12b-2 of the Exchange Act. (Check one):
Large
accelerated filer o
|
Accelerated
filer o
|
Non-accelerated
filer o
|
Smaller
reporting company x
|
Indicate
by check mark whether the registrant is a shell company (as defined in Rule
12b-2 of the Act). Yes ¨
No x
The
aggregate market value of the voting and non-voting common equity held by
non-affiliates, computed by reference to the price at which the common equity
was last sold or the average bid and asked price of such common equity, as
of
the last business day of the registrant’s most recently completed second fiscal
quarter was $74,961,490. There were 13,158,477 shares of common stock
outstanding as of March 1, 2008.
DOCUMENTS
INCORPORATED BY REFERENCE
The
definitive proxy statement relating to the registrant’s Annual Meeting of
Stockholders, to be held on April 29, 2008, is incorporated by reference in
Part
III to the extent described therein.
CLEVELAND
BIOLABS, INC.
FORM
10-K
03/21/08
Cleveland
BioLabs, Inc.
Form
10-K
For
the Fiscal Year Ended December 31, 2007
INDEX
Page
|
||
PART
I
|
||
Item
1
|
Description
of Business
|
2
|
Item
2
|
Description
of Property
|
21
|
Item
3
|
Legal
Proceedings
|
21
|
Item
4
|
Submission
of Matters to a Vote of Security Holders
|
21
|
PART
II
|
||
Item
5
|
Market
for Registrant’s Common Equity, Related Stockholder Matters and Issuer
Purchases of Equity Securities
|
22
|
Item
6
|
Selected
Financial Data
|
22
|
Item
7
|
Management’s
Discussion and Analysis of Financial Condition and Results of
Operations
|
22
|
Item
8
|
Financial
Statements and Supplementary Data
|
32
|
Item
9
|
Changes
in and Disagreements with Accountants on Accounting and Financial
Disclosures
|
33
|
Item
9A
|
Controls
and Procedures
|
33
|
Item
9B
|
Other
Information
|
33
|
PART
III
|
||
Item
10
|
Directors,
Executive Officers and Corporate Governance
|
34
|
Item
11
|
Executive
Compensation
|
34
|
Item
12
|
Security
Ownership of Certain Beneficial Owners and Management and Related
Stockholder Matters
|
34
|
Item
13
|
Certain
Relationships and Related Transactions, and Director
Independence
|
34
|
Item
14
|
Principal
Accountant Fees and Services
|
34
|
PART
IV
|
||
Item
15
|
Exhibits
and Financial Statement Schedules
|
34
|
SIGNATURES
|
37
|
1
FORWARD-LOOKING
STATEMENTS
This
Annual Report on Form 10-K contains forward-looking statements that involve
risks and uncertainties. In particular, statements about our expectations,
beliefs, plans, objectives, assumptions or future events or performance are
contained or incorporated by reference in this report. We have based these
forward-looking statements on our current expectations about future events.
While we believe these expectations are reasonable, such forward-looking
statements are inherently subject to risks and uncertainties, many of which
are
beyond our control. The actual future results for Cleveland BioLabs, Inc. may
differ materially from those discussed here for various reasons. Given these
risks and uncertainties, you are cautioned not to place undue reliance on such
forward-looking statements. The forward-looking statements included in this
report are made only as of the date hereof. We do not undertake and specifically
decline any obligation to update any such statements or to publicly announce
the
results of any revisions to any of such statements to reflect future events
or
developments. When used in the report, unless otherwise indicated, “CBLI,” “we,”
“our” and “us” refers to Cleveland BioLabs, Inc.
PART
I
Item 1.
Description
of Business
GENERAL OVERVIEW
CBLI
was
incorporated in Delaware and commenced business operations in June 2003 as
a
development-stage, biotechnology company, with a very specific and targeted
focus on radiation drug discovery. We have devoted substantially all of our
resources to the identification, development and commercialization of new types
of drugs for protection of normal tissues from exposure to radiation and other
stresses, such as toxic chemicals and cancer treatments. CBLI’s
pipeline includes products from two primary families of compounds: protectans
and curaxins. We are developing protectans as drug candidates that protect
healthy tissues from acute stresses such as radiation, chemotherapy and ischemia
(pathologies
developed as a result of blocking blood flow to a part of the body).
Curaxins are being developed as anticancer agents that could act as mono-therapy
drugs or in combination with other existing anticancer agents.
On
July
20, 2006, we sold 1,700,000 shares of common stock, par value $0.005 per share,
in our initial public offering at a per share price of $6.00. After our initial
public offering, our common stock was listed on the NASDAQ Capital Market under
the symbol “CBLI” and on the Boston Stock Exchange under the symbol “CFB.” Our
trading symbol on the Boston Stock Exchange was later changed to “CBLI.” On
August 28, 2007, trading of our common stock transferred from the NASDAQ Capital
Market to the NASDAQ Global Market. In September 2007, we ceased our listing
on
the Boston Stock Exchange.
TECHNOLOGY
Our
development efforts are based on discoveries made in connection with the
investigation of the cell-level process known as apoptosis. Apoptosis is a
highly specific and tightly regulated form of cell death that can occur in
response to external events such as exposure to radiation, toxic chemicals
or
internal stresses. Apoptosis is a major determinant of tissue damage caused
by a
variety of medical conditions including cerebral stroke, heart attack and acute
renal failure. Conversely, apoptosis is also an important protective mechanism
that allows the body to shed itself of defective cells, which otherwise can
cause cancerous growth.
Research
has demonstrated that apoptosis is sometimes suppressed naturally. For example,
most cancer cells develop resistance to apoptotic death caused by drugs or
natural defenses of the human body. Our research is geared towards identifying
the means by which apoptosis can be affected and manipulated depending on the
need.
2
If
the
need is to protect healthy tissues against an external event such as exposure
to
radiation, we focus our research efforts on attempting to temporarily and
reversibly suppress apoptosis in those healthy tissues, thereby imitating the
apoptotic-resistant tendencies displayed by cancer cells. A drug with this
effect would also be useful in ameliorating the often severe side effects of
anticancer drugs and radiation that cause collateral damage to healthy tissues
during cancer treatment. Because the severe side effects of anticancer drugs
and
radiation often limit their dosage in cancer patients, an apoptosis suppressant
drug may enable a more aggressive treatment regimen using anticancer drugs
and
radiation and thereby increase their effectiveness.
On
the
other hand, if the need is to destroy cancerous cells, we focus our research
efforts on restoring apoptotic mechanisms that are suppressed in tumors, so
that
those cancerous cells will once again become vulnerable to apoptotic death.
In
this regard, we believe that our drug candidates could have significant
potential for improving, and becoming vital to, the treatment of cancer
patients.
Through
our research and development, or R&D, and our strategic partnerships, we
have established a technological foundation for the development of new
pharmaceuticals and their rapid preclinical evaluation. We spent $17,429,652
and
$6,989,804 on R&D in the fiscal years ended December 31, 2007 and December
31, 2006, respectively.
We
have
acquired rights to develop and commercialize the following prospective
drugs:
·
|
Protectans
- modified factors of microbes and tumors that protect cells from
apoptosis, and which therefore have a broad spectrum of potential
applications. The potential applications include both non-medical
applications such as protection from exposure to radiation, whether
as a
result of military or terrorist action or as a result of a nuclear
accident, as well as medical applications such as reducing cancer
treatment side effects.
|
·
|
Curaxins
- small molecules designed to kill tumor cells by simultaneously
targeting
two regulators of apoptosis. Initial test results indicate that curaxins
can be effective against a number of malignancies, including renal
cell
carcinoma, or RCC (a highly fatal form of kidney cancer), soft-tissue
sarcoma, and hormone-refractory prostate
cancer.
|
In
the
area of radiation protection, we have achieved high levels of protection in
animal models. With respect to cancer treatment, the biology of cancer is such
that there is no single drug that can be successfully used to treat 100% or
even
50% of all cancer patients. This means that there likely will be a need for
additional anticancer drugs for each type of cancer.
These
drug candidates demonstrate the value of our scientific foundation. Based on
the
expedited approval process currently available for non-medical applications
such
as protection from exposure to radiation, our most advanced drug candidate,
Protectan CBLB502, may be approved for such applications within 24 to 36 months.
Another drug candidate, Curaxin CBLC102, entered Phase IIa clinical trials
earlier this year.
INDUSTRY
CBLI
is a
biotechnology, or biotech, company focused on developing cancer treatment,
tissue protection and biodefense drugs. Historically, biotech was defined by
newly discovered “genetic engineering” technology, which was first developed in
universities and new startup biotech companies in the mid-1970s. Later, other
technologies (based on a constant flow of discoveries in the field of biology)
started playing a leading role in biotech development. Medicine, and
specifically drug development, is a lucrative field for use of these
technologies. Large pharmaceutical, or Pharma, companies joined the biotech
arena through licensing, sponsored research, and corporate agreement
relationships. Today biotech is a $300 billion industry (based on total
market capitalization) and includes large companies such as Amgen, Inc. and
Genentech, Inc.
3
The
traditional biotech business model is a derivative of the long drug development
process. Typical biotech companies go through the following stages:
·
|
During
the first stage, biotech companies fund their development through
equity
or debt financings while conducting R&D, which culminates in phased
drug trials.
|
·
|
During
the second stage, when their lead drug candidates enter the drug
trials,
biotech companies may start licensing their drug candidates to Pharma
companies in order to (1) generate revenue, (2) gain access to additional
expertise, and (3) establish relations with Pharma companies in the
market
who can eventually take a leading role in distributing successful
drugs.
|
·
|
At
the most advanced stage, biotech companies generate revenues by selling
drugs or other biotech products to consumers or through alliances
of
equals.
|
The
Project BioShield Act, which was signed into law in July 2004, allocated
$5.6 billion over ten years to fund the research, development and
procurement of drugs, biological products or devices to treat or prevent injury
from exposure to biological, chemical, radiological or nuclear agents as a
result of a military, terrorist or nuclear attack. The legislation provides
for
a more expedited approval process by allowing for approval based on Phase I
safety studies in humans and efficacy studies in two animal species (rodents
and
non-human primates) instead of Phase II and III human clinical trials. With
the
Project BioShield Act, biotech companies now have greater access to grants
and
contracts with the U.S. government. Several biotech companies have secured
grants and contracts from the U.S. government to develop drugs and vaccines
as
medical countermeasures against potential terrorist attacks. For biotech
companies focused on these types of drugs and vaccines, this type of funding,
together with the scaled down Food and Drug Administration, or FDA, approval
process, are major departures from the traditional biotech business model.
The
principal provisions of this law are to:
·
|
Facilitate
R&D efforts of biomedical countermeasures by the NIH;
|
·
|
Provide
for the procurement of needed countermeasures through a special reserve
fund of $5.6 billion over ten years; and
|
·
|
Authorize,
under limited circumstances, the emergency use of medical products
that
have not been approved by the FDA.
|
While
there are a number of biotech and Pharma companies that are attempting to
develop new anti-radiation and anti-cancer drugs to treat these medical
conditions, these areas are nevertheless considered unmet medical needs, which
means that there are currently no existing methods to satisfactorily treat
these
medical conditions.
STRATEGIES
AND OBJECTIVES
Our
primary objective is to become a leading developer of drugs for the protection
of human tissues against radiation and other stresses and for cancer treatment.
Key elements of our strategy include:
·
|
Aggressively
working towards the commercialization of Protectan
CBLB502.
Our most advanced drug candidate, Protectan CBLB502, offers the potential
to protect normal tissues against exposure to radiation. Because
of the
potential military and defense implications of such a drug, the normally
lengthy FDA approval process for these non-medical applications is
substantially abbreviated resulting in a large cost savings to us.
We
anticipate having a developed drug available for these non-medical
applications within 18-30 months. The FDA approval process is estimated
to
take an additional six months.
|
4
·
|
Leveraging
our relationship with leading research and clinical development
institutions.
The Cleveland Clinic Foundation, one of the top research medical
facilities in the world, is one of our co-founders. In addition to
providing us with drug leads and technologies, the Cleveland Clinic
will
share valuable expertise with us as clinical trials are performed on
our drug candidates. In January 2007, we entered into a strategic
research
partnership with Roswell Park Cancer Institute, or RPCI, in Buffalo,
New
York. This partnership will enhance the speed and efficiency of our
clinical research and provide us with access to the state-of-the-art
clinical development facilities of a globally recognized cancer research
center.
|
·
|
Utilizing
governmental initiatives to target our markets.
Our focus on drug candidates such as Protectan CBLB502, which has
applications that have been deemed useful for military and defense
purposes, provides us with a built-in market for our drug candidates.
This
enables us to invest less in costly retail and marketing resources.
In an
effort to improve our responsiveness to military and defense needs,
we
have established a collaborative relationship with the Armed Forces
Radiobiology Research Institute.
|
·
|
Utilizing
other strategic relationships.
We
have collaborative relationships with other leading organizations
that
enhance our drug development and marketing efforts. For example,
one of
our founders, with whom we maintain a strategic partnership, is ChemBridge
Corporation. Known for its medicinal chemistry expertise and synthetic
capabilities, ChemBridge provides valuable resources to our drug
development research.
|
PRODUCTS
IN DEVELOPMENT
Protectans
We
are
exploring a new natural source of factors that suppress the programmed cell
death (apoptosis) response in human cells, which can be rapidly developed into
therapeutic products. These inhibitors are anti-apoptotic factors developed
by
microorganisms of human microflora throughout millions of years of co-evolution
with mammalian host. We are using the same strategy that was applied for
the discovery of antibiotics, one of the biggest medical achievements of the
20th
century. We have established a technological pipeline for screening of
such factors, named protectans, and their rapid preclinical
evaluation.
Such
inhibitors can be used as protection from cancer treatment side effects and
antidotes against injuries induced by radiation and other stresses associated
with severe pathologies (i.e., heart attack or stroke).
Protectan
CBLB502
Protectan
CBLB502 is our leading radioprotectant molecule in the protectans series.
Protectan CBLB502 represents a rationally-designed derivative of the microbial
protein, flagellin.
Flagellin is secreted by Salmonella
typhimurium and
many
other Gram-negative bacteria, and in nature, arranges itself in a hollow
cylinder to form the filament in bacterial flagellum and
acts
as a natural activator of NF-kB (nuclear factor-kappa B), a protein complex
widely used by cells as a regulator of genes that control cell proliferation
and
cell survival. Thus,
Protectan CBLB502 reduces
injury from acute stresses by mobilizing several natural cell protecting
mechanisms, including inhibition of apoptosis, reduction of oxidative damage
and
induction of factors (cytokines) that induce protection and regeneration of
stem
cells in bone marrow and intestine.
Potential
applications for Protectan CBLB502 include reduction of radiation therapy or
chemotherapy side effects in cancer patients, protection
from Acute Radiation Syndrome (ARS) in defense scenarios, and protection from
acute organ failure.
Protectan
CBLB502 is administered through intramuscular injection.
5
Biodefense
Applications
Our
scientists have demonstrated that injecting Protectan CBLB502 into mice, rats
and non-human primates protects them from lethal doses of total body gamma
radiation. An important advantage of Protectan CBLB502, above any other
radioprotectant known to us, is the ability to effectively protect not only
the
hematopoietic system, but also the gastrointestinal, or GI, tract, which are
among the most sensitive areas of the human body to radiation. High levels
of
radiation, among other effects, induce moderate to severe bone marrow damage.
The immune and blood stem cells are also depleted and death is caused by anemia,
infection, bleeding and poor wound healing. Protectan CBLB502’s ability to
effectively protect the hematopoietic system and GI tract may make Protectan
CBLB502 uniquely useful as a radioprotective antidote. Protectan CBLB502 was
shown to be safe at
its
therapeutic doses in rodents and non-human primates.
In
addition, Protectan CBLB502 has proved to be a stable compound for storage
purposes. It can be stored at temperatures close to freezing, room temperature
or extreme heat. Manufacture of Protectan CBLB502 is relatively inexpensive,
due
to its high yield bacterial producing strain and simple purification
process.
Our
research has also demonstrated that a single injection of less than 1% of the
maximum tolerable dose of Protectan CBLB502 protected greater than 80% of
National Institutes of Health, or NIH, Swiss mice from exposure to as high
as 13
Gy of total body irradiation. No other known compounds in development show
this
degree of protective effect from this level of radiation exposure.
Protectan
CBLB502 also showed strong radioprotective efficacy as a single therapy in
non-human primates, enabling the survival of 70% of the CBLB502-treated animals
that received whole-body radiation versus the non-treated control group, in
which 75% of the animals died. Of the non-human primates in the control group
that survived, none were without significant abnormalities. In contrast, the
surviving non-human primates treated with CBLB502 possessed no significant
structural abnormalities in their bone marrow, immune system organs, or small
intestines after 40 days. This is consistent with data previously obtained
from
trials on mice. Irradiated mice treated with CBLB502 survived to their normal
life span without developing any significant abnormalities and while preserving
the normal formation of blood cells (hematopoiesis). This data suggests that
CBLB502 may offer true protection from gamma-irradiation induced ARS, including
the lethal effects on both the GI and hematopoietic systems.
A
study
completed in late 2007 demonstrated the efficacy of Protectan CBLB502 as a
mitigator of hematopoietic (bone marrow/blood production) damage up to 48 hours
after radiation exposure. This was the first primate study pointing towards
CBLB502's high utility in protection of civil populations, where countermeasures
would be stockpiled and then distributed.
In
the
study, five groups of ten rhesus primates received 5 Gy (approximately 20%
of
lethal dose) of gamma radiation. The control group received a placebo, while
the
four experimental groups received a single intramuscular injection of Protectan
CBLB502 at one of the following times: 1, 16, 24 or 48 hours after irradiation.
No mortality was observed in CBLB502-treated groups after 30 days, while 20%
mortality was observed in the control group. Thrombocytopenia has been shown
to
be the best predictor of primate post-irradiation mortality in recent studies.
The
duration and occurrence of severe thrombocytopenia (a decrease of
platelets,
the
blood cells that prevent bleeding) was strongly reduced by CBLB502. The average
number of severe thrombocytopenia (< 50,000 platelets/ul) days per primate
was drastically reduced from 4.3 in the control group to 0.6-1.5 in all four
CBLB502-treated groups.
In
addition, duration and occurrence of severe neutropenia (a decrease in white
blood cells, which serve as the primary defense against infections) was also
reduced by CBLB502. For example, an average number of days of extremely severe
neutropenia (< 100 neutrophils/ul) per primate was reduced from 2.7 in the
control group to 0.3-1.5 in the experimental groups.
We
submitted Protectan CBLB502 in response to a Request for Information, or RFI,
from the Department of Health and Human Services, or HHS, in July 2007, which
noted the agency's intention to pursue an initial acquisition of 100,000
treatment courses of a medical countermeasure for neutropenia arising as a
consequence of ARS. The RFI further stated that there would be options for
up to
an additional 100,000 treatment courses to meet HHS’s requirement of at least
200,000 treatment courses.
6
We
intend
to initiate a human safety study in the first half of 2008 for Protectan CBLB502
in ARS, which is the only stage of human testing required for approval in this
indication.
Regulatory
Status
Extraordinary
radioprotective properties, an excellent toxicity profile, outstanding stability
and inexpensive production of Protectan CBLB502 make it a primary candidate
for
entering formal preclinical and clinical studies. Initially, Protectan CBLB502
will be developed for non-medical purposes — as a radioprotectant antidote for
the protection of people from severe doses of ionizing radiation. Our drug
development strategy complies with the recently adopted FDA rules for
investigational drugs that address situations such as radiation injury, where
it
would be unethical to conduct efficacy studies in humans. While Phase II and
Phase III human clinical trials are normally required for the approval of
marketing an investigational drug, under the FDA rules, Protectan CBLB502 would
be considered for approval for this indication based on Phase I safety studies
in humans and efficacy studies in two animal species (rodents and non-human
primates). Based upon this expedited approval process, Protectan CBLB502 could
be approved for non-medical applications within 24 to 36 months. Because Phase
II and Phase III testing involves applying a drug candidate to a large numbers
of participants who suffer from the targeted disease and condition and can
last
for a total of anywhere from three to six or additional years, bypassing these
phases represents a significant time and cost savings in receiving FDA approval.
As
part
of this expedited approval process, the FDA has indicated that it intends to
engage in a highly interactive review of Investigational New Drug, or IND,
applications and New Drug Applications, or NDAs, and to provide for accelerated
review or approval of certain medical products for counterterrorism
applications, including granting eligible applications “Fast Track” approval
status. The Fast Track designation ordinarily allows a product to be considered
for accelerated approval through the use of surrogate endpoints to demonstrate
effectiveness. As a result of these provisions, the FDA has broader authority
to
consider evidence of partial tumor shrinkage or other surrogate endpoints of
clinical benefit in deciding on approval. This new policy is intended to
facilitate the study of cancer therapies and shorten the total time required
for
marketing approvals. In cases where priority review is given to Fast Track
applications, the applicant is permitted to submit applications on a rolling
basis.
As
part
of the process to receive final FDA approval for Protectan CBLB502 for
non-medical applications, we have completed
Good Manufacturing Practices compliant (cGMP) manufacturing of Protectan
CBLB502. The
yields from the process and the purity of the final product exceeded our
expectations. We were able to develop a complicated, high-yield manufacturing
process for CBLB502 because of the excellent work of our in-house team and
consultants, and our subcontractor, SynCo Bio Partners B.V, which was able
to
prototype the process and resolve multiple challenges during the industrial
development. We currently have drug substance corresponding to over 100,000
projected human doses, or potentially many more, depending on the final
therapeutic dose to be used, which will be determined in the coming months
through our Phase I safety trial. The process we developed gives us the ability
to manufacture up to five million estimated doses within a year without any
additional scale-up; and if necessary, scale-up could be implemented relatively
easily.
In
order
for us to receive final FDA approval for Protectan CBLB502 for non-medical
applications, we need to:
·
|
Submit
an IND application and receive approval from the
FDA;
|
·
|
Perform
a Phase I dose-escalation human study on a small number of
volunteers;
|
·
|
Conduct
pivotal animal efficacy studies with the GMP manufactured drug
candidate;
|
·
|
Perform
a human safety study in a larger number of volunteers using the dose
of
CBLB502 previously shown to be safe in humans and efficacious in
animals;
and
|
·
|
File
a Biologic License Application, or
BLA.
|
7
In
our
most optimistic business scenario, all of these steps could be accomplished
in
18 months. In a more conservative business scenario, it may take up to 30 months
or more to complete the development and file the BLA for the approval of
Protectan CBLB502 for non-medical applications.
The
Project BioShield Act of 2004, which further expedites the approval of drug
candidates for certain uses, is intended to bolster our nation’s ability to
provide protections and countermeasures against biological, chemical,
radiological or nuclear agents that may be used in a military, terrorist or
nuclear attack.
This
law
also allows for the use of expedited peer review when assessing the merit of
grants and contracts of up to $1,500,000 for countermeasure research. We have
been awarded a $1,500,000 research grant pursuant to this law.
The
Defense Threat Reduction Agency of the U.S. Department of Defense, or DoD,
awarded us a $1.3 million grant in March 2007, to fund “development leading to
the acquisition” of Protectan CBLB502 as a radiation countermeasure, in
collaboration with the Armed Forces Radiobiology Research Institute, which
has
also received significant independent funding for work on Protectan CBLB502.
The
DoD
also recently awarded a $1 million grant to our founding partner, the Cleveland
Clinic, to conduct pre-clinical studies on Protectan CBLB502 for use in
tourniquet and other ligation-reperfusion battlefield injuries where blood
flow
is stopped and then restored after a prolonged period of time.
Market
Opportunities
Protectan
CBLB502 is a candidate for procurement by the DoD. In general, the procurement
process is conducted on the basis of full and open competition that cannot
be
limited, unless the DoD determines that the public requesting policy would
otherwise seriously jeopardize national security.
Prior
to
determining the best treatment, the DoD issues a Request for Information, or
RFI, for treatments available or in development for a specific condition
resulting from an identified threat. The RFI provides an incentive for companies
to research and develop countermeasures that are superior to those selected
for
stockpiling. Through the RFI, companies may compete for future contracts that
will revise and update stockpile content for emerging threats and to discover
advanced technologies and new countermeasures.
Following
its review of the responses it receives, the DoD issues a Request for Proposal,
or RFP. The RFP solicits proposals for the manufacturing of specified treatments
for a defined number of doses to be delivered within a specified time frame
(a
maximum of eight years). A contract may be awarded once the review of the RFP
responses has been completed, though payments by the government are made only
upon product delivery.
If
the
product or the use indicated in the RFP of an approved product is not approved,
licensed, or cleared for commercial distribution at completion of the review,
the DoD has the authority to procure the required amount if it has:
·
|
Determined
that sufficient and satisfactory clinical experience or research
data
(including data, if available, from pre-clinical and clinical trials)
support a reasonable conclusion that the countermeasure will qualify
for
approval or licensing within eight years after the date of a
determination, and
|
·
|
Determined
that the product is authorized for emergency use.
|
8
In
February 2007, the DoD, through the U.S. Army Space and Missile Defense Command,
issued a RFP for the Advanced Development of Medical Radiation Countermeasures,
or MRC. According to the RFP, the objective of the MRC project is to develop
a
post-exposure MRC through a Phase I clinical trial and, pending successful
completion of the Phase I clinical trial, develop the MRC product through
approval/licensure with the FDA and procure quantities of the MRC sufficient
to
achieve Initial Operational Capability, or IOC. The RFP stated that the MRC
must
have the following characteristics: be safe, efficacious, quick acting, free
from performance-decrementing side effects, relatively non-invasive, compatible
with current military countermeasures, and usable on the battle field. The
MRC
also should not require refrigeration, nor have other significant logistical
burdens, and should have a relatively long shelf life. The solicitation
specifically requested a drug/biologic intended for use after exposure to
ionized radiation, or IR, has occurred.
In
January 2008, we learned that Protectan CBLB502 was not selected for award
under
the RFP. We
intend
to further develop CBLB502 and obtain FDA approval and will respond to future
DoD solicitations as they are announced. We plan to continue our discussions
with the DoD, HHS, and other friendly governments, who are interested in
CBLB502’s potential to protect against terrorist threats and nuclear disaster.
Our goal is to achieve FDA approval for CBLB502 in 2009 and market it as an
effective and affordable radiation protector for defense use on the battlefield
or for first responders of civilian emergencies.
Medical
Applications
In
addition to its military or other non-medical applications, we have found that
Protectan CBLB502 has been observed to dramatically increase the efficacy of
radiotherapy of experimental tumors in mice. Protectan CBLB502 appears to
increase the tolerance of mice to radiation while having no effect on the
radiosensitivity of tumors, thus opening the possibility of combining
radiotherapy with Protectan CBLB502 treatment to improve the overall anticancer
efficacy of radiotherapy. Our animal efficacy studies have demonstrated that
up
to 100% of mice treated with Protectan CBLB502 prior to being exposed to
radiation survived without any associated signs of toxicity. This compares
to a
100% mortality rate in the animal group that received a placebo drug.
Another
recent study demonstrated the ability of Protectan CBLB502 to reduce the side
effects of a chemotherapeutic drug, Platinol (cisplatin), broadly used for
the
treatment of ovarian, endometrial, head and neck, lung, stomach and other types
of cancer. Platinol
treatment was used in the study as an example of chemotherapy-associated
toxicity. Platinol injected at toxic doses is known to induce myelosuppression
(suppression of bone marrow) and nephrotoxicity (kidney damage). The severity
of
these injuries in mice can be monitored by the degree of weight loss and, in
the
case of severe adverse effects, the proportion of fatalities in treated groups.
In
the
study, Protectan CBLB502 was injected 30 minutes before Platinol was
administered at a dose of 1 mcg/mouse (0.04 mg/kg, which is less than 1% of
maximal tolerable dose). Platinol was injected either at the maximal tolerable
dose or at double maximal tolerable dose. Mice were monitored daily for weight
and behavioral abnormalities for 30 days, or until death. Mice losing more
than
25% of their weight were sacrificed, per conventional ethical guidelines for
animal treatment.
In
mice
that received the maximal tolerable dose of Platinol, CBLB502-treated animals
showed neither weight loss nor behavioral signs of morbidity, both of which
were
seen in all of the control group mice.
In
mice
that received twice the maximal tolerable dose of Platinol, the majority of
mice
in the control group died or were sacrificed due to loss of more than 25% of
their weight by day 10, while all of the animals that received Protectan CBLB502
before Platinol survived, never reaching more than 20% weight loss.
Thus
in
both of these dose groups, a single injection of CBLB502 prior to treatment
with
Platinol strongly reduced the toxicity of the drug, as indicated by less severe
weight loss and lack of behavioral changes in treated mice.
9
The
prospect of increasing patients' tolerance to chemotherapeutic drugs and
optimizing treatment regimens would be a significant paradigm shift in cancer
treatment. It is estimated that approximately 40% of the roughly $50 billion
annually spent on cancer treatment represents supportive care addressing side
effects of various treatments, including chemotherapy.
We
plan
to initiate a Phase I/II study in the second half of 2008 for Protectan CBLB502
in head and neck cancer patients. The endpoint of the study will be the
reduction of side effects of radiation and chemotherapy, such as mucositis
(a
painful
inflammation and ulceration of oral mucosa causing difficulties with speaking
and eating). Mucositis
weakens
the patient by not allowing for the oral intake of nutrients and fluids and
forces the temporary suspension of radiotherapy and chemotherapy until the
tissues of the mouth and throat have healed. Due to the ability of head and
neck
cancer cells to regrow during periods of interrupted treatment, any interruption
in radiotherapy should be avoided. Since the main cause of treatment
interruptions in radiotherapy or combinations of chemotherapy and radiotherapy
treatment regimens of head and neck cancer is acute mucositis, the ability
to
prevent mucositis, and therefore, interruptions in treatment, could actually
result in better outcomes for patients with cancers of the head and
neck.
Protectan
CBLB502 has also shown efficacy as a potential adjuvant for radiation therapy
in
mouse models of sarcoma and our researchers, in collaboration with investigators
from Cleveland Clinic, have demonstrated that a single injection of Protectan
CBLB502 effectively prevents acute renal failure and subsequent death in a
mouse
model of ischemia-reperfusion renal injury.
In
contrast to the non-medical applications of CBLB502, the use of Protectan
CBLB502 to ameliorate the side effects of radiation treatment and anticancer
drugs will be subject to the full FDA approval process.
Protectan
CBLB612
Protectan
CBLB612 is a modified lipopeptide mycoplasma that acts as a powerful stimulator
and mobilizer of hematopoietic (bone marrow/blood production) stem cells, or
HSC, to peripheral blood.
Potential
applications for Protectan CBLB612 include accelerated hematopoietic recovery
during chemotherapy and during donor preparation for bone marrow
transplantation.
Our
research indicates that Protectan CBLB612 is not only a potent stimulator of
bone marrow stem cells, but also causes their mobilization and proliferation
throughout the blood. A single administration of Protectan CBLB612 resulted
in a
three-fold increase in the number of progenitor stem cells in mouse bone marrow
within 24 hours after administration. Furthermore, the number of these stem
cells in peripheral blood was increased ten-fold within four days of
administration.
Protectan
CBLB612 was also found to be highly efficacious in stimulating proliferation
and
mobilization of hematopoietic stem cells into peripheral blood in primate model
(Rhesus macaques). A single injection of Protectan CBLB612 in Rhesus
macaques resulted in a 20-fold increase of hematopoietic progenitor cells in
blood. At the peak of the effect (48-72 hours post-injection) the
proportion of free-floating CD34+ cells in the total white blood cell count
reached 30% (compared with 1.5% in normal blood). CD34 is a molecule
present
on certain cells within the human body. Cells expressing CD34, otherwise
known as CD34+ cells, are normally found in the umbilical
cord
and
bone
marrow
as
hematopoietic cells.
This
discovery opens a new and innovative way for us to address a broad spectrum
of
human diseases, some of which currently lack effective treatment. Direct
comparisons of Protectan CBLB612 and the market leading drug used for
stimulation of blood regeneration, G-CSF (Neupogen®, Amgen, Inc., Thousand Oaks,
California), demonstrated a stronger efficacy of Protectan CBLB612 as a
propagator and mobilizer of HSC in peripheral blood.
Protectan
CBLB612's strength as a stem cell stimulator was further demonstrated by the
outcome of its combined use with G-CSF and AMD3100 (a promising clinical-stage
stem cell mobilizer from Genzyme Corporation (Cambridge, Massachusetts)), where
the addition of Protectan CBLB612 resulted in eight to ten times higher yields
of HSC in peripheral blood in comparison with the standard
protocol.
In
this
study, a single injection of Protectan CBLB612 was given to mice in combination
with the current standard methodology for stem cell donor isolation, which
is
four daily injections of G-CSF followed by one injection of AMD3100. The
addition of Protectan CBLB612 to the protocol yielded eight to ten times higher
concentrations of short-term and long-term HSC in peripheral blood compared
to
the standard protocol. Even a single administration of Protectan CBLB612 in
combination with AMD3100 yielded twice as many mobilized HSC compared with
that
of the standard regimen. Furthermore, Protectan CBLB612 mobilized all major
classes of HSC to peripheral blood, suggesting that no HSC classes would be
lost
in the enrichment process.
10
A
report
published by the NIH division of HHS entitled "Regenerative Medicine 2006,"
notes that hematopoietic stem cells have been used clinically since 1959 and
are
used increasingly routinely for transplantations, albeit almost exclusively
in a
non-pure form. Currently, the main indications for bone marrow transplantation
are either hematopoietic cancers (leukemias and lymphomas), or the use of
high-dose chemotherapy for nonhematopoietic malignancies (cancers in other
organs). Other indications include diseases that involve genetic or acquired
bone marrow failure, such as aplastic anemia, thalassemia sickle cell anemia,
and increasingly, autoimmune diseases. Producing a ready supply of hematopoietic
stem cells for an individual, without painful procedures, risk of contamination,
or side effects, would be tantamount to enabling the body to repair itself
from
any damage to its blood-forming system.
In
addition to efficacy in stimulation and mobilization of stem cells, Protectan
CBLB612 was found to be highly effective in
an
animal bone marrow stem cell transplantation model.
Blood
from healthy mice treated by Protectan CBLB612 was transplanted into mice that
received a lethal dose of radiation that killed hematopoietic (bone marrow/blood
production) stem cells. A small amount of blood from the Protectan CBLB612
treated mice successfully rescued the mice with radiation-induced bone marrow
stem cell deficiency. 100% of the deficient mice transplanted with blood
from CBLB612
treated
mice survived past the 60-day mark, while 85% of the untreated deficient mice
died within the first three weeks of the experiment. The 60-day mark is
considered to be the critical point in defining the presence of long-term,
adult
bone marrow stem cells, which are capable of completely restoring lost or
injured bone marrow function. The rescuing effect of the peripheral blood
of the treated mice was equivalent to that of conventional bone marrow
transplantation.
Adult
hematological bone marrow stem cell transplantation is currently used for
hematological disorders (malignant and non-malignant), as well as some
non-hematological diseases, such as breast
cancer, testicular cancer, neuroblastoma, ovarian cancer, Severe Combined Immune
Deficiency (SCID), Wiskott-Aldrich syndrome, and Chediak-Higashi syndrome.
Protectan
CBLB612 also has been shown to provide protection in a mouse model from lethal
hematopoietic-induced ARS when administered between 48 hours prior or up to
24
hours after radiation exposure.
Protectan
CBLB612 does not display
any significant toxicity at its therapeutic doses in rodents and non-human
primates.
With
efficacy and non-GLP safety already studied in mice and monkeys, Protectan
CBLB612 entered formal pre-clinical safety and manufacturing development in
February 2008. Its first human trials are projected for 2009. The development
of
our Protectan CBLB612 has been supported by a grant from the Defense Advanced
Research Projects Agency of the Department of Defense.
Curaxins
Curaxins
are small molecules that destroy tumor cells by simultaneously targeting two
regulators of apoptosis. Our initial test results indicate that curaxins can
be
effective against a number of malignancies, including renal cell carcinoma,
or
RCC, soft-tissue sarcoma, and hormone-refractory prostate cancer.
11
The
original focus of our drug development program was to develop drugs to treat
one
of the most treatment-resistant types of cancer, RCC. Unlike many cancer types
that frequently mutate or delete p53, one of the major tumor suppressor genes,
RCC belongs to a rare category of cancers that typically maintain a wild type
form of this protein. Nevertheless, RCC cells are resistant to apoptosis,
suggesting that in spite of its normal structure, p53 is functionally disabled.
The work of our founders has shown that p53 function is indeed inhibited in
RCC
by an unknown dominant factor. We have established a drug discovery program
to
identify small molecules that selectively destroy tumor cells by restoring
the
normal function to functionally impaired p53 in RCC. This program yielded a
series of chemicals with the desirable properties named curaxins (CBLC100
series). We have isolated three chemical classes of curaxins. One of them
includes relatives of 9-aminoacridine, the compound that is the core structure
of many existing drugs. Pre-existing information about this compound has allowed
us to bypass the preclinical development and Phase I studies and bring one
of
our drug candidates into Phase IIa clinical trials, saving years of R&D
efforts and improving the probability of success.
One
of
the most important outcomes of this drug discovery program was the
identification of the mechanism by which curaxins deactivate NF-kB. This
mechanism of action makes curaxins potent inhibitors of the production and
the
activity of NF-kB not only in its stimulated form, but also in its basal form.
The level of active NF-kB is usually also increased in cancer cells. Moreover,
due to curaxin-dependent functional conversion of NF-kB-DNA complexes, the
cells
with the highest basal or induced NF-kB activity are supposed to be the most
significantly affected by curaxins. Clearly, this paradoxical activity makes
deactivation of NF-kB by curaxins more advantageous compared to conventional
strategies targeting NF-kB activators.
The
discovery of the mechanism of action of curaxins allowed us to predict and
later
experimentally verify that curaxins could be used for treatment of multiple
forms of cancers, including hormone-refractory prostate cancer, hepatocellular
carcinoma, multiple myeloma, acute lymphocytic leukemia, acute myeloid leukemia,
soft-tissue sarcomas and several others.
Curaxin
CBLC102
One
of
the curaxins from the 9-aminoacridine group is a long-known, anti-infective
compound known as quinacrine, which we refer to as Curaxin CBLC102. It has
been
used for over 40 years to treat malaria, osteoarthritis and autoimmune
disorders. However, we have discovered new mechanisms of action for quinacrine
in the area of apoptosis. Through assay testing performed at Dr. Andrei Gudkov’s
laboratories at the Cleveland Clinic beginning in 2002, which included testing
in a variety of human tumor-derived cell lines representing cancers of different
tissue origin (including RCC, sarcomas, prostate, breast and colon carcinomas),
we have observed that Curaxin CBLC102 behaves as a potent NF-kB suppressor
and
activator of p53 in these types of cancer cells. It has favorable
pharmacological and toxicological profiles and demonstrates the anticancer
effect in transplants of human cancer cells into primates. These features make
Curaxin CBLC102 our prime IND drug candidate among other curaxins.
We
have
applied for a patent covering the use of Curaxin CBLC102 as an anticancer agent
based on a newly-discovered mechanism of action.
We
have
an agreement with Regis Technologies, Inc., a GMP manufacturer, to produce
sufficient quantities of Curaxin CBLC102 according to the process previously
used for the production of this drug when it was in common use. On May 26,
2006,
we filed our IND application with the FDA to begin clinical trials in patients
with androgen-independent, prostate cancer. On June 26, 2006, the FDA advised
us
that we may initiate clinical Phase II studies after making minor modifications
to the protocol for such clinical studies.
A
Phase
II efficacy clinical trial using Curaxin CBLC102 in patients with advanced
hormone-refractory (androgen-independent) prostate cancer started in January
2007 at the University of Chicago, Cleveland Clinic, and Case Western Reserve
University Hospitals. We are applying CBLC102 as the monotherapy to patients
who
have failed to respond satisfactorily after undergoing established cancer
treatments and will use the suppression of tumor growth and prolonged patient
survival as major endpoints. Reducing the prostate-specific antigen, or
PSA, level is an additional endpoint (elevated PSA levels are indicative of
the
progression of prostate cancer). We expect to report the results of this trial
in mid-2008.
12
We
intend
to seek orphan drug status with respect to Curaxin CBLC102. The orphan drug
provisions of the Federal Food, Drug, and Cosmetic Act provide incentives to
drug and biologic manufacturers to develop and manufacture drugs for the
treatment of rare diseases, currently defined as diseases that exist in fewer
than 200,000 individuals in the U.S. or, for a disease that affects more than
200,000 individuals in the U.S., where the sponsor does not realistically
anticipate that its drug will become profitable. We believe that Curaxin CBLC102
may qualify as an orphan drug for purposes of treatment of RCC, soft tissue
sarcoma,
and
multiple myeloma - all diseases that affects
fewer than 200,000 individuals in the U.S.
Under
these provisions, a manufacturer of a designated orphan drug can seek tax
benefits, and the holder of the first designated orphan drug approved by the
FDA
will be granted a seven-year period of marketing exclusivity for that drug.
There is no assurance that we will receive orphan drug status for Curaxin
CBLC102. Even if we do receive orphan drug status, while the marketing
exclusivity of an orphan drug would prevent other sponsors from obtaining
approval of the same compound for the same indication, it would not prevent
other types of drugs from being approved for the same indication and therefore
may not provide sufficient protection against competitive products.
Other
Curaxins
As
mentioned above, screening of the chemical library for compounds capable of
restoring normal function to wild type p53 in the context of RCC yielded three
chemical classes of compounds. Generation of focused chemical libraries around
the hits from one of these classes and their structure-activity optimization
brought about a new generation of curaxins. As the part of this program
performed in the partnership with ChemBridge Corporation, more than 800
proprietary compounds were screened for p53 activation, efficacy in animal
tumor
models, selective toxicity and metabolic stability in the presence of rat and
human microsomes. The most active compounds were efficacious in preventing
tumor
growth in models for colon carcinoma, melanoma, ovarian cancer, RCC, and breast
cancer. In February 2008, three lead candidates were chosen for
preclinical development based on their efficacy, low toxicity profiles, high
stability and suitability for human administration.
COLLABORATIVE
RESEARCH AGREEMENTS
Cleveland
Clinic Foundation
We
have a
unique opportunity to accelerate our development by utilizing intellectual
property, drug leads, new research technologies, technical know-how and original
scientific concepts derived from 25 years of research achievements relevant
to
cancer by Dr. Gudkov and his research team while at the Cleveland Clinic.
Pursuant to an Exclusive License Agreement we entered into with the Cleveland
Clinic effective as of July 1, 2004, we were granted an exclusive license to
the
Cleveland Clinic’s research base underlying our therapeutic platform (the
CBLC100, CBLB500 and CBLB600 series). In consideration for obtaining this
exclusive license, we agreed to:
·
|
Issue
to the Cleveland Clinic 1,341,000 shares of common
stock;
|
·
|
Make
certain milestone payments (ranging from $50,000 to $4,000,000, depending
on the type of drug and the stage of such drug’s
development);
|
·
|
Make
royalty payments (calculated as a percentage of the net sales of
the drugs
ranging from 1-2%); and
|
·
|
Make
sublicense royalty payments (calculated as a percentage of the royalties
received from the sublicenses ranging from 5-35%).
|
13
The
schedule of milestone payments is as follows:
File
IND application for Protectan CBLB502
|
$
|
50,000
|
||
Complete
Phase I studies for Protectan CBLB502
|
$
|
100,000
|
||
File
NDA application for Protectan CBLB502
|
$
|
350,000
|
||
Receive
regulatory approval to sell Protectan CBLB502
|
$
|
1,000,000
|
||
File
IND application for Curaxin CBLC102 (completed May 2006)
|
$
|
50,000
|
||
Commence
Phase II clinical trials for Curaxin CBLC102 (completed January
2007)
|
$
|
250,000
|
||
Commence
Phase III clinical trials for Curaxin CBLC102
|
$
|
700,000
|
||
File
NDA application for Curaxin CBLC102
|
$
|
1,500,000
|
||
Receive
regulatory approval to sell Curaxin CBLC102
|
$
|
4,000,000
|
Under
this license agreement, we may exclusively license additional technologies
discovered by Dr. Gudkov in this field by providing the Cleveland Clinic with
notice within 60 days after receiving an invention disclosure report from the
Cleveland Clinic relating to any such additional technologies. We believe that
this relationship will prove valuable, not only for the purposes of developing
the discoveries of Dr. Gudkov and his colleagues, but also as a source of
additional new technologies. We also expect that the Cleveland Clinic will
play
a critical role in validating therapeutic concepts and in conducting trials.
The
Cleveland Clinic may terminate the license upon a material breach by us, as
specified in the agreement. However, we may avoid such termination if we cure
the breach within 90 days of receipt of a termination notice.
In
August
2004, we entered into a cooperative research and development agreement, or
CRADA, with (i) the Uniformed Services University of the Health Sciences, which
includes the Armed Forces Radiobiology Research Institute, or AFRRI, (ii) the
Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.,
and
(iii) the Cleveland Clinic, to evaluate one of our radioprotective drug
candidates and its effects on intracellular and extracellular signaling
pathways. As a collaborator under this agreement, we are able to use the
laboratories of the Armed Forces Radiobiology Research Institute to evaluate
Protectan CBLB502 and its effects on intracellular and extracellular signaling
pathways in order to improve countermeasures to lethal doses of radiation.
Under
the terms of the agreement, all parties are financially responsible for their
own expenses related to the agreement. The agreement has a five-year term,
but
may be unilaterally terminated by any party upon 30 days prior written notice
with or without cause.
In
February 2008, the terms of the agreement were extended by an additional two
years expiring August 15, 2010 and an additional scope of the research to be
performed under the CRADA has been added. As the part of the extended research
plan AFRRI will perform additional experiments in non-human primates to evaluate
radioprotection efficacy of Protectan CBLB502 and perform analysis of
hematopoietic stem cell mobilization by Protectan CBLB612.
Roswell
Park Cancer Institute
In
January 2007, we entered into a strategic research partnership with Roswell
Park
Cancer Institute, or RPCI, to develop our anticancer and radioprotectant drug
candidates.
RPCI,
founded in 1898, is a world-renowned cancer research hospital and the nation's
first cancer research, treatment and education center. RPCI is a member of
the
prestigious National Comprehensive Cancer Network, an alliance of the nation's
leading cancer centers, and is one of only ten free-standing cancer centers
in
the nation.
14
RPCI
and
various agencies of the state of New York will provide us with up to $5 million
of grant and other funding. We established a major research/clinical facility
at
the RPCI campus in Buffalo, New York, which has become the foundation for
several of our advanced research and clinical trials.
Our
partnership with RPCI will enhance the speed and efficiency of our clinical
research, and will provide us with access to state-of-the-art clinical
development facilities in partnership with a globally recognized cancer research
center. We believe that our proprietary technology, combined with the assistance
of RPCI, and our continuing strong relationship with the Cleveland Clinic,
will
position us to become a leading oncology company. A key element of our long-term
business strategy is to partner with world-class institutions to aid us in
accelerating our drug development timeline. We believe that our firm alliances
with both RPCI and the Cleveland Clinic provide us with a significant
competitive advantage.
ChemBridge
Corporation
Another
vital component of our drug development capabilities is our strategic
partnership with ChemBridge Corporation, an established leader in combinatorial
chemistry and in the manufacture of diverse chemical libraries.
On
April
27, 2004, we entered into a library access agreement with ChemBridge that,
in
exchange for shares of our common stock and warrants, provides us with continual
access to a chemical library of 214,000 compounds. Under the library access
agreement, we have also agreed to collaborate with ChemBridge in the future
on
two optimization projects, wherein ChemBridge will have the responsibility
of
providing the chemistry compounds for the project and we will have the
responsibility of providing the pharmacological/biological compounds. Upon
providing ChemBridge with our data after at least two positive repeat screening
assays, which have been confirmed in at least one additional functional assay,
ChemBridge will have the option to select such compound as one of the two
optimization projects. ChemBridge will retain a 50% ownership interest in two
lead compounds selected by ChemBridge and all derivative compounds thereof.
The
parties will jointly manage the development and commercialization of any
compounds arising from an optimization project. The parties are discussing
the
possibility of entering into an additional project arising from the optimization
project. There can be no assurance the parties will agree to proceed with such
project on favorable terms, or at all. The library access agreement does not
have a specified term or any termination provisions.
We
have a
strong working relationship with ChemBridge. This relationship has already
resulted in the isolation of bioactive small molecules with clinical potential
that helped to establish either new therapeutic concepts (p53 inhibitors) or
identify molecules for important indications acting through previously unknown
mechanisms (novel class of inhibitors of multidrug transporters). Both lines
of
study have resulted in high visibility publications and are slated for further
exploration by us.
PATENTS
As
a
result of the license agreement with the Cleveland Clinic, we have filed, on
the
Cleveland Clinic’s behalf, thirteen patent applications covering new classes of
anticancer and radiation-protecting compounds, their utility and mode of action.
Our
intellectual property platform is based primarily on these thirteen
patent
applications exclusively licensed to us by the Cleveland Clinic and three patent
applications, which we have filed and own exclusively.
The
aforementioned thirteen
patent
applications licensed from the Cleveland Clinic are as follows:
·
|
Methods
of Inhibiting Apoptosis Using Latent
TFGß
|
·
|
Methods
of Identifying Modulators of Apoptosis From Parasites and Uses
Thereof;
|
15
·
|
Methods
of Inhibiting Apoptosis Using Inducers of
NF-kB;
|
·
|
Methods
of Protecting Against Radiation Using Inducers of
NF-kB;
|
·
|
Methods
of Protecting Against Radiation Using
Flagellin;
|
·
|
Small
Molecules Inhibitors of MRP1 and Other Multidrug
Transporters;
|
·
|
Flagellin
Related Polypeptides and Uses
Thereof;
|
·
|
Modulation
of Apoptosis Using Aminoacridines;
|
·
|
Modulation
of Immune Responses;
|
·
|
Activation
of p53 and Inhibition of NF-kB for Cancer
Treatment;
|
·
|
Methods
of Protecting Against Apoptosis Using
Lipopeptides;
|
·
|
Modulation
of Cell Growth; and
|
·
|
Mitochondrial
Cytochrome B.
|
The
aforementioned three patent applications, which we filed, are as
follows:
·
|
Quinacrine
Isomers;
|
·
|
Modulation
of Androgen Receptor for Treatment of Prostate Cancer;
and
|
·
|
Method
of Increasing Hematopoietic Stem
Cells.
|
MANUFACTURING
We
do not
intend to establish or operate facilities to manufacture our drug candidates,
and therefore will be dependent upon third parties to do so. As we develop
new
products or increase sales of any existing product, we must establish and
maintain relationships with manufacturers to produce and package sufficient
supplies of our finished pharmaceutical products. We have
established a relationship with SynCo Bio Partners B.V., a leading
biopharmaceutical manufacturer, to produce Protectan CBLB502 under cGMP
specifications, and have completed an agreement to produce sufficient amounts
for clinical trials and a commercial launch. For CBLC102, we have contracted
with Regis Technologies, Inc. to manufacture sufficient amounts for clinical
trials.
Reliance
on third party manufacturing presents several risks, including the
following:
·
|
Delays
in the delivery of quantities needed for multiple clinical trials
or
failure to manufacture such quantities to our specifications, either
of
which could cause delays in clinical trials, regulatory submissions
or
commercialization of our drug
candidates;
|
·
|
Inability
to fulfill our needs in the event market demand for our drug candidates
suddenly increases, which may require us to seek new manufacturing
arrangements, which, in turn, could be expensive and time consuming;
and
|
·
|
Ongoing
inspections by the FDA or other regulators and other regulatory
authorities for compliance with rules, regulations and standards,
the
failure to comply with which may subject us to, among other things,
product seizures, recalls, fines, injunctions, suspensions or revocations
of marketing licenses, operating restrictions and criminal
prosecution.
|
16
COMPETITION
Non-Medical
Applications
In
the
area of radiation-protective antidotes, various companies, such as RxBio, Inc.,
Exponential Biotherapies Inc., Osiris Therapeutics, Inc., ImmuneRegen
BioSciences, Inc. and Humanetics Corporation are developing biopharmaceutical
products that potentially directly compete with our non-medical application
drug
candidates even though their approaches to such treatment are different.
We
believe that due to the global political environment, the level of development
advancement is the critical factor in the marketing of an effective medical
radiation countermeasure to federal agencies, such as DoD and HHS. New
developments in this area are expected to continue at a rapid pace in both
industry and academia. For these reasons, we believe that competition will
be
driven by the level of development advancement of MRC.
Anticancer
Applications
The
arsenal of medical radiation-protectors is limited to ETHYOL™ (amifostine), sold
by MedImmune, and recently acquired by AstraZeneca International. This
radiation-protector is limited because of the serious side effects of the drug.
Other radiation-protectors may enter the market.
Biomedical
research for anticancer therapies is a large industry, with many companies,
universities, research institutions and foreign government-sponsored companies
competing for market share. The top ten public U.S.-based companies involved
in
cancer therapy have a combined market capitalization exceeding $1 trillion.
In
addition, there are several hundred biotech companies who have as their mission
anticancer drug development. These companies account for the approximately
150
anticancer compounds currently in drug trials. However, despite the numerous
companies in this field, there is still a clear, unmet need in the anticancer
drug development market.
Each
of
the approximately 200 types of cancer recognized by the National Cancer
Institute, or NCI, has dozens of subtypes, both etiological and on a treatment
basis. Due to this market segmentation, the paradigm of a one-size-fits-all,
super-blockbuster approach to drug treatments does not work well in cancer
therapy. Currently, even the most advanced therapeutics on the market do not
provide substantial health benefits.
This
suggests that innovative anticancer therapies are driven by the modest success
of current therapeutics, the need for an improved understanding of the
underlying science, and a shift in the treatment paradigm towards more
personalized medicine. Our technology addresses this need for an improved
understanding of the underlying science and implements a fundamental shift
in
the approach to developing anticancer therapies.
Stem
Cell Mobilization
G-CSF
(Neupogen® and Neulast@, Amgen, Inc., Thousand Oaks, California) is the current
standard against which all other mobilization agents for stem cells are
measured. This is because it has been shown to both mobilize more CD34+ stem
cells and have less toxicity than any other single agent against which it has
been tested to date. Use of G-SCF caused deaths attributed to thrombosis (acute
myocardial infarction and stroke) in sibling donors. Other side effects include
pain, nausea, vomiting, diarrhea, insomnia, chills, fevers, and night
sweats.
Sargramostim
(Bayer HealthCare Pharmaceuticals Inc., Wayne, New Jersey) as a single agent
is
used less often today for mobilization than G-CSF, because it mobilizes somewhat
less well than G-CSF and because of a relatively higher incidence of both mild
and severe side effects. Erythropoietin (Amgen, Inc.), now commonly used among
cancer patients undergoing chemotherapy to maintain hemoglobin in the near
normal range, also has some ability to mobilize CD34+ cells.
17
Other
Sources of Competition
In
addition to the direct competition outlined above, there is potential for
adverse market effects from other outside developments. For example, producing
a
new drug with fewer side effects reduces the need for anti-side effects
therapies. Because of this, we must monitor a broad area of anticancer R&D
and be ready to fine-tune our development as needed.
The
biotechnology and biopharmaceutical industries are characterized by rapid
technological developments and intense competition. This competition comes
both
from biotech firms and from major pharmaceutical and chemical companies. Many
of
these companies have substantially greater financial, marketing and human
resources than we do (including, in some cases, substantially greater experience
in clinical testing, manufacturing and marketing of pharmaceutical products).
Our drug candidates’ competitive position among other biotech and
biopharmaceutical companies may be based on, among other things, patent
position, product efficacy, safety, reliability, availability, patient
convenience, delivery devices, and price, as well as the development and
marketing of new competitive products.
We
also
experience competition in the development of our drug candidates from
universities and other research institutions and compete with others in
acquiring technology from such universities and institutions. In addition,
certain of our drug candidates may be subject to competition from products
developed using other technologies, some of which have completed numerous
clinical trials. As a result, our actual or proposed drug candidates could
become obsolete before we recoup any portion of our related R&D and
commercialization expenses. However, we believe our competitive position is
enhanced by our commitment to research leading to the discovery and development
of new products and manufacturing methods.
Some
of
our competitors are actively engaged in R&D in areas where we also are
developing drug candidates. The competitive marketplace for our drug candidates
is significantly dependent upon the timing of entry into the market. Early
entrants may have important advantages in gaining product acceptance and market
share contributing to the product’s eventual success and profitability.
Accordingly, in some cases, the relative speed with which we can develop
products, complete the testing, receive approval, and supply commercial
quantities of the product to the market is vital towards establishing a strong
competitive position.
Our
ability to sell to the government also can be influenced by indirect competition
from other providers of products and services. For instance, a major
breakthrough in an unrelated area of biodefense could cause a major reallocation
of government funds from radiation protection. Likewise, an outbreak or
threatened outbreak of some other form of disease or condition may also cause
a
reallocation of funds away from the condition that Protectan CBLB502 is intended
to address.
GOVERNMENT
REGULATION
The
R&D, manufacturing and marketing of drug candidates are subject to
regulation, primarily by the FDA in the U.S. and by comparable authorities
in
other countries. These national agencies and other federal, state, local and
foreign entities regulate, among other things, R&D activities (including
testing in primates and in humans) and the testing, manufacturing, handling,
labeling, storage, record keeping, approval, advertising and promotion of the
products that we are developing. Noncompliance with applicable requirements
can
result in various adverse consequences, including approval delays or refusals
to
approve drug licenses or other applications, suspension or termination of
clinical investigations, revocation of approvals previously granted, fines,
criminal prosecution, recalls or seizures of products, injunctions against
shipping drugs, and total or partial suspension of production and/or refusal
to
allow a company to enter into governmental supply contracts.
The
process of obtaining FDA approval for a new drug may take many years and
generally involves the expenditure of substantial resources. The steps required
before a new drug can be produced and marketed for human use include clinical
trials and the approval of an NDA.
18
Preclinical
Testing
In
the
preclinical phase of development, the promising compound is subjected to
extensive laboratory and animal testing to determine if the compound is
biologically active and safe.
Investigational
New Drug (IND)
Before
human tests can start, the drug sponsor must file an IND application with the
FDA, showing how the drug is made and the results of animal testing. IND status
allows initiation of clinical investigation within 30 days of filing if the
FDA
does not respond with questions during the 30-day period.
Human
Clinical Testing
The
human
clinical testing program usually involves three phases that generally are
conducted sequentially, but which, particularly in the case of anti-cancer
and
other life-saving drugs, may overlap or be combined. Clinical trials are
conducted in accordance with protocols that detail the objectives of the study,
the parameters to be used to monitor safety and the efficacy criteria to be
evaluated. Each protocol is submitted to the FDA as part of the IND filing.
Each
clinical study is conducted under the direction of an independent Institutional
Review Board, or IRB, for each institution at which the study will be conducted.
The IRB will consider, among other things, all existing pharmacology and
toxicology information on the product, ethical factors, the risk to human
subjects and the potential benefits of therapy relative to risk.
In
Phase
I clinical trials, studies usually are conducted on healthy volunteers or,
in
the case of certain terminal illnesses such as advanced prostate cancer,
patients with the disease who have failed to respond to other treatment, to
determine the maximum tolerated dose, side effects and pharmacokinetics of
a
product. Phase II studies are conducted on a small number of patients having
a
specific disease to determine initial efficacy in humans for that specific
disease, the most effective doses and schedules of administration, and possible
adverse effects and safety risks. Phase II/III differs from Phase II in that
the
trials involved may include more patients and, at the sole discretion of the
FDA, be considered the “pivotal” trials, or trials that will form the basis for
FDA approval. Phase III normally involves the pivotal trials of a drug,
consisting of wide-scale studies on patients with the same disease, in order
to
evaluate the overall benefits and risks of the drug for the treated disease
compared with other available therapies. The FDA continually reviews the
clinical trial plans and results, and may suggest design changes or may
discontinue the trials at any time if significant safety or other issues
arise.
As
described above, for several of the product opportunities we are pursuing,
we
may apply for approval based upon a rule adopted by the FDA in 2002, titled
“Approval of New Drugs When Human Efficacy Studies Are Not Ethical or Feasible”
(Part 314, Subpart I), which is also referred to as the two animal rule.
Pursuant to this new rule, in situations where it would be unethical to conduct
traditional Phase II and Phase III efficacy studies in humans, as is the case
with countermeasures to a number of weapons of mass destruction, the FDA will
review new drugs for approval on the basis of safety in humans and efficacy
in
relevant animal models.
New
Drug Application (NDA)
Upon
successful completion of Phase III clinical trials, the drug sponsor files
an
NDA with the FDA for approval, containing all information that has been
gathered. The NDA must include the chemical composition of the drug, scientific
rationale, purpose, animal and laboratory studies, results of human tests,
formation and production details, and proposed labeling.
Following
any initial regulatory approval of any drugs we may develop, we will also be
subject to continuing regulatory review, including the review of adverse
experiences and clinical results that are reported after our drug candidates
are
made commercially available. This will include results from any post-marketing
tests or vigilance required as a condition of approval. The manufacturer and
manufacturing facilities we use to make any of our drug candidates will also
be
subject to periodic review and inspection by the FDA. The discovery of any
previously unknown problems with the drug, manufacturer or facility may result
in restrictions on the drug, manufacturer or facility, including withdrawal
of
the drug from the market. We do not have, and currently do not intend to
develop, the ability to manufacture material for our clinical trials or on
a
commercial scale. Reliance on third-party manufacturers entails risks to which
we would not be subject if we manufactured drugs ourselves, including reliance
on the third-party manufacturer for regulatory compliance. Our drug promotion
and advertising is also subject to regulatory requirements and continuing FDA
review.
19
The
testing and approval process is likely to require substantial time and effort,
and there can be no assurance that any FDA approval will be granted on a timely
basis, if at all. The approval process is affected by a number of factors,
primarily the side effects of the drug (safety) and its therapeutic benefits
(efficacy). Additional preclinical or clinical trials may be required during
the
FDA review period and may delay marketing approval. The FDA may also deny an
NDA
if applicable regulatory criteria are not met.
The
FDA
reviews the results of the clinical trials and may order the temporary or
permanent discontinuation of clinical trials at any time if it believes the
drug
candidate exposes clinical subjects to an unacceptable health risk.
Investigational drugs used in clinical studies must be produced in compliance
with current GMP rules pursuant to FDA regulations.
Sales
outside the U.S. of products that we develop will also be subject to regulatory
requirements governing human clinical trials and marketing for drugs and
biological products and devices. The requirements vary widely from country
to
country, but typically the registration and approval process takes several
years
and requires significant resources. In most cases, even if the FDA has not
approved a product for sale in the U.S., the product may be exported to any
country if it complies with the laws of that country and has valid marketing
authorization by the appropriate authority. There are specific FDA regulations
that govern this process.
We
also
are subject to the following risks and obligations, among others:
·
|
The
FDA or foreign regulators may interpret data from pre-clinical testing
and
clinical trials differently than we interpret
them;
|
·
|
If
regulatory approval of a product is granted, the approval may be
limited
to specific indications or limited with respect to its distribution.
In
addition, many foreign countries control pricing and coverage under
their
respective national social security
systems;
|
·
|
The
FDA or foreign regulators may not approve our manufacturing processes
or
manufacturing facilities;
|
·
|
The
FDA or foreign regulators may change their approval policies or adopt
new
regulations;
|
·
|
Even
if regulatory approval for any product is obtained, the marketing
license
will be subject to continual review, and newly discovered or developed
safety or effectiveness data may result in suspension or revocation
of the
marketing license;
|
·
|
If
regulatory approval of the product candidate is granted, the marketing
of
that product would be subject to adverse event reporting requirements
and
a general prohibition against promoting products for unapproved or
“off-label” uses;
|
·
|
In
some foreign countries, we may be subject to official release requirements
that require each batch of the product we produce to be officially
released by regulatory authorities prior to its distribution by us;
and
|
·
|
We
will be subject to continual regulatory review and periodic inspection
and
approval of manufacturing modifications, including compliance with
current
GMP regulations.
|
20
The
manufacturing and marketing of our proposed products and our R&D activities
are and will continue to be subject to regulation by federal, state and local
governmental authorities in the U.S. and other countries. In the U.S.,
pharmaceuticals are subject to rigorous regulation by the FDA, which reviews
and
approves the marketing of drugs. The Federal Food, Drug and Cosmetic Act, the
regulations promulgated thereunder, and other federal and state statutes and
regulations govern, among other things, the testing, manufacturing, labeling,
storage, record keeping, advertising and promotion of our potential
products.
EMPLOYEES
As
of
March 1, 2008, we had 48 employees, 46 of whom were full-time
employees.
Item 2.
Description of Property
Our
corporate headquarters is located at 73 High Street, Buffalo, New York 14203.
We
have approximately 28,000 square feet of laboratory and office space under
a
five year lease through June of 2012. This space serves as the corporate
headquarters and primary research facilities. In addition, we have leased
approximately 2,500 square feet of office space located at 9450 W. Bryn Mawr
Rd., Rosemont, Illinois, 60018 through July 2011. We do not own any real
property.
Item 3.
Legal
Proceedings
As
of
March 1, 2008, we were not a party to any litigation or other legal proceeding.
Item 4.
Submission of Matters to a Vote of Security Holders
Not
applicable.
21
PART
II
Item 5.
Market for Registrant’s Common Equity, Related Stockholder Matters and Issuer
Purchases of Equity Securities
From
July
21, 2006 (our first day of trading) until August 28, 2007, our common stock
was
traded on the NASDAQ Capital Market under the symbol “CBLI.” Our common stock
also traded on the Boston Stock Exchange, first under the symbol “CFB” and then
under the symbol “CBLI” until September 2007. On August 28, 2007, trading of our
stock moved from the NASDAQ Capital Market to the NASDAQ Global
Market.
The
following table sets forth the quarterly high and low selling prices for our
common stock on the NASDAQ Capital Market or NASDAQ Global Market, as
applicable, for the full quarterly periods within the fiscal years ended
December 31, 2007 and December 31, 2006.
Common
Stock
|
|||||||
2007
|
|||||||
High
|
Low
|
||||||
4th
Quarter
|
$
|
13.07
|
$
|
6.64
|
|||
3rd
Quarter
|
$
|
13.89
|
$
|
9.10
|
|||
2nd
Quarter
|
$
|
11.98
|
$
|
8.00
|
|||
1st
Quarter
|
$
|
13.99
|
$
|
4.49
|
|||
2006
|
|||||||
|
High
|
Low
|
|||||
4th
Quarter
|
$
|
5.87
|
$
|
4.25
|
|||
3rd
Quarter
|
$
|
5.58
|
$
|
4.17
|
As
of
March 1, 2008, there were approximately 44
stockholders of record of our common stock. We have not paid cash dividends
on
our common stock and do not intend to do so in the foreseeable
future.
We
made
no repurchases of our securities during the year ended December 31,
2007.
Item 6.
Selected Financial Data
Not
applicable.
Item 7.
Management’s Discussion and Analysis of Financial Condition and Results of
Operations
This
management’s discussion and analysis of financial condition and results of
operations and other portions of this filing contain forward-looking information
that involves risks and uncertainties. Our actual results could differ
materially from those anticipated by the forward-looking information. Factors
that may cause such differences include, but are not limited to, availability
and cost of financial resources, results of our R&D efforts and clinical
trials, product demand, market acceptance and other factors discussed in the
Company’s other SEC filings under the heading “Risk Factors.” This management’s
discussion and analysis of financial condition and results of operations should
be read in conjunction with our financial statements and the related notes
included elsewhere in this filing.
22
Overview
We
incorporated in Delaware and commenced business operations in June 2003. We
secured a $6,000,000 investment via a private placement of Series A Preferred
Stock in March 2005. On July 20, 2006,
we
sold 1,700,000 shares of common stock in our initial public offering at $6.00
per share. The net proceeds from this offering were approximately $8,300,000.
In
connection with the initial public offering, we issued warrants to purchase
170,000 shares of common stock to the underwriters and their designees. Those
warrants have an exercise price of $8.70 per share. Beginning July 21, 2006,
our
common stock was listed on the NASDAQ Capital Market and on the Boston Stock
Exchange under the symbols “CBLI” and “CFB” respectively. On August 28, 2007,
trading of our stock moved from the NASDAQ Capital Market to the NASDAQ Global
Market. In September 2007, we ceased our listing on the Boston Stock Exchange.
On
September 21, 2006, the SEC declared effective a registration statement of
the
Company registering up to 4,453,601 shares of common stock for resale from
time
to time by the selling stockholders named in the prospectus contained in the
registration statement. We will not receive any proceeds from the sale of the
underlying shares of common stock, although to the extent the selling
stockholders exercise warrants for the underlying shares of common stock, we
will receive the exercise price of those warrants. The registration statement
was filed to satisfy registration rights that we had previously granted in
connection with our Series A Preferred transaction.
On
March
16, 2007, we consummated a transaction with various accredited investors
pursuant to which we agreed to sell to the investors, in a
private placement, an aggregate of approximately 4,288,712 shares of
Series B Convertible Preferred Stock, par value $0.005 per share, and Series
B
Warrants to purchase approximately 2,144,356 shares of our common
stock pursuant to a Securities Purchase Agreement of the same date. The
aggregate purchase price paid by the investors for the Series B Preferred
and Series B Warrants was approximately $30,000,000. After related
fees and expenses, we received net proceeds of approximately
$29,000,000. We intend to use the proceeds for general corporate and
working capital purposes.
The
Series B Preferred have an initial conversion price of $7.00 per share, and
in
the event of a conversion at such conversion price, one share of Series B
Preferred would convert into one share of common stock. Based on the closing
price of our stock on March 16, 2007 of $10.19, the Series B Preferred sold
to
investors and issued to certain of the Agents had a market value of $46,660,112.
The Series B Warrants have an exercise price of $10.36 per share, the closing
bid price on the day prior to the private placement. To the extent, however,
that the conversion price of the Series B Preferred or the exercise price of
the
Series B Warrants is reduced as a result of certain anti-dilution protections,
the number of shares of common stock into which the Series B Preferred are
convertible and for which the Series B Warrants are exercisable may
increase.
We
also
issued to the placement agents in the private placement, as compensation for
their services, Series B Preferred, Series B Warrants, and Series C Warrants.
The Agents collectively received Series B Preferred that are convertible into
an
aggregate of 290,298 shares of common stock, Series B Warrants that are
exercisable for an aggregate of 221,172 shares of our common stock, and Series
C
Warrants that are exercisable for 267,074 shares of our common stock. The Series
C Warrants have an exercise price of $11.00 per share, and are also subject
to
antidilution protections that could increase the number of shares of common
stock for which they are exercisable.
In
total,
the securities issued in the private placement were convertible into, or
exercisable for, up to approximately 7,211,612 shares of common stock (subject
to adjustments for stock splits, anti-dilution, etc.). As of March 1, 2008
the
securities issued in the transaction, in the aggregate, were convertible into
or
exercisable for approximately 6,249,469
shares of common stock (subject to adjustments for stock splits,
anti-dilution, etc.).
Proceeds
from these transactions, together with grants we have received, have supported
our R&D activities to date. We are actively seeking new grants and
co-development contacts with premier pharmaceutical partners to support further
development of other promising leads resulting from our R&D
program.
23
On
December 11, 2007, the SEC declared effective a registration statement of the
Company registering up to 5,514,999 shares of common stock for resale from
time
to time by the selling stockholders named in the prospectus contained in the
registration statement. This number represents 5,514,999 shares of common stock
issuable upon the conversion or exercise of the securities issued the Company’s
March 2007 private placement at the current conversion and exercise prices.
Of
these 5,514,999 shares of common stock, 3,717,515 shares are issuable upon
conversion of Series B Preferred and 1,797,484 shares are issuable upon
exercise of the Series B Warrants. We will not receive any proceeds from the
sale of the underlying shares of common stock, although to the extent the
selling stockholders exercise warrants for the underlying shares of common
stock, we will receive the exercise price of those warrants. The registration
statement was filed to satisfy registration rights that we had previously
granted. Subsequent to the effectiveness of the registration statement,
708,743 Series B Preferred were converted and $60,789 in dividends earned were
paid as of December 31, 2007.
Critical
Accounting Policies
Our
management’s discussion and analysis of our financial condition and results of
operations is based upon our financial statements, which have been prepared
in
accordance with generally accepted accounting principles in the U.S., or GAAP.
The preparation of these financial statements requires us to make estimates
and
judgments that affect the reported amounts of our assets, liabilities, revenues,
expenses and other reported disclosures. We base our estimates on historical
experience and on various other assumptions that we believe are reasonable
under
the circumstances.
Note
2 to
our financial statements include disclosure of our significant accounting
policies. While all decisions regarding accounting policies are important,
we
believe that our policies regarding revenue recognition, R&D expenses,
intellectual property related costs and stock-based compensation expense could
be considered critical.
Revenue
Recognition
We
recognize revenue in accordance with Staff Accounting Bulletin No. 104, “Revenue
Recognition.” Our revenue sources consist of government grants, government
contracts and commercial development contracts.
Grant
revenue is recognized using two different methods depending on the type of
grant. Cost reimbursement grants require us to submit proof of costs incurred
that are invoiced by us to the government agency, which then pays the invoice.
In this case, grant revenue is recognized at the time of submitting the invoice
to the government agency.
Fixed-cost
grants require no proof of costs and are paid as a request for payment is
submitted for expenses. The grant revenue under these fixed cost grants is
recognized using a percentage-of-completion method, which uses assumptions
and
estimates. These assumptions and estimates are developed in coordination with
the principal investigator performing the work under the government fixed-cost
grants to determine key milestones, expenses incurred, and deliverables to
perform a percentage-of-completion analysis to ensure that revenue is
appropriately recognized. Critical estimates involved in this process include
total costs incurred and anticipated to be incurred during the remaining life
of
the grant.
Government
contract revenue is recognized periodically upon delivery of an invoice for
allowable R&D expenses according to the terms of the contract. Commercial
development revenues are recognized when the service or development is
delivered.
For
the
grant from Roswell Park Cancer Institute through the State of New York for
collaborative research with the RPCI, we use SFAS 116 to guide the revenue
recognition. In accordance with SFAS 116, contributions received are recorded
as
revenue upon receipt, unless they contain donor-imposed conditions which must
be
met by the recipient, in which case the contributions are deferred until the
conditions are met. Although we currently project the anticipated use of the
funds received, and currently do not expect an event to occur that would result
in repayment of the funds, technically we have not earned the funds until the
qualifying expenses are incurred. The deferred revenue should be recognized
as
the approved direct and indirect costs are incurred, inclusive of our general
overhead allocation.
24
R&D
Expenses
R&D
costs are expensed as incurred. These expenses consist primarily of our
proprietary R&D efforts, including salaries and related expenses for
personnel, costs of materials used in our R&D, costs of facilities and costs
incurred in connection with our third-party collaboration efforts. Pre-approved
milestone payments made by us to third parties under contracted R&D
arrangements are expensed when the specific milestone has been achieved. As
of
December 31, 2007, we had made $300,000 in milestone payments. Once a drug
receives regulatory approval, we will record any subsequent milestone payments
in identifiable intangible assets, less accumulated amortization, and amortize
them evenly over the remaining agreement term or the expected drug life cycle,
whichever is shorter. We expect our R&D expenses to increase as we continue
to develop our drug candidates.
Intellectual
Property Related Costs
We
capitalize costs associated with the preparation, filing and maintenance of
our
intellectual property rights. Capitalized intellectual property is reviewed
annually for impairment. If a patent application is approved, costs paid by
us
associated with the preparation, filing and maintenance of the patent will
be
amortized on a straight line basis over the shorter of 17 years or the
anticipated useful life of the patent. If the patent application is not
approved, costs paid by us associated with the preparation, filing and
maintenance of the patent will be expensed as part of general and administrative
expenses at that time.
Through
December 31, 2006, we had capitalized $252,978 in expenditures associated with
the preparation, filing and maintenance of certain of our patents. For the
year
ending December 31, 2007, we capitalized an additional $206,124 relating to
these costs, totaling $459,102.
Stock-based
Compensation
The
Financial Accounting Standards Board (FASB) issued SFAS No. 123(R) requiring
all
share-based payments to employees, including grants of employee stock options,
be recognized in the statement of operations based at their fair values.
Accordingly, effective January 1, 2005, we value employee stock based
compensation under the provisions of SFAS 123(R) and related
interpretations.
The
fair
value of each stock option granted is estimated on the grant date using the
Black-Scholes option valuation model or the Monte Carlo Simulation depending
on
the terms and conditions present within the specific option being valued. The
assumptions used to calculate the fair value of options granted are evaluated
and revised, as necessary, to reflect our experience. We use a risk-free rate
based on published rates from the St. Louis Federal Reserve at the time of
the
option grant; assume a forfeiture rate of zero; assume an expected dividend
yield rate of zero based on our intent not to issue a dividend in the
foreseeable future; use an expected life based on our best judgment; and compute
an expected volatility based on similar high-growth, publicly-traded,
biotechnology companies. Compensation expense is recognized using the
straight-line amortization method for all stock-based awards.
On
March
1, 2006, we granted 116,750 options pursuant to stock award agreements to
certain employees and key consultants. On July 20, 2006, we granted a total
of
45,000 fully-vested, stock options to our new independent board members (Messrs.
Antal, Kasten, and Perez) pursuant to stock award agreements.
In
the
fiscal year ended December 31, 2007, we granted 520,000 options pursuant to
stock award agreements to certain employees and key consultants. On June 12,
2007 we granted 140,000 fully- vested stock options to the independent board
members (Messrs. Antal, DiCorleto, Kasten, and Perez) pursuant to stock award
agreements.
25
The
Black-Scholes option valuation model was developed for use in estimating the
fair value of traded options that have no vesting restrictions and are fully
transferable. In addition, the Black-Scholes valuations model requires the
input
of highly subjective assumptions including the expected stock price volatility.
Because our employee stock options have characteristics significantly different
from those of traded options, and because changes in subjective input
assumptions can materially affect the fair value estimate, in management’s
opinion, the existing models do not necessarily provide a reliable single
measure of the fair value of our options. For those stock options where
market
conditions are present within the stock options, we utilize Monte Carlo
simulation to value the stock options. There was one issuance throughout the
year for a total of 90,000 options to an outside consultant where Monte Carlo
simulation was used to value the issuance.
We
recognized a total of $3,401,499, $506,078, and $318,111 in expense for options
for the years ended December 31, 2007, 2006, and 2005 respectively.
The
weighted average, estimated grant date fair values of stock options granted
during the years ended December 31, 2007 and 2006 were $6.08 and $3.14,
respectively.
Impact
of Recently Issued Accounting Pronouncements
On
January 1, 2007, the Financial Accounting Standards Board, or FASB,
issued FIN 48, Accounting for Uncertainty in Income Taxes, an
interpretation of FASB Statement No. 109. FIN 48 prescribes a minimum
recognition threshold and measurement methodology that a tax position taken
or
expected to be taken in a tax return is required to meet before being recognized
in the financial statements. The minimum recognition threshold is defined in
FIN
48 as a tax position that is more likely than not to be sustained upon
examination by the applicable taxing authority, including resolution of any
related appeals or litigation processes, based on the technical merits of the
position. If a tax benefit meets this threshold, it is measured and recognized
based on an analysis of the cumulative probability of the tax benefit being
ultimately sustained. There was no impact on our financial statements upon
adoption of FIN 48.
In
December 2007, the FASB issued Statement No. 160, Noncontrolling Interests
in Consolidated Financial Statements — An Amendment of ARB No. 51, or
SFAS 160. SFAS 160 establishes new accounting and reporting standards for
the
noncontrolling interest in a subsidiary and for the deconsolidation of a
subsidiary. Specifically, SFAS 160 requires the recognition of a noncontrolling
interest (minority interest) as equity in the consolidated financial statements
and separate from the parent's equity. The amount of net income attributable
to
the noncontrolling interest will be included in consolidated net income on
the
face of the income statement. In addition, SFAS 160 requires that a parent
recognize a gain or loss in net income when a subsidiary is deconsolidated.
Such
gain or loss will be measured using the fair value of the noncontrolling
equity
investment on the deconsolidation date. SFAS 160 also includes expanded
disclosure requirements regarding the interests of the parent and its
noncontrolling interest. SFAS 160 is effective for fiscal years, and interim
periods within those fiscal years, beginning on or after December 15, 2008.
We do not expect a material impact from the adoption of SFAS 160.
In
December 2007, the FASB issued Statement No. 141 (revised 2007),
Business
Combinations
("SFAS
141(R)"), which replaces SFAS 141. SFAS 141(R) requires an acquiring entity
to
recognize all the assets acquired and liabilities assumed in a transaction
at
the acquisition-date fair value with limited exceptions. In addition, SFAS
141(R) will require acquisition costs to be expensed as incurred, acquired
contingent liabilities will be recorded at fair value at the acquisition
date
and subsequently measured at either the higher of such amount or the amount
determined under existing guidance for non-acquired contingencies, in-process
research and development will be recorded at fair value as an indefinite-lived
intangible asset at the acquisition date, restructuring costs associated
with a
business combination will be generally expensed subsequent to the acquisition
date and changes in deferred tax asset valuation allowances and income tax
uncertainties after the acquisition date generally will affect income tax
expense. SFAS 141(R) also includes a substantial number of new disclosure
requirements. SFAS 141(R) is effective prospectively to business combinations
for which the acquisition date is on or after the beginning of the first
annual
reporting period beginning on or after December 15, 2008. We anticipate
that the prospective application of the provisions of SFAS 141(R) could have
a
material impact on the fair values assigned to assets and liabilities of
future
acquisitions.
26
In
February 2007, the FASB issued Statement No. 159, The Fair Value Option for
Financial Assets and Financial Liabilities, Including an amendment of FASB
Statement No. 115. The Statement permits entities to choose to measure many
financial instruments and certain other items at fair value. The objective
of
the Statement is to improve financial reporting by providing entities with
the
opportunity to mitigate volatility in reporting earnings caused by measuring
related assets and liabilities differently without having to apply complex
hedge
accounting provisions. The Statement is effective as of the beginning of
an
entity’s first fiscal year that begins after November 15, 2007. We are
currently evaluating the Statement to determine what impact, if any, it will
have upon adoption on January 1, 2008.
In
September 2006, the FASB issued Statement No. 157, Fair Value Measurements
(“SFAS 157). SFAS 157 clarifies the principle that fair value should be based
on
the assumptions market participants would use when pricing an asset or liability
and establishes a fair value hierarchy that prioritizes the information used
to
develop those assumptions. Under the Statement, fair value measurements would
be
separately disclosed by level within the fair value hierarchy. SFAS 157 is
effective for financial statements issued for fiscal years beginning after
November 15, 2007 and interim periods within those fiscal years, with early
adoption permitted. We are currently evaluating the Statement to determine
what
impact, if any, it will have on the Company’s consolidated financial statements
upon adoption on January 1, 2008.
Results
of Operations
Our
operating results for the past three fiscal years have been nominal. The
following table sets forth our statement of operations data for the years ended
December 31, 2007, 2006 and 2005, and should be read in conjunction with our
financial statements and the related notes appearing elsewhere in this annual
report on Form 10-K.
|
Year
Ended
December
31,
2007
|
Year
Ended
December 31,
2006
|
Year
Ended
December 31,
2005
|
|||||||
Revenues
|
$
|
2,018,558
|
$
|
1,708,214
|
$
|
1,138,831
|
||||
Operating
expenses
|
27,960,590
|
9,126,315
|
3,626,664
|
|||||||
Net
interest expense (income)
|
(1,003,766
|
)
|
(195,457
|
)
|
(101,378
|
)
|
||||
Other
expense
|
2,058,236
|
-
|
-
|
|||||||
Net
income (loss)
|
$
|
(26,996,502
|
)
|
$
|
(7,222,644
|
)
|
$
|
(2,386,455
|
)
|
Year
Ended December 31, 2007 Compared to Year Ended December 31,
2006
Revenue
Revenue
increased from $1,708,214 for the year ended December 31, 2006 to $2,018,558
for
the year ended December 31, 2007, representing an increase of $310,344 or 18.2%,
resulting
primarily from an increase in revenue from various grants including the
sponsored research agreement with RPCI, the DTRA contract, and the NCI contract.
As
the
term of the BioShield grant ended, the proceeds from the BioShield grant were
$0
for the year ended December 31, 2007 as compared to $1,100,293 for the year
ended December 31, 2006.
27
See
the
table below for further details regarding the sources of our grant and
government contract revenue:
Agency
|
Program
|
|
|
Amount
|
|
|
Period
of Performance
|
|
|
Revenue
2007
|
|
|
Revenue
2006
|
|||
NIH
|
BioShield
program
|
$
|
1,500,000
|
07/2005-01/2007
|
$
|
-
|
$
|
1,100,293
|
||||||||
NIH
|
Phase
I SBIR program
|
$
|
100,000
|
08/2005-01/2006
|
$
|
-
|
$
|
33,334
|
||||||||
NASA
|
Phase
I NASA STTR program
|
$
|
100,000
|
01/2006-01/2007
|
$
|
33,197
|
$
|
66,393
|
||||||||
NIH
|
Phase
II SBIR program
|
$
|
750,000
|
07/2006-06/2008
|
$
|
459,621
|
$
|
212,713
|
||||||||
NIH
|
NCI
Contract
|
$
|
750,000
|
09/2006-08/2008
|
$
|
440,028
|
$
|
90,481
|
||||||||
NY
State / RPCI
|
Sponsored
Research Agreement
|
$
|
3,000,000
|
01/2007-01/2012
|
$
|
329,390
|
$
|
-
|
||||||||
DTRA
|
DTRA
Contract
|
$
|
1,263,836
|
03/2007-03/2010
|
$
|
466,322
|
$
|
-
|
||||||||
$
|
1,728,558
|
$
|
1,503,214
|
We
anticipate our revenue over the next year to be derived mainly from government
grants and contracts. In addition, it is common in our industry for companies
to
enter into licensing agreements with large pharmaceutical companies. To the
extent we enter into such licensing arrangements, we will receive additional
revenue from licensing fees.
Operating
Expenses
Operating
expenses have historically consisted of costs relating to R&D and general
and administrative expenses. R&D expenses have consisted mainly of
supporting our R&D teams, process development, sponsored research at the
Roswell Park Cancer Institute and the Cleveland Clinic, clinical trials and
consulting fees. General and administrative expenses include all corporate
and
administrative functions that serve to support our current and future operations
while also providing an infrastructure to support future growth. Major items
in
this category include management and staff salaries, rent/leases, professional
services and travel-related expenses. We expect these expenses to increase
as a
result of increased legal and accounting fees anticipated in connection with
our
compliance with ongoing reporting and accounting requirements of the SEC and
also to support the expansion of our business.
Operating
expenses increased from $9,126,315 for the year ended December 31, 2006 to
$27,960,590 for the year ended December 31, 2007. This represents an increase
of
$18,834,275 or 206.4%. We
recognized a total of $7,789,305 of non-cash compensation for stock based
compensation for the year December 31, 2007 compared to $506,078 for the year
ended December 31, 2006. If these non-cash stock based compensation expenses
were excluded, operating expenses would have increased from $8,620,237 for
the
year ended December 31, 2006 to $20,171,285 for the year ended December 31,
2007. This represents an increase in operating expenses of $11,551,048 or
134.0%.
This
increase resulted primarily from an increase in R&D expenses from $6,989,804
for the year ended December 31, 2006 to $17,429,652 for the year ended December
31, 2007, an increase of $10,439,848 or 149.4%. The
higher R&D expenses were incurred as a result of increasing the number of
research and development personnel, commencing clinical trials for CBLC102
and
completing the cGMP manufacturing of CBLB502. We recognized a total of $250,682
of non-cash compensation for R&D stock based compensation for the year ended
December 31, 2006 compared to $1,836,787 for the year ended December 31, 2007.
Without the non-cash stock based compensation, the R&D expenses increased
from $6,739,122 for the year ended December 31, 2006 to $15,592,865 for the
year
ended December 31, 2007; an increase of $8,853,743 or 131.4%.
In
addition, general and administrative expenses increased from $2,136,511 for
the
year ended December 31, 2006 to $10,530,938, for the year ended December 31,
2007. This represents an increase of $8,394,427 or 392.9%. These higher general
and administrative expenses were incurred as a result of creating and improving
the infrastructure of the company and the costs associated with being a publicly
traded company.
We
recognized a total of $255,396 of non-cash stock-based compensation for general
and administrative compensation for the year ended December 31, 2006 compared
to
$5,952,517 for the year ended December 31, 2007. Without the non-cash stock
based compensation, the general and administrative expenses increased from
$1,881,115 for the year ended December 31, 2006 to $4,578,421 for the year
ended
December 31, 2007; an increase of $2,697,306 or 143.4%.
28
Until
we
introduce a product to the market, expenses in the categories mentioned above
will be the largest component of our income statement.
Year
Ended December 31, 2006 Compared to Year Ended December 31,
2005
Revenue
Revenue
increased from $1,138,831 for the year ended December 31, 2005 to $1,708,214
for
the year ended December 31, 2006, representing an increase of $569,383 or 50%,
resulting primarily from an increase in proceeds from the $1,500,000 BioShield
grant. The proceeds from the BioShield grant were $1,100,293 for the year ended
December 31, 2006 as compared to $999,556 for all grant proceeds for the year
ended December 31, 2005. Also, we realized $205,000 for the year ended December
31, 2006 through a commercial contract with Peprotech Inc. to develop chemical
compounds compared to $139,275 for the year ended December 31,
2005.
Operating
Expenses
Operating
expenses increased from $3,626,664 for the year ended December 31, 2005 to
$9,126,315 for the year ended December 31, 2006. This represents an increase
of
$5,499,651 or 151.6%. This increase resulted primarily from an increase in
R&D expenses from $2,640,240 for the year ended December 31, 2005 to
$6,989,804 for the year ended December 31, 2006, an increase of $4,349,564
or
164.7%, as we increased the number of research scientists and related projects
and started a number of clinical trials. In addition, general and administrative
expenses increased from $986,424 for the year ended December 31, 2005 to
$2,136,511, for the year ended December 31, 2006. This represents an increase
of
$1,150,087 or 116.6%. These higher general and administrative expenses were
incurred as a result of creating and improving the infrastructure of the Company
and the costs associated with being a publicly traded company.
Liquidity
and Capital Resources
We
have
incurred annual operating losses since our inception, and, as of December 31,
2007, we had an accumulated deficit of $40,641,743. Our
principal sources of liquidity have been cash provided by sales of our
securities, and government grants, contracts and agreements. Our
principal uses of cash have been R&D and working capital. We
expect
our future sources of liquidity to be primarily government grants, equity
financing, licensing fees and milestone payments in the event we enter into
licensing agreements with third parties, and research collaboration fees in
the
event we enter into research collaborations with third parties.
Net
cash
used in operating activities totaled $16,607,922 for the year ended December
31,
2007, compared to $6,653,602 used in operating activities for the same period
in
2006. Net cash used in operating activities totaled $1,730,513 for the same
period in 2005. For all periods, the increase in cash used was primarily
attributable to increased R&D activities and creating and maintaining the
infrastructure necessary to support these R&D activities.
Net
cash
used in investing activities was $442,523 for the year ended December 31, 2007
and $14,281 used for the same period in 2006. The increase in cash used for
investing activities resulted primarily from the maturing of short-term
investments that converted to cash. Net cash used in investing activities was
$2,805,113 for the same period in 2005. The decrease from 2005 to 2006 resulted
from maturity of investments in long-term certificates of deposit.
Net
cash
provided by financing activities totaled $28,200,591 for the year ended December
31, 2007, compared to $8,523,414 provided by financing activities for the same
period in 2006. The increase in cash provided by financing activities was
attributed to the proceeds from the issuance of Series B Preferred in connection
with our private placement offering. Net cash provided by financing activities
totaled $5,647,347 for the same period in 2005. The funds provided for the
year
ended December 31, 2005 were attributable primarily to the net proceeds from
our
initial public offering in July 2006.
29
Under
our
exclusive license agreement with the Cleveland Clinic, we may be responsible
for
making milestone payments to the Cleveland Clinic in amounts ranging from
$50,000 to $4,000,000. The milestones and corresponding payments for Protectan
CBLB502 and Curaxin CBLC102 are set forth below:
$
|
50,000
|
|||
Complete
Phase I studies for Protectan CBLB502
|
$
|
100,000
|
||
File
NDA application for Protectan CBLB502
|
$
|
350,000
|
||
Receive
regulatory approval to sell Protectan CBLB502
|
$
|
1,000,000
|
||
File
IND application for Curaxin CBLC102 (completed May 2006)
|
$
|
50,000
|
||
Commence
Phase II clinical trials for Curaxin CBLC102 (completed January
2007)
|
$
|
250,000
|
||
Commence
Phase III clinical trials for Curaxin CBLC102
|
$
|
700,000
|
||
$
|
1,500,000
|
|||
Receive
regulatory approval to sell Curaxin CBLC102
|
$
|
4,000,000
|
As
of
December 31, 2007, we had accrued and paid $50,000
for the milestone payment relating to the filing of the IND application for
Curaxin CBLC102 and $250,000 for the milestone payment relating to starting
a
Phase II hormone-refractory prostate cancer clinical trial for Curaxin
CBLC102.
Our
agreement with CCF also provides for payment by us to the CCF of royalty
payments calculated as a percentage of the net sales of the drug candidates
ranging from 1-2%, and sublicense royalty payments calculated as a percentage
of
the royalties received from the sublicenses ranging from 5-35%. However, any
royalty payments and sublicense royalty payments assume that we will be able
to
commercialize our drug candidates, which are subject to numerous risks and
uncertainties, including those associated with the regulatory approval process,
our R&D process and other factors. Accrued milestone payments, royalty
payments and sublicense royalty payments are payable upon achievement of the
milestone.
To
more
effectively match short-term investment maturities with cash flow requirements,
we have obtained a working capital line of credit, which is fully secured by
our
short-term investments. This line of credit has an interest rate of prime,
a
borrowing limit of $1,000,000 and expires on September 20, 2008. At December
31,
2007, there were no outstanding borrowings under this credit
facility.
Although
we believe that existing cash resources will be sufficient to finance our
currently planned operations for the near-term (approximately 12 months), such
amounts will not be sufficient to meet our longer-term cash requirements,
including our cash requirements for the commercialization of certain of our
drug
candidates currently in development. We may be required to issue equity or
debt
securities or enter into other financial arrangements, including relationships
with corporate and other partners, in order to raise additional capital.
Depending upon market conditions, we may not be successful in raising sufficient
additional capital for our long-term requirements. In such event, our business,
prospects, financial condition and results of operations could be materially
adversely affected.
The
following factors, among others, could cause actual results to differ from
those
indicated in the above forward-looking statements: the results of our R&D
efforts, the timing and success of preclinical testing, the timing and success
of any clinical trials we may commence in the future, the timing of and
responses to regulatory submissions, the amount of cash generated by our
operations, the amount of competition we face, and how successful we are in
obtaining any required licenses and entering into collaboration arrangements.
30
Impact
of Inflation
We
believe that our results of operations are not dependent upon moderate changes
in inflation rates.
Impact
of Exchange Rate Fluctuations
We
believe that our results of operations are somewhat dependent upon changes
in
foreign currency exchange rates. We have entered into agreements with foreign
third parties to produce one of our drug compounds and are required to make
payments in the foreign currency. As a result, our financial results could
be
affected by changes in foreign currency exchange rates.
As of
December 31, 2007, we are obligated to make payments under these agreements
of
9,715 Euros and 86,412 Australian dollars. We have established means to purchase
forward contracts to hedge against this risk. As of December 31, 2007, we had
9,715 Euros and 86,412 Australian dollars in contracts outstanding.
Off-Balance
Sheet Arrangements
We
have
not entered into any off-balance sheet arrangements.
31
Item 8.
Financial Statements and Supplementary Data
TABLE
OF CONTENTS
Page
|
|
Report
of Independent Registered Public Accounting Firm
|
F-1
|
Financial
Statements:
|
|
Balance
Sheets
|
F-2
|
Statements
of Operations
|
F-4
|
Statements
of Stockholders' Equity and Comprehensive Loss
|
F-5
|
Statement
of Cash Flows
|
F-8
|
Notes
to Financial Statements
|
F-9
|
32
REPORT
OF INDEPENDENT REGISTERED PUBLIC ACCOUNTING FIRM
Board
of
Directors and Stockholders of
Cleveland
BioLabs, Inc.
We
have
audited the accompanying balance sheets of CLEVELAND BIOLABS, INC. as of
December 31, 2007 and 2006, and the related statements of operations,
stockholders' equity and comprehensive income, and cash flows for each of the
years in the three-year period ended December 31, 2007. Cleveland BioLabs,
Inc.'s management is responsible for these financial statements. Our
responsibility is to express an opinion on these financial statements based
on
our audits.
We
conducted our audits in accordance with the standards of the Public Company
Accounting Oversight Board (United States). Those standards require that we
plan
and perform the audit to obtain reasonable assurance about whether the financial
statements are free of material misstatement. The Company is not required to
have, nor were we engaged to perform, an audit of its internal control over
financial reporting. Our audit included consideration of internal control over
financial reporting as a basis for designing audit procedures that are
appropriate in the circumstances, but not for the purpose of expressing an
opinion on the effectiveness of the Company's internal control over financial
reporting. Accordingly, we express no such opinion. An audit also includes
examining, on a test basis, evidence supporting the amounts and disclosures
in
the financial statements, 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 financial position of Cleveland BioLabs Inc. as of
December 31, 2007 and 2006, and the results of its operations and its cash
flows
for each of the years in the three-year period ended December 31 , 2007 in
conformity with accounting principles generally accepted in the United States
of
America.
MEADEN
& MOORE, LTD.
Certified
Public Accountants
Cleveland,
Ohio
March
13,
2008
F-1
CLEVELAND
BIOLABS, INC.
BALANCE
SHEETS
December
31, 2007 and December 31, 2006
December
31
2007
|
December
31
2006
|
||||||
ASSETS
|
|||||||
CURRENT
ASSETS
|
|||||||
Cash
and equivalents
|
$
|
14,212,189
|
$
|
3,061,993
|
|||
Short-term
investments
|
1,000,000
|
1,995,836
|
|||||
Accounts
receivable:
|
|||||||
Trade
|
163,402
|
159,750
|
|||||
Interest
|
50,042
|
42,479
|
|||||
Notes
receivable - Orbit Brands
|
-
|
50,171
|
|||||
Prepaid
expenses
|
325,626
|
434,675
|
|||||
Total
current assets
|
15,751,259
|
5,744,904
|
|||||
EQUIPMENT
|
|||||||
Computer
equipment
|
258,089
|
132,572
|
|||||
Lab
equipment
|
966,517
|
347,944
|
|||||
Furniture
|
274,903
|
65,087
|
|||||
1,499,509
|
545,603
|
||||||
Less
accumulated depreciation
|
313,489
|
142,011
|
|||||
1,186,020
|
403,592
|
||||||
OTHER
ASSETS
|
|||||||
Intellectual
property
|
459,102
|
252,978
|
|||||
Deposits
|
25,445
|
15,055
|
|||||
484,547
|
268,033
|
||||||
TOTAL
ASSETS
|
$
|
17,421,826
|
$
|
6,416,529
|
F-2
CLEVELAND
BIOLABS, INC.
BALANCE
SHEETS
December
31, 2007 and December 31, 2006
December
31
2007
|
December
31
2006
|
||||||
LIABILITIES
AND STOCKHOLDERS' EQUITY
|
|||||||
CURRENT
LIABILITIES
|
|||||||
Accounts
payable
|
$
|
710,729
|
$
|
644,806
|
|||
Deferred
revenue
|
1,670,610
|
-
|
|||||
Dividends
payable
|
396,469
|
-
|
|||||
Accrued
expenses
|
449,774
|
128,569
|
|||||
Total
current liabilities
|
3,227,582
|
773,375
|
|||||
LONG-TERM
LIABILITIES
|
|||||||
Milestone
payable (long-term)
|
-
|
50,000
|
|||||
STOCKHOLDERS'
EQUITY
|
|||||||
Series
B convertible preferred stock, $.005 par value
|
|||||||
Authorized
- 10,000,000 shares at December 31, 2007 and December 31,
2006
|
|||||||
Issued
and outstanding 3,870,267 and 0 shares at December 31, 2007 and December
31, 2006, respectively
|
19,351
|
-
|
|||||
Additional
paid-in capital
|
24,383,695
|
-
|
|||||
Common
stock, $.005 par value
|
|||||||
Authorized
- 40,000,000 shares at December 31, 2007 and December 31,
2006
|
|||||||
Issued
and outstanding 12,899,241 and 11,826,389 shares at December 31,
2007 and
December 31, 2006, respectively
|
64,496
|
59,132
|
|||||
Additional
paid-in capital
|
30,764,914
|
18,314,097
|
|||||
Accumulated
other comprehensive income (loss)
|
-
|
(4,165
|
)
|
||||
Accumulated
deficit
|
(41,038,212
|
)
|
(12,775,910
|
)
|
|||
Total
stockholders' equity
|
14,194,244
|
5,593,154
|
|||||
|
|
||||||
TOTAL
LIABILITIES AND STOCKHOLDERS' EQUITY
|
$
|
17,421,826
|
$
|
6,416,529
|
F-3
CLEVELAND
BIOLABS, INC.
STATEMENT
OF OPERATIONS
Years
Ended December 31, 2007, 2006, and 2005
December
31
2007
|
December
31
2006
|
December
31
2005
|
||||||||
REVENUES
|
||||||||||
Grant
|
$
|
1,728,558
|
$
|
1,503,214
|
$
|
999,556
|
||||
Service
|
290,000
|
205,000
|
139,275
|
|||||||
2,018,558
|
1,708,214
|
1,138,831
|
||||||||
OPERATING
EXPENSES
|
||||||||||
Research
and development
|
17,429,652
|
6,989,804
|
2,640,240
|
|||||||
Selling,
general and administrative
|
10,530,938
|
2,136,511
|
986,424
|
|||||||
Total
operating expenses
|
27,960,590
|
9,126,315
|
3,626,664
|
|||||||
|
|
|
||||||||
LOSS
FROM OPERATIONS
|
(25,942,032
|
)
|
(7,418,101
|
)
|
(2,487,833
|
)
|
||||
OTHER
INCOME
|
||||||||||
Interest
income
|
1,004,853
|
206,655
|
119,371
|
|||||||
Sublease
revenue
|
4,427
|
-
|
-
|
|||||||
OTHER
EXPENSE
|
||||||||||
Interest
expense
|
1,087
|
11,198
|
17,993
|
|||||||
Corporate
relocation
|
1,741,609
|
-
|
-
|
|||||||
Loss
on disposal of fixed assets
|
15,575
|
-
|
-
|
|||||||
Loss
on investment
|
305,479
|
-
|
-
|
|||||||
NET
LOSS
|
(26,996,502
|
)
|
(7,222,644
|
)
|
(2,386,455
|
)
|
||||
DIVIDENDS
ON CONVERTIBLE PREFERRED STOCK
|
(1,265,800
|
)
|
(214,928
|
)
|
(291,914
|
)
|
||||
NET
LOSS AVAILABLE TO COMMON STOCKHOLDERS
|
$
|
(28,262,302
|
)
|
$
|
(7,437,572
|
)
|
$
|
(2,678,369
|
)
|
|
NET
LOSS AVAILABLE TO COMMON STOCKHOLDERS PER SHARE OF COMMON STOCK -
BASIC
AND DILUTED
|
$
|
(2.34
|
)
|
$
|
(0.84
|
)
|
$
|
(0.43
|
)
|
|
WEIGHTED
AVERAGE NUMBER OF SHARES USED IN CALCULATING NET LOSS PER SHARE,
BASIC AND
DILUTED
|
12,090,430
|
8,906,266
|
6,250,447
|
F-4
CLEVELAND
BIOLABS, INC.
STATEMENTS
OF STOCKHOLDERS' EQUITY AND COMPREHENSIVE LOSS
Period
From January 1, 2005 to December 31, 2007
Stockholders'
Equity
|
|||||||||||||
Common
Stock
|
|||||||||||||
Additional
|
|||||||||||||
Paid-in
|
Penalty
|
||||||||||||
Shares
|
Amount
|
Capital
|
Shares
|
||||||||||
Balance
at January 1, 2005
|
5,960,000
|
29,800
|
2,255,954
|
-
|
|||||||||
Issuance
of shares - Series A financing
|
308,000
|
1,540
|
588,122
|
-
|
|||||||||
Issuance
of shares - stock dividend
|
69,201
|
346
|
138,056
|
-
|
|||||||||
Issuance
of options (383,840 options issued, 324,240 outstanding)
|
-
|
-
|
318,111
|
-
|
|||||||||
Exercise
of options (59,600 options exercised)
|
59,600
|
298
|
118,902
|
-
|
|||||||||
Accrue
unissued shares
|
-
|
-
|
(81,125
|
)
|
81,125
|
||||||||
Net
loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Unrealized
holding gains (losses) arising during period
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance
at December 31, 2005
|
6,396,801.00
|
31,984
|
3,338,020
|
81,125
|
|||||||||
Issuance
of shares -previously accrued penalty shares
|
54,060
|
270
|
80,855
|
(81,125
|
)
|
||||||||
Issuance
of shares - stock dividend
|
184,183
|
922
|
367,445
|
-
|
|||||||||
Issuance
of penalty shares
|
15,295
|
76
|
(76
|
)
|
-
|
||||||||
Issuance
of shares -initial public offering
|
1,700,000
|
8,500
|
10,191,500
|
-
|
|||||||||
Fees
associated with initital public offering
|
-
|
-
|
(1,890,444
|
)
|
-
|
||||||||
Conversion
of preferred stock to common stock
|
3,351,219
|
16,756
|
5,291,385
|
-
|
|||||||||
Conversion
of notes payable to common stock
|
124,206
|
621
|
312,382
|
-
|
|||||||||
Issuance
of options
|
-
|
-
|
506,078
|
-
|
|||||||||
Exercise
of options
|
625
|
3
|
2,810
|
-
|
|||||||||
Issuance
of warrants
|
-
|
-
|
114,032
|
-
|
|||||||||
Proceeds
from sales of warrants
|
-
|
-
|
110
|
-
|
|||||||||
Net
loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses) arising during period
|
-
|
-
|
-
|
-
|
|||||||||
Less
reclassification adjustment for (gains) losses included in net loss
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance
at December 31, 2006
|
11,826,389
|
$
|
59,132
|
$
|
18,314,097
|
$
|
-
|
||||||
Issuance
of options
|
-
|
-
|
3,401,499
|
-
|
|||||||||
Options
to be issued in 2008
|
-
|
-
|
2,687,355
|
-
|
|||||||||
Issuance
of shares - Series B financing
|
-
|
-
|
-
|
-
|
|||||||||
Fees
associated with Series B Preferred offering
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of restricted shares
|
190,000
|
950
|
1,699,500
|
-
|
|||||||||
Exercise
of options
|
126,046
|
630
|
110,650
|
-
|
|||||||||
Exercise
of warrants
|
48,063
|
240
|
90,275
|
-
|
|||||||||
Conversion
of Series B Preferred Shares to Common
|
708,743
|
3,544
|
4,461,537
|
-
|
|||||||||
Dividends
on Series B Preferred shares
|
-
|
-
|
-
|
-
|
|||||||||
Net
Loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses) arising during period
|
-
|
-
|
-
|
-
|
|||||||||
Less
reclassification adjustment for (gains) losses included in
net loss
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance
at December 31, 2007
|
|
12,899,241
|
$
|
64,496
|
|
$
|
30,764,914 |
|
$ |
-
|
F-5
CLEVELAND
BIOLABS, INC.
STATEMENTS
OF STOCKHOLDERS' EQUITY AND COMPREHENSIVE LOSS
Period
From January 1, 2005 to December 31, 2007
Stockholders'
Equity
|
|||||||||||||
Preferred
Stock
|
|||||||||||||
Additional
|
|||||||||||||
Paid-in
|
Penalty
|
||||||||||||
Shares
|
Amount
|
Capital
|
Shares
|
||||||||||
Balance
at January 1, 2005
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of shares -Series A financing
|
3,051,219
|
15,256
|
5,292,885
|
-
|
|||||||||
Issuance
of shares - stock dividend
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of options (383,840 options issued, 324,240 outstanding)
|
-
|
-
|
-
|
-
|
|||||||||
Exercise
of options (59,600 options exercised)
|
-
|
-
|
-
|
-
|
|||||||||
Accrue
unissued shares
|
(360,000
|
)
|
360,000
|
||||||||||
Net
loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Unrealized
holding gains (losses) arising during period
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance
at December 31, 2005
|
3,051,219
|
15,256
|
4,932,885
|
360,000
|
|||||||||
Issuance
of shares - previously accrued penalty shares
|
240,000
|
1,200
|
358,800
|
(360,000
|
)
|
||||||||
Issuance
of shares - stock dividend
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of penalty shares
|
60,000
|
300
|
(300
|
)
|
-
|
||||||||
Issuance
of shares -initial public offering
|
-
|
-
|
-
|
-
|
|||||||||
Fees
associated with initital public offering
|
-
|
-
|
-
|
-
|
|||||||||
Conversion
of preferred stock to common stock
|
(3,351,219
|
)
|
(16,756
|
)
|
(5,291,385
|
)
|
-
|
||||||
Conversion
of notes payable to common stock
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of options
|
-
|
-
|
-
|
-
|
|||||||||
Exercise
of options
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of warrants
|
-
|
-
|
-
|
-
|
|||||||||
Proceeds
from sales of warrants
|
-
|
-
|
-
|
-
|
|||||||||
Net
loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses) arising during
period
|
-
|
-
|
-
|
-
|
|||||||||
Less
reclassification adjustment for (gains) losses included in net
loss
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance
at December 31, 2006
|
-
|
$ |
-
|
$
|
-
|
$
|
-
|
||||||
Issuance
of options
|
-
|
-
|
-
|
-
|
|||||||||
Options
to be issued in 2008
|
-
|
-
|
-
|
-
|
|||||||||
Issuance
of shares -Series B financing
|
4,579,010
|
22,895
|
32,030,175
|
-
|
|||||||||
Fees
associated with Series B Preferred offering
|
-
|
-
|
(3,184,943
|
)
|
-
|
||||||||
Issuance
of restricted shares
|
-
|
-
|
-
|
-
|
|||||||||
Exercise
of options
|
-
|
-
|
-
|
-
|
|||||||||
Exercise
of warrants
|
-
|
-
|
-
|
-
|
|||||||||
Conversion
of Series B Preferred Shares to Common
|
(708,743
|
)
|
(3,544
|
)
|
(4,461,537
|
)
|
-
|
||||||
Dividends
on Series B Preferred shares
|
-
|
-
|
-
|
-
|
|||||||||
Net
Loss
|
-
|
-
|
-
|
-
|
|||||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses)
|
|||||||||||||
arising
during period
|
-
|
-
|
-
|
-
|
|||||||||
Less
reclassification adjustment for (gains) losses
|
|||||||||||||
included
in net loss
|
-
|
-
|
-
|
-
|
|||||||||
Comprehensive
loss
|
|||||||||||||
Balance at December 31, 2007 |
3,870,267
|
$
|
19,351 |
$
|
24,383,695
|
$
|
-
|
F-6
STATEMENTS
OF STOCKHOLDERS' EQUITY AND COMPREHENSIVE LOSS
Period
From January 1, 2005 to December 31, 2007
Stockholders'
Equity
|
|||||||||||||
Other
Comprehensive
Income/(Loss)
|
Accumulated
Deficit
|
Total
|
Comprehensive
Income
(Loss)
|
||||||||||
Balance
at January 1, 2005
|
-
|
(2,659,968)
|
(374,214)
|
||||||||||
Issuance
of shares - Series A financing
|
-
|
-
|
5,897,803
|
||||||||||
Issuance
of shares - stock dividend
|
-
|
(138,433
|
)
|
(31
|
)
|
||||||||
Issuance
of options (383,840 options issued, 324,240 outstanding)
|
-
|
-
|
318,111
|
||||||||||
Exercise
of options (59,600 options exercised)
|
-
|
-
|
119,200
|
||||||||||
Accrue
unissued shares
|
-
|
-
|
-
|
||||||||||
Net
loss
|
-
|
(2,386,455
|
)
|
(2,386,455
|
)
|
(2,386,455
|
)
|
||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Unrealized
holding gains (losses) arising during period
|
(17,810
|
)
|
-
|
(17,810
|
)
|
$
|
(17,810
|
)
|
|||||
Comprehensive
loss
|
$
|
(2,404,265
|
)
|
||||||||||
Balance
at December 31, 2005
|
(17,810
|
)
|
(5,184,856
|
)
|
3,556,604
|
||||||||
Issuance
of shares - previously accrued penalty shares
|
-
|
-
|
-
|
||||||||||
Issuance
of shares - stock dividend
|
-
|
(368,410
|
)
|
(43
|
)
|
||||||||
Issuance
of penalty shares
|
-
|
-
|
-
|
||||||||||
Issuance
of shares - initial public offering
|
-
|
-
|
10,200,000
|
||||||||||
Fees
associated with initital public offering
|
-
|
-
|
(1,890,444
|
)
|
|||||||||
Conversion
of preferred stock to common stock
|
-
|
-
|
-
|
||||||||||
Conversion
of notes payable to common stock
|
-
|
-
|
313,003
|
||||||||||
Issuance
of options
|
-
|
-
|
506,078
|
||||||||||
Exercise
of options
|
-
|
-
|
2,813
|
||||||||||
Issuance
of warrants
|
-
|
-
|
114,032
|
||||||||||
Proceeds
from sales of warrants
|
-
|
-
|
110
|
||||||||||
Net
loss
|
-
|
(7,222,644
|
)
|
(7,222,644
|
)
|
(7,222,644
|
)
|
||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses) arising during period
|
6,678
|
-
|
6,678
|
$
|
6,678
|
||||||||
Less
reclassification adjustment for (gains) losses included in net
loss
|
6,967
|
-
|
6,967
|
$
|
6,967
|
||||||||
Comprehensive
loss
|
$
|
(7,208,999
|
)
|
||||||||||
Balance
at December 31, 2006
|
$
|
(4,165
|
)
|
$
|
(12,775,910
|
)
|
$
|
5,593,154
|
|||||
Issuance
of options
|
-
|
-
|
3,401,499
|
||||||||||
Options
to be issued in 2008
|
-
|
-
|
2,687,355
|
||||||||||
Issuance
of shares - Series B financing
|
-
|
-
|
32,053,070
|
||||||||||
Fees
associated with Series B Preferred offering
|
-
|
-
|
(3,184,943
|
)
|
|||||||||
Issuance
of restricted shares
|
-
|
-
|
1,700,450
|
||||||||||
Exercise
of options
|
-
|
-
|
111,280
|
||||||||||
Exercise
of warrants
|
-
|
-
|
90,515
|
||||||||||
Conversion
of Series B Preferred Shares to Common
|
-
|
-
|
-
|
||||||||||
Dividends
on Series B Preferred shares
|
-
|
(1,265,800
|
)
|
(1,265,800
|
)
|
||||||||
Net
Loss
|
-
|
(26,996,502
|
)
|
(26,996,502
|
)
|
(26,996,502
|
)
|
||||||
Other
comprehensive income
|
|||||||||||||
Unrealized
gains (losses) on short term investments
|
|||||||||||||
Changes
in unrealized holding gains (losses) arising during period
|
-
|
-
|
-
|
$
|
-
|
||||||||
Less
reclassification adjustment for (gains) losses included in net
loss
|
4,165
|
-
|
4,165
|
$
|
4,165
|
||||||||
Comprehensive
loss
|
$
|
(26,992,337
|
)
|
||||||||||
Balance
at December 31, 2007
|
$
|
-
|
$
|
(41,038,212
|
)
|
$
|
14,194,244
|
F-7
STATEMENTS
OF CASH FLOWS
For
the
Years Ended December 31, 2007, 2006 and 2005
2007
|
2006
|
2005
|
||||||||
CASH
FLOWS FROM OPERATING ACTIVITIES
|
||||||||||
Net
loss
|
$
|
(26,996,502
|
)
|
$
|
(7,222,644
|
)
|
$
|
(2,386,455
|
)
|
|
Adjustments
to reconcile net loss to net cash used by operating
activities:
|
||||||||||
Depreciation
|
188,395
|
94,931
|
44,762
|
|||||||
Noncash
interest expense
|
-
|
9,929
|
17,993
|
|||||||
Noncash
salaries and consulting expense
|
7,789,305
|
620,119
|
437,311
|
|||||||
Deferred
compensation
|
-
|
5,886
|
9,141
|
|||||||
Loss
on disposal of fixed assets
|
15,575
|
-
|
-
|
|||||||
Loss
on investments
|
305,479
|
-
|
-
|
|||||||
Changes
in operating assets and liabilities:
|
||||||||||
Accounts
receivable - trade
|
(3,652
|
)
|
(159,750
|
)
|
225,013
|
|||||
Accounts
receivable - interest
|
(12,870
|
)
|
(5,616
|
)
|
(37,035
|
)
|
||||
Prepaid
expenses
|
109,049
|
(422,427
|
)
|
(12,249
|
)
|
|||||
Deposits
|
(10,390
|
)
|
(3,750
|
)
|
(3,734
|
)
|
||||
Accounts
payable
|
65,923
|
380,023
|
10,869
|
|||||||
Deferred
revenue
|
1,670,610
|
(100,293
|
)
|
100,293
|
||||||
Accrued
expenses
|
321,206
|
99,990
|
(136,421
|
)
|
||||||
Milestone
payments
|
(50,000
|
)
|
50,000
|
-
|
||||||
Total
adjustments
|
10,388,630
|
569,042
|
655,942
|
|||||||
Net
cash (used by) provided by operating activities
|
(16,607,872
|
)
|
(6,653,602
|
)
|
(1,730,513
|
)
|
||||
CASH
FLOWS FROM INVESTING ACTIVITIES
|
||||||||||
Purchase
of short-term investments
|
(1,000,000
|
)
|
(4,800,000
|
)
|
(2,400,000
|
)
|
||||
Sale
of short-term investments
|
2,000,000
|
5,200,000
|
-
|
|||||||
Issuance
of notes receivable
|
(250,000
|
)
|
(50,000
|
)
|
-
|
|||||
Purchase
of equipment
|
(987,649
|
)
|
(187,660
|
)
|
(328,756
|
)
|
||||
Sale
of equipment
|
1,250
|
-
|
-
|
|||||||
Costs
of patents pending
|
(206,124
|
)
|
(176,621
|
)
|
(76,357
|
)
|
||||
Net
cash (used in) provided by investing activities
|
(442,523
|
)
|
(14,281
|
)
|
(2,805,113
|
)
|
||||
CASH
FLOWS FROM FINANCING ACTIVITIES
|
||||||||||
Issuance
of preferred stock
|
30,020,984
|
-
|
6,000,000
|
|||||||
Financing
costs
|
(1,152,857
|
)
|
(1,679,456
|
)
|
(402,622
|
)
|
||||
Dividends
|
(869,331
|
)
|
(43
|
)
|
(31
|
)
|
||||
Issuance
of common stock
|
-
|
10,200,000
|
-
|
|||||||
Exercise
of stock options
|
111,280
|
2,813
|
-
|
|||||||
Exercise
of warrants
|
90,515
|
-
|
-
|
|||||||
Issuance
of warrants
|
-
|
100
|
-
|
|||||||
Proceeds
from convertible notes payable
|
-
|
-
|
50,000
|
|||||||
Net
cash (used in) provided by financing activities
|
28,200,591
|
8,523,414
|
5,647,347
|
|||||||
INCREASE
(DECREASE) IN CASH AND EQUIVALENTS
|
11,150,196
|
1,855,531
|
1,111,721
|
|||||||
CASH
AND EQUIVALENTS AT BEGINNING OF PERIOD
|
3,061,993
|
1,206,462
|
94,741
|
|||||||
CASH
AND EQUIVALENTS AT END OF PERIOD
|
$
|
14,212,189
|
$
|
3,061,993
|
$
|
1,206,462
|
||||
Supplemental
disclosures of cash flow information:
|
||||||||||
Cash
paid during the period for interest
|
$
|
1,087
|
$
|
1,269
|
$
|
-
|
||||
Cash
paid during the year for income taxes
|
$
|
-
|
$
|
-
|
$
|
-
|
||||
Supplemental
schedule of noncash financing activities:
|
||||||||||
Issuance
of stock options to employees, consultants, and independent board
members
|
$
|
3,401,499
|
$
|
506,078
|
$
|
318,511
|
||||
Stock
options due to employees and a consultant
|
$
|
2,687,355
|
$
|
-
|
$
|
-
|
||||
Issuance
of shares to consultants
|
$
|
1,700,450
|
$
|
368,367
|
$
|
-
|
||||
Issuance
of non-cash financing fees
|
$
|
2,032,086
|
$
|
-
|
$
|
-
|
||||
Conversion
of preferred stock to common stock
|
$
|
4,465,081
|
$
|
5,308,141
|
$
|
-
|
||||
Accrual
of preferred stock dividends
|
$
|
396,469
|
$
|
-
|
$
|
-
|
||||
Common
stock issued as financing fees on issuance of preferred
shares
|
$
|
-
|
$
|
-
|
$
|
589,662
|
||||
Conversion
of notes payable and accrued interest to preferred stock
|
$
|
-
|
$
|
-
|
$
|
102,438
|
||||
Issuance
of warrants to consultant
|
$
|
-
|
$
|
114,042
|
$
|
-
|
||||
Exercise
of stock options into 59,600 common shares by consultant
|
$
|
-
|
$
|
-
|
$
|
119,200
|
||||
Issuance
of common stock dividend to preferred shareholders
|
$
|
-
|
$
|
-
|
$
|
138,402
|
||||
Unissued
shares to preferred shareholders for penalty per agreement
|
$
|
-
|
$
|
-
|
$
|
441,125
|
||||
Conversion
of notes payable and accrued interest to common stock
|
$
|
-
|
$
|
313,003
|
$
|
-
|
F-8
CLEVELAND
BIOLABS, INC.
NOTES
TO FINANCIAL STATEMENTS
Note
1. Organization
Cleveland
BioLabs, Inc. (“CBLI” or the “Company”) is engaged in the discovery, development
and commercialization of products for cancer treatment and protection of normal
tissues from radiation and toxins. The Company was incorporated under the laws
of the State of Delaware on June 5, 2003 and is headquartered in Buffalo, New
York. CBLI is a drug discovery and development company leveraging its
proprietary discoveries about programmed cell death to treat cancer and protect
normal tissues from exposure to radiation and other stresses. To date, the
Company has not developed any commercial products, but the Company has developed
and produced biological compounds under a single commercial development
contract.
Note
2. Summary of Significant Accounting Policies
A.
|
Cash
and Equivalents – The Company considers highly liquid debt
instruments with original maturities of three months or less to be
cash
equivalents. In addition, the Company maintains cash and equivalents
at
financial institutions, which may exceed federally insured amounts
at
times and which may, at times, significantly exceed balance sheet
amounts
due to outstanding checks.
|
B.
|
Marketable
Securities and Short Term Investments – The Company considers investments
with a maturity date of more than three months to maturity to be
short-term investments and has classified these securities as
available-for-sale. Such investments are carried at fair value, with
unrealized gains and losses included as accumulated other comprehensive
income (loss) in stockholders’ equity. The cost of available-for-sale
securities sold is determined based on the specific identification
method.
|
C.
|
Accounts
Receivable – The Company extends unsecured credit to customers under
normal trade agreements, which generally require payment within 30
days.
Management estimates an allowance for doubtful accounts which is
based
upon management’s review of delinquent accounts and an assessment of the
Company’s historical evidence of collections. There is no allowance for
doubtful accounts as of December 31, 2007, and
2006.
|
D.
|
Notes
Receivable – On December 7, 2006 the Company entered into an agreement
with the Orbit Brands Corporation (Borrower) and its subsidiaries
whereby
the Company would lend up to $150,000 each on two promissory notes
to the
Borrower at a rate of 5% per annum with a maturity date of one year.
The
proceeds of the loans were to be used by the Borrower solely to cover
expenses associated with converting the notes into common stock and
preparing the lending motions for the bankruptcy case involving the
Borrower. The
Company is under no obligation to fund or loan any additional amount
to
the Borrower. As
of December 31, 2006 the balance outstanding was $50,000 plus accrued
interest of $171. At
September 30, 2007, the Company wrote off the balance outstanding
of
$300,000 plus accrued interest of $5,479 due to the fact that the
Securities and Exchange Commission has initiated proceedings to
permanently suspend trading in the shares of Borrower and to revoke
its
registration under the Securities Exchange Act of 1934. On December
11,
2007, the SEC revoked the registrations of all classes of securities
of
Orbit Brands Corp. pursuant to Section 12(j) of the Securities Exchange
Act of 1934. In addition, the Borrower does not appear to have sufficient
funds to emerge from its bankruptcy
proceedings.
|
F-9
E.
|
Deferred
Compensation – The Company realized deferred compensation upon the
valuation of restricted stock granted to the founding stockholders.
This
deferred compensation was expensed over the three-year vesting period
from
the grant of the stock. The Company expensed $0, $5,887, and $9,140
in
compensation expense in 2007, 2006 and 2005,
respectively.
|
F.
|
Equipment
– Equipment is stated at cost and depreciated over the estimated useful
lives of the assets (generally five years) using the straight-line
method.
Leasehold improvements are depreciated on the straight-line method
over
the shorter of the lease term or the estimated useful lives of the
assets.
Expenditures for maintenance and repairs are charged to expense as
incurred. Major expenditures for renewals and betterments are capitalized
and depreciated. Depreciation expense was $188,395, $94,931, and
$44,762
for the years ended December 31, 2007, 2006, and 2005
respectively.
|
G.
|
Impairment
of Long-Lived Assets – In accordance with Statements of Financial
Accounting Standards, or SFAS, No. 144, Accounting for the Impairment
or
Disposal of Long-Lived Assets, long-lived assets to be held and used,
including equipment and intangible assets subject to depreciation
and
amortization, are reviewed for impairment whenever events or changes
in
circumstances indicate that the carrying amounts of the assets or
related
asset group may not be recoverable. Determination of recoverability
is
based on an estimate of discounted future cash flows resulting from
the
use of the asset and its eventual disposition. In the event that
such cash
flows are not expected to be sufficient to recover the carrying amount
of
the asset or asset group, the carrying amount of the asset is written
down
to its estimated net realizable
value.
|
H.
|
Intellectual
Property – The Company capitalizes the costs associated with the
preparation, filing, and maintenance of certain intellectual property
rights. Capitalized intellectual property is reviewed annually for
impairment.
|
A
portion
of this intellectual property is owned by the Cleveland Clinic Foundation,
or
CCF,
and
granted to the Company through an exclusive licensing agreement. As part of
the
licensing
agreement, CBLI agrees to bear the costs associated with the preparation, filing
and maintenance of patent applications relating to this intellectual property.
If the patent application is approved, the costs paid by the Company are
amortized on a straight-line basis over the shorter of 17 years or the
anticipated useful life of the patent. If the patent application is not
approved, the costs associated with the preparation, filing and maintenance
of
the patent application by the Company on behalf of CCF will be expensed as
part
of selling, general and administrative expenses. Gross capitalized patents
pending costs were $407,425 and $222,789 on behalf of CCF for 13 patent
applications as of December 31, 2007 and December 31, 2006, respectively. All
of
the CCF patent applications are still pending approval.
F-10
The
Company also has submitted three patent applications as a result of intellectual
property exclusively developed and owned by the Company. If the patent
applications are approved, costs paid by the Company associated with the
preparation, filing, and maintenance of the patents will be amortized on a
straight-line basis over the shorter of 17 years or the anticipated useful
life
of the patent. If the patent application is not approved, the costs associated
with the preparation, filing and maintenance of the patent application will
be
expensed as part of selling, general and administrative expenses at that time.
Gross capitalized patents pending costs were $51,677 and $30,189 on behalf
of
the Company for three patent applications as of December 31, 2007 and December
31, 2006, respectively. The patent applications are still pending
approval.
I.
|
Line
of Credit – The Company has a working capital line of credit that is fully
secured by short-term investments. This fully-secured, working capital
line of credit carries an interest rate of prime, a borrowing limit
of
$1,000,000, and expires on September 20, 2008. At December 31, 2007
and
December 31, 2006, there were no outstanding
borrowings.
|
J.
|
Fair
Value of Financial Instruments – Financial instruments, including cash and
equivalents, accounts receivable, notes receivable, accounts payable
and
accrued liabilities, are carried at net realizable
value.
|
K.
|
Use
of Estimates – The preparation of financial statements in conformity with
accounting principles generally accepted in the U.S. requires management
to make estimates and assumptions that affect the reported amounts
of
assets and liabilities and disclosure of contingent assets and liabilities
at the date of the financial statements and the reported amounts
of
revenues and expenses during the reporting period. The Company bases
its
estimates on historical experience and on various other assumptions
that
the Company believes to be reasonable under these circumstances.
Actual
results could differ from those
estimates.
|
L.
|
Revenue
Recognition – The Company recognizes revenue in accordance with Staff
Accounting Bulletin No. 104, “Revenue Recognition.” Revenue sources
consist of government grants, government contracts and commercial
development contracts.
|
Revenues
from government grants and contracts are for research and development purposes
and are recognized in accordance with the terms of the award and the government
agency. Grant revenue is recognized in one of two different ways depending
on
the grant. Cost reimbursement grants require us to submit proof of costs
incurred that are invoiced by us to the government agency, which then pays
the
invoice. In this case, grant revenue is recognized at the time of submitting
the
invoice to the government agency. Fixed cost grants require no proof of costs
and are paid as a request for payment is submitted for expenses. The grant
revenue under these fixed costs grants is recognized using a
percentage-of-completion method, which uses assumptions and estimates. These
assumptions and estimates are developed in coordination with the principal
investigator performing the work under the government fixed-cost grants to
determine key milestones, expenses incurred, and deliverables to perform a
percentage-of-completion analysis to ensure that revenue is appropriately
recognized. Critical estimates involved in this process include total costs
incurred and anticipated to be incurred during the remaining life of the grant.
The
Company recognizes revenue related to the funds received in 2007 from the State
of New York under the sponsored research agreement with the Roswell Park Cancer
Institute (RPCI) in accordance with SFAS 116. The principles of SFAS 116 result
in the recognition of revenue as allowable costs are incurred. The Company
recognizes revenue on research laboratory services and the purchase and
subsequent use of related equipment. The amount paid as a payment toward future
services related to the equipment is recognized as a prepaid asset and will
be
recognized as revenue as the services are performed and the prepaid asset is
recognized as expense.
F-11
Government
contract revenue is recognized periodically upon delivery of an invoice for
allowable R&D expenses according to the terms of the contract. The Company
has recognized grant revenue from the following agencies: the Defense Threat
Reduction Agency (DTRA), the U.S. Army (DARPA), National Aeronautics and Space
Administration (NASA), the National Institutes of Health (NIH) and the
Department of Health and Human Services (HHS). Commercial development revenues
are recognized when the service or development is delivered.
M. |
Deferred
Revenue – Deferred revenue results when payment is received in advance of
revenue being earned. The Company makes a determination as to whether
the
revenue has been earned by applying a percentage-of-completion analysis
to
compute the need to recognize deferred revenue. The percentage of
completion method is based upon (1) the total income projected for
the
project at the time of completion and (2) the expenses incurred to
date.
The percentage-of-completion can be measured using the proportion
of costs
incurred versus the total estimated cost to complete the
contract.
|
The
Company received $2,000,000 in funds from the State of New York through the
Roswell Park Cancer Institute during the second quarter of 2007 and is
recognizing this revenue over the terms and conditions of the sponsored research
agreement. The Company recognizes revenue on research laboratory services and
the purchase and subsequent use of related equipment. The amount paid as a
payment toward future services related to the equipment is recognized as a
prepaid asset and will be recognized as revenue as the services are performed
and the prepaid asset is recognized as expense. For the year ended December
31,
2007, the Company recognized $329,390 as revenue resulting in a balance of
deferred revenue of $1,670,610 at December 31, 2007. At December 31, 2006 and
December 31, 2005, the Company had $0 and $100,293 deferred revenue. The balance
of $100,293 at December 31, 2005 was related to a federal grant and recognized
as revenue in the first quarter of 2006.
N.
|
Research
and Development – Research and development expenses consist primarily of
costs associated with the clinical trials of drug candidates, compensation
and other expenses for research and development, personnel, supplies
and
development materials, costs for consultants and related contract
research
and facility costs. Expenditures relating to research and development
are
expensed as incurred.
|
O.
|
Other
Expense – Other expense consists primarily of costs associated with the
relocation of the Company facilities and employees from Cleveland,
Ohio to
Buffalo, New York. For the years ended December 31, 2007, 2006, and
2005,
relocation costs were $1,741,609, $0 and $0, respectively. Other
expense
also includes the loss on the Orbit Brands investment (see Note 1.D
for
more information), loss on disposal of assets and interest expense.
The
loss on disposal of fixed assets was $15,575, $0 and $0 for the years
ended December 31, 2007, 2006 and 2005, respectively. Interest expense
was
$1,087, $11,198 and $17,993 for the years ended December 31, 2007,
2006
and 2005, respectively.
|
P.
|
Employee
Benefit Plan – The Company maintains a 401(k) retirement savings plan that
is available to all full-time employees who have reached age 21.
The plan
is intended to qualify under Section 401(k) of the Internal Revenue
Code
of 1986, as amended. The plan provides that each participant may
contribute up to a statutory limit of their pre-tax compensation,
which
was $15,500 for employees under age 50 and $20,500 for employees
50 and
older in calendar year 2007. Employee contributions are held in the
employees’ name and invested by the plan trustee. The plan currently
provides for the Company to make matching contributions, subject
to
established limits. The Company made matching contributions of $99,530,
$48,858, and $0 for 2007, 2006, and 2005,
respectively.
|
F-12
Q.
|
2006
Equity Incentive Plan - On May 26, 2006, the Company’s Board of
Directors adopted the 2006 Equity Incentive Plan (“Plan”) to attract and
retain persons eligible to participate in the Plan, motivate Participants
to achieve long-term Company goals, and further align Participants’
interests with those of the Company’s other stockholders. The Plan expires
on May 26, 2016 and allows up to 2,000,000 shares of stock to be
awarded.
For the year ended December 31, 2006, 45,000 options were granted
to
independent board members. On February 14, 2007, the 2,000,000
shares were registered with the SEC by filing a Form S-8 registration
statement. For the year ended December 31, 2007, 190,000 stock awards
and
660,000 options were granted to executives, independent board members,
employees and key consultants. At December 31, 2007, stock awards
of
190,000 and option awards of 705,000 have been awarded under the
Plan
leaving 1,015,000 shares of stock to be
awarded.
|
R.
|
2007
Executive Compensation Plan - On May 11, 2007, the Compensation
Committee (the “Compensation
Committee”)
of the Board of Directors approved an executive compensation program
designed to reward each of the Company’s Chief Executive Officer,
Chief
Operating Officer,
Chief Financial Officer
and Chief Scientific Officer (the “Executive Officers”) for the
achievement of certain pre-determined milestones. The purpose of
the
program is to link each Executive Officer’s compensation to the
achievement of key Company milestones that the Compensation Committee
believes have a strong potential to create long-term stockholder
value.
|
Under
the
terms of this program, after each fiscal year beginning with the fiscal year
ended December 31, 2007, each component of our Executive Officers’
compensation packages
- base
salary, cash bonus and stock option awards -
will be
measured against the Company’s achievement of (1) stock performance milestones,
(2) scientific milestones, (3) business
milestones and (4) financial milestones, each of which will be weighted equally.
The milestones will be set at the beginning of each fiscal year. Each set of
milestones has a threshold level, a target level and a high performance level.
For base salary, increases will range between 2% for threshold performance
to 6%
for high performance. For cash bonuses, increases will range between 15% for
threshold performance and 60% for high performance. For stock option awards,
awards will range between 50,000 stock options for threshold performance and
300,000 for high performance.
As
of
December 31, 2007, the Company has accrued $185,288 in cash bonuses and
$2,687,355 in non-cash, stock-based compensation for the stock options to be
awarded by the Compensation Committee under the 2007 Executive Compensation
Plan.
S.
|
Stock-Based
Compensation - The FASB issued SFAS No. 123(R) (revised December
2004),
Share Based Payment, which is a revision of SFAS No. 123 Accounting
for
Stock-Based Compensation. SFAS 123(R) requires all share-based payments
to
employees, including grants of employee stock options, to be recognized
in
the statement of operations based on their fair values. The Company
values
employee non-cash, stock-based compensation under the provisions
of SFAS
123(R) and related interpretations.
|
The
fair
value of each stock option granted is estimated on the grant date. The Black
Scholes model is used for standard stock options, but if market conditions
are
present within the stock options, the Company utilizes Monte Carlo simulation
to
value the stock options. The assumptions used to calculate the fair value of
options granted are evaluated and revised, as necessary, to reflect the
Company’s experience. The Company uses a risk-free rate published by the St.
Louis Federal Reserve at the time of the option grant, assumes a forfeiture
rate
of zero, assumes an expected dividend yield rate of zero based on the Company’s
intent not to issue a dividend in the foreseeable future, uses an expected
life
based on the safe harbor method, and computes an expected volatility based
on
similar high-growth, publicly-traded, biotechnology companies. The Company
does
not include the use of its own stock in the volatility calculation at this
time
because of the brief history of the stock as a publicly traded security on
a
listed exchange. The Company recognizes the fair value of share-based
compensation in net income on a straight-line basis over the requisite service
period. A summary of the values for these assumptions appears
below:
F-13
|
2007
|
2006
|
2005
|
|||||||
Risk-free
interest rate
|
3.38-5.11
|
%
|
4.66-5.04
|
%
|
3.95-4.46
|
%
|
||||
Expected
dividend yield
|
0
|
%
|
0
|
%
|
0
|
%
|
||||
Expected
life
|
2.74-6
years
|
5
years
|
5
Years
|
|||||||
Expected
volatility
|
71.86-76.29
|
%
|
71.43-75.11
|
%
|
78.0-81.75
|
%
|
The
fair
value of warrants issued in 2006 to a key consultant in exchange for services
was also estimated using the Black-Scholes option valuation model with the
same
assumptions.
The
Company issued 660,000, 161,750 and 383,840 stock options during the years
ended
December 31, 2007, 2006, and 2005, respectively,
pursuant to various stock award agreements. The Company recognized a total
of
$3,401,499, $506,078, and $318,111 in expense related to options for the years
ended December 31, 2007, 2006, and 2005, respectively. The weighted average,
estimated grant date fair values of stock options granted was $6.08, $3.14,
and
$1.65 during the years ended December 31, 2007, 2006, and 2005,
respectively.
The
following tables summarize the stock option activity for the years
ended
December 31, 2007 and December 31, 2006, respectively.
|
Shares
|
Weighted
Average
Exercise
Price per
Share
|
Weighted
Average
Remaining
Contractual
Term (in Years)
|
|||||||
Outstanding,
December 31, 2006
|
483,490
|
$
|
2.17
|
|||||||
Granted
|
660,000
|
$
|
9.85
|
|||||||
Exercised
|
131,750
|
$
|
1.34
|
|||||||
Forfeited,
Canceled
|
0
|
n/a
|
||||||||
Outstanding,
December 31, 2007
|
1,011,740
|
$
|
7.29
|
8.80
|
||||||
Exercisable,
December 31, 2007
|
646,930
|
$
|
6.89
|
8.75
|
F-14
Shares
|
Weighted
Average
Exercise
Price per
Share
|
Weighted
Average
Remaining
Contractual
Term (in Years)
|
||||||||
Outstanding,
December 31, 2005
|
324,240
|
$
|
0.82
|
|||||||
Granted
|
161,750
|
$
|
4.92
|
|||||||
Exercised
|
625
|
$
|
4.50
|
|||||||
Forfeited,
Canceled
|
1,875
|
$
|
4.50
|
|||||||
Outstanding,
December 31, 2006
|
483,490
|
$
|
2.17
|
8.77
|
||||||
Exercisable,
December 31, 2006
|
243,183
|
$
|
2.27
|
8.78
|
In
addition, the Company recognized $1,700,450 in expense for shares issued under
the Plan to various consultants during the year ended December 31, 2007. For
the
years ended December 31, 2006 and 2005, there was no compensation expense
recognized due to no share issuance.
The
Black-Scholes option valuation model was developed for use in estimating the
fair value of traded options that have no vesting restrictions and are fully
transferable. In addition, the Black-Scholes valuation model requires the input
of highly subjective assumptions including the expected stock price volatility.
Because the Company’s employee stock options and warrants have characteristics
significantly different from those of traded derivative securities, and because
changes in subjective input assumptions can materially affect the fair value
estimate, in management’s opinion, the existing models do not necessarily
provide a reliable single measure of the fair value of the Company’s options and
warrants.
T.
|
Income
Taxes – The Company utilizes Statement of Financial Accounting Standards
No. 109, “Accounting for Income Taxes,” which requires an asset and
liability approach to financial accounting and reporting for income
taxes.
Deferred tax assets and liabilities are recognized for the estimated
future tax consequences attributable to operating loss and tax credit
carryforwards, and temporary differences between the financial statement
carrying amounts of existing assets and liabilities and their respective
tax bases. Deferred tax assets and liabilities are measured using
enacted
tax rates expected to apply to taxable income in the years in which
those
operating loss carryforward and temporary differences are expected
to be
recovered or settled. The effect on deferred income tax assets and
liabilities of a change in tax rates is recognized in income in the
period
that includes the enactment date. A valuation allowance is established,
if
necessary, to reduce the deferred tax asset to the amount that will,
more
likely than not, be realized. The Company accounts for interest and
penalties related to uncertain tax positions as part of its provision
for
income taxes.
|
F-15
U.
|
Net
Loss Per Share – Basic and diluted net loss per share has been computed
using the weighted-average number of shares of common stock outstanding
during the period.
|
The
following table presents the basic and diluted earnings per share:
2007
|
2006
|
2005
|
||||||||
Net
loss available to common stockholders
|
$
|
(28,262,302
|
)
|
$
|
(7,437,572
|
)
|
$
|
(2,678,369
|
)
|
|
Net
loss per share, basic and diluted
|
$
|
(2.34
|
)
|
$
|
(0.84
|
)
|
$
|
(0.43
|
)
|
|
Weighted-average
shares used in computing net loss per share, basic and
diluted
|
12,090,430
|
8,906,266
|
6,250,447
|
The
Company has included $1,265,800, $214,928 and $291,914 in the numerator to
account for cumulative dividends for Series A and B preferred stock that were
recognized for 2007, 2006 and 2005, respectively. The
Company has excluded all outstanding warrants and options from the calculation
of diluted net loss per share because all such securities are antidilutive
for
all applicable periods presented.
The
total
number of shares excluded from the calculations of diluted net loss per share,
prior to application of the treasury stock method for warrants,
was
3,453,268, 814,424, and 594,424 for the years ended December 31, 2007, 2006
and
2005, respectively. Such securities, had they been dilutive, would have been
included in the computation of diluted earnings per share.
The
total
number of shares excluded from the calculations of diluted net loss per share,
prior to the application of the treasury stock method for options,
was
1,011,740, 483,490, and 324,240 for the years ended December 31, 2007, 2006
and
2005, respectively. Such securities, had they been dilutive, would have been
included in the computation of diluted earnings per share.
V.
|
Concentrations
of Risk – Grant revenue was comprised wholly from grants issued by the
federal and state governments and accounted for 85.6%, 88.0%, and
88.9% of
total revenue for the years ended December 31, 2007, 2006 and 2005,
respectively. Although the Company anticipates ongoing grant revenue,
there is no guarantee that this revenue stream will continue in the
future.
|
Financial
instruments that potentially subject us to a significant concentration of credit
risk consist primarily of cash and cash equivalents and securities
available-for-sale. The Company maintains deposits in federally insured
institutions in excess of federally insured limits. The Company does not believe
it is exposed to significant credit risk due to the financial position of the
depository institutions in which those deposits are held. Additionally, the
Company has established guidelines regarding diversification of its investment
portfolio and maturities of investments, which are designed to meet safety
and
liquidity.
F-16
W.
|
Foreign
Currency Exchange Rate Risk – The Company has entered into agreements with
foreign third parties to advance the Company’s research and development
efforts and is required to make payments in the foreign currency.
As a
result, the Company’s financial results could be affected by changes in
foreign currency exchange rates. As of December 31, 2007, the Company
is
obligated to make payments under the agreement of 9,715 Euros and
86,412
Australian dollars. The Company has established means to purchase
forward
contracts to hedge against this risk. As of December 31, 2007, the
Company
has 9,715 Euros and 86,412 Australian dollars in contracts outstanding.
The
estimated fair values of forward contracts are based on quoted market
prices.
|
X.
|
Comprehensive
Income/(Loss) – The Company applies Statement of Financial Accounting
Standards (SFAS) No. 130, “Reporting Comprehensive Income.” SFAS No. 130
requires disclosure of all components of comprehensive income on
an annual
and interim basis. Comprehensive income is defined as the change
in equity
of a business enterprise during a period from transactions and other
events and circumstances from non-owner
sources.
|
Y.
|
Segment
Reporting – As of December 31, 2007, the Company has determined that it
operates in only one segment. Accordingly, no segment disclosures
have
been included in the notes to the consolidated financial
statements.
|
Z.
|
Effect
of New Accounting Standards – Effective January 1, 2007, the Company
adopted Financial Accounting Standards Board FIN 48, Accounting for
Uncertainty in Income Taxes, an interpretation of FASB Statement
No. 109.
FIN 48 prescribes a minimum recognition threshold and measurement
methodology that a tax position taken or expected to be taken in
a tax
return is required to meet before being recognized in the financial
statements. The minimum recognition threshold is defined in FIN 48
as a
tax position that is more likely than not to be sustained upon examination
by the applicable taxing authority, including resolution of any related
appeals or litigation processes, based on the technical merits of
the
position. If a tax benefit meets this threshold, it is measured and
recognized based on an analysis of the cumulative probability of
the tax
benefit being ultimately sustained. There was no impact to the financial
statements upon adoption of FIN 48.
|
Note
3. Significant Alliances and Related Parties
The
Cleveland Clinic Foundation
Effective
July 2004, the Company entered into a strategic alliance with CCF. Under the
agreement, the Company received an exclusive license to use CCF licensed patents
and CCF technology for the benefit of the Company for research and drug
development. The Company has primary responsibility to fund all newly developed
patents; however, CCF retains patent ownership on those contained in the
agreement. The Company also has the responsibility to secure applicable
regulatory approvals. In partial consideration of this agreement, in December
2004, the Company issued 1,341,000 shares of its common stock to CCF and
recognized $2,250,000 as non-cash research and development expense in exchange
for the stock. The calculation of this expense was based in part on an estimate
of the Company’s value based on discussions in 2004 with potential investors, in
which the Company was estimated to have a value of approximately $12,500,000.
This valuation was reflected in an agreement between the Company and an
investment bank dated September 30, 2004. This agreement set forth the terms
on
which the investment bank was to raise equity capital for the Company. In light
of the preliminary and subjective nature of that estimate, the Company
discounted that estimate to arrive at a valuation of $10,000,000.
F-17
CCF
will
receive milestone payments for each product developed with CCF technology as
development passes through major developmental stages. In addition, the Company
will pay CCF royalties and sublicense royalties as a percentage of net sales
of
all commercial products developed with CCF technology. Milestone payments,
royalties or sublicense royalties have been paid amount to $300,000, $0, and
$0
for the years ended December 31, 2007, 2006, and 2005,
respectively.
The
Company also incurred $927,347, $1,142,290, and $475,934 in subcontract expense
to CCF related to research grants and agreements for the years ended December
31, 2007, 2006 and 2005, respectively. The balance remaining is $70,539 in
accrued payables at December 31, 2007. Finally, the Company recognizes a balance
of $10,227 in accounts receivable at December 31, 2007 related to a prepayment
of a research agreement.
The
Company also rented office and laboratory space from an entity related to CCF
on
a month to month basis through May of 2005. Rent to this entity related to
CCF
was $0, $0, and $11,121 in 2007, 2006 and 2005, respectively.
Roswell
Park Cancer Institute
In
January 2007, the Company entered into a sponsored research agreement with
RPCI
to
develop the Company’s cancer and radio-protectant drug candidates.
The
Company received $2,000,000 in funds from RPCI during the second quarter of
2007
and is scheduled to receive an additional $1,000,000 in the second quarter
of
2008. This money was funded by the State of New York as part of an incentive
package for the Company to relocate and establish
a major research/clinical facility in
Buffalo,
New York. The Company will have an open-ended license to any basic research
conducted within, or in collaboration with RPCI.
ChemBridge
Corporation
In
April
2004, ChemBridge Corporation acquired 357,600 shares of the Company’s common
stock valued at $6,081 (subject to antidilution provisions for future equity
issues) and holds warrants to purchase an additional 264,624 shares of the
Company’s common stock for $1.13 per share. The warrants expire in April 2010.
Under the agreement, ChemBridge has agreed to provide chemical technology and
expertise for the benefit of the Company for research and drug
development.
In
April
2004, the Company entered into a chemical libraries license agreement with
ChemBridge. Under the terms of the agreement, the Company has a non-exclusive
worldwide license to use certain chemical compound libraries for drug research
conducted on its own or in collaboration with others. In return, ChemBridge
will
receive royalty payments on any revenue received by the Company for all
contracts, excluding CCF, in which the libraries are used. No revenues or
royalties have been paid through the year ended December 31, 2007.
The
Company has also agreed to collaborate with ChemBridge on two optimization
projects, wherein ChemBridge will have the responsibility of providing the
chemistry compounds of the project and the Company will have the responsibility
of providing the biological expertise. ChemBridge will retain a 50% ownership
interest in two selected “confirmed hits” that make up the optimization
projects.
The
parties will jointly manage the development and commercialization of any
compounds arising from an optimization project. No “confirmed hits” have been
selected during the year ended December 31, 2007.
In
addition, the Company paid ChemBridge $41,780, $29,910, and $3,913 for the
purchase of chemical compounds in the normal course of business in 2007, 2006
and 2005, respectively.
F-18
Cooperative
Research and Development Agreement
In
August
2004, the Company entered into a five-year cooperative research and development
agreement (CRADA) with the Uniformed Service University of the Health Sciences,
which includes the Armed Forces Radiobiology Research Institute, the Henry
M.
Jackson Foundation for the Advancement of Military Medicine, Inc., and CCF,
to
evaluate the Company’s radioprotective drug candidates and their effects on
intracellular and extracellular signaling pathways. Under the terms of the
agreement, all parties are financially responsible for their own expenses
related to the agreement. The agreement may be unilaterally terminated by any
party upon 30 days prior written notice.
In
August
2007, the Company entered into an additional one-year CRADA with the Uniformed
Service University of the Health Sciences to evaluate the Company’s
radioprotective drug candidate Protectan CBLB502 in non-human primates. Under
the terms of the agreement, the Company paid $222,769 to Henry M. Jackson
Foundation for the Advancement of Military Medicine, Inc to purchase, house
and
irradiate animals and perform blood and cytokine analysis. The agreement may
be
unilaterally terminated by any party upon 30 days prior written
notice.
Sunrise
Securities Corp.
The
Company engaged Sunrise
Securities Corporation,
or
SSC,
to act
as the investment banker for the private placement that took place in March
2005,
as a
lead underwriter for the initial public offering in 2006,
and
as placement agent for its private placement of Series B Convertible Preferred
Stock, or Series B Preferred. SSC and its related
parties
are owners of both common stock and warrants of the Company as a result of
the
private placement and the initial public offering. The Company paid SSC $75,000
as an initial retainer for underwriting work associated with the initial public
offering and SSC
received $945,000
in
underwriting commissions from
the
initial public offering. In addition, the Company paid SSC $95,000 related
to
legal fees incurred in the March 2007 Series B Preferred
offering.
Consultants
In
addition, one Company
stockholder, who serves as our Chief Scientific Officer, received payments
for
consulting services performed on certain grant awards and internal research
and
development. Total cash subcontract expense made to this person amounted to
$120,580, $104,168, and $100,250 for the years ended December 31, 2007, 2006
and
2005, respectively. In addition, the Company incurred $198,375 in non-cash,
stock-based compensation expense in 2007 to this consultant and also accrued
an
additional $732,915 in non-cash, stock-based compensation and an additional
$19,215 in subcontractor expense related to the 2007 Executive Compensation
Plan.
One
Company stockholder, who serves as our Vice President of Research -
Radioprotectant Group, received payment for consulting services performed
related to the Company’s research efforts. Total consultant expense made to this
person amounted to $95,520, $84,330, and $49,135 for the years ended December
31, 2007, 2006, and 2005, respectfully.
Note
4. Stock Transactions
In
March
2005, the Company issued 3,000,000 shares of Series A Participating Convertible
Preferred Stock (Series A) for $6 million in gross proceeds. These shares were
convertible into common stock on a one-for-one basis and earn a dividend of
6%
payable biannually on February 1 and August 1 in cash or common stock. In
conjunction with the issuance of the Series A shares, $50,000 of convertible
notes held at December 31, 2004 and a $50,000 note issued February 3, 2005,
including accrued interest, were converted into 51,219 shares of Series A
preferred stock. The Company also issued 308,000 shares of common stock and
300,000 warrants to purchase 300,000 shares of common stock with an exercise
price of $2.00 per share to Sunrise Securities Corp., the private placement
agent, and its designees as partial consideration for their services rendered.
295,850 of these warrants expire on March 15, 2010 and 4,150 expire on March
28,
2010 resulting from two closing dates.
F-19
In
March
2005, the Company issued 10,000 stock options under a non-qualified stock option
agreement to a consultant who works for the company on an ongoing basis. These
options allow for the purchase of common stock at a price of $3.00 per share.
These options have a thirteen month vesting schedule and expire on March 1,
2015. The value of the options is being recognized as consulting expense over
the vesting period based on the Black-Scholes option pricing model.
In
July
2005, the Company issued 294,240 stock options to various employees of the
Company under non-qualified stock option agreements. These options allow for
the
purchase of 190,000 shares of common stock at a price of $.66 and 104,240 shares
of common stock at a price of $0.67 per share, respectively. These options
have
a three-year vesting schedule and expire on June 30, 2015. The value of the
options is being recognized as compensation expense over the vesting period
based on the Black-Scholes option pricing model.
In
July
2005, the company issued fully vested options to purchase 59,600 shares of
common stock under a non-qualified stock option agreement to an outside
consultant who works for the company on an ongoing basis. These stock options
were exercised at a price of $2.00 per share and the company recorded $119,200
in consulting fees as a result of the issuance of these stock
options.
On
August
1, 2005, the Company paid a stock dividend of 69,201 shares of common stock
to
holders of record of the outstanding Series A preferred stock.
In
December 2005, the Company issued 20,000 stock options under a non-qualified
stock option agreement to a consultant who works for the company on an ongoing
basis. These options allow for the purchase of common stock at a price of $2.00
per share with a two-year vesting schedule and expire on November 30,
2015.
As
a
condition of the issuance of the Series A preferred stock in March 2005, all
holders of Series A preferred stock received an additional 2% of all preferred
stock, common stock and warrants that each Series A preferred stockholder owns
for each 30 day period that a delay occurs in a required transaction. These
penalty shares are not subject to compounding or prorating based on the number
of days of delay. They are earned at the end of each 30-day penalty period.
For
the first quarter of 2006, one penalty period occurred in which 60,000 shares
of
Series A preferred stock were earned at $120,000. In addition, 13,515 shares
of
common stock were earned at $27,030. The penalty shares were issued in January
2006.
Pursuant
to an Amendment to the Series A Rights Agreement, dated as of February 17,
2006,
the Company’s obligation to issue penalty shares was suspended for a period of
70 days, subject to a one-time 45-day extension, while the Company’s
registration statement was being reviewed by the SEC.
On
February 1, 2006, the Company paid a common stock dividend of 91,776 shares
to
holders of the Series A preferred stock to satisfy the dividend requirement
of
the preferred stock issuance.
On
March
1, 2006, the Company issued 116,750 stock options to various employees and
consultants of the Company under non-qualified stock option agreements. These
options allow for the purchase of 116,750 shares of common stock at a price
of
$4.50. These options have a three-year vesting schedule and expire on February
29, 2016.
On
June
21, 2006, after the expiration of the 115-day extension and an additional 30-day
period, the Company incurred one additional penalty period in which 60,000
shares of Series A preferred stock were earned at $120,000 and 15,295 shares
of
common stock were earned at $30,590. The Company has not incurred any further
obligation to issue penalty shares since these issuances.
F-20
On
July
20, 2006, the Company sold 1,700,000 shares of common stock in its initial
public offering at $6.00 per share. The net proceeds to the Company from this
offering were approximately $8,300,000. Beginning July 21, 2006, the Company’s
shares were quoted on the NASDAQ Capital Market and listed on the Boston Stock
Exchange under the symbols “CBLI” and “CFB” respectively. On August 28, 2007,
trading of the Company’s common stock moved from the NASDAQ
Capital
Market to the NASDAQ
Global
Market. In September 2007, we ceased our listing on the Boston Stock Exchange.
In connection with its initial public offering, the Company sold warrants to
purchase 170,000 shares of common stock to the underwriters and their designees
at a cost of $100.00. The warrants have an exercise price of $8.70 per
share.
On
July
20, 2006, the effective date of the Company’s initial public offering, the
Company issued 92,407 shares of common stock as accumulated dividends to the
Series A preferred stockholders. On the same date, all of the Company’s Series A
Preferred shares automatically converted on a one-for-one basis into 3,351,219
shares of common stock and notes of the Company in the principal amount of
$283,500 plus accrued interest of $29,503 automatically converted into 124,206
shares of common stock. In connection with their appointment to the Board,
the
Company issued to each of the Company’s three new independent directors, options
to purchase 15,000 shares of common stock with an exercise price of $6.00 per
share.
On
September 21, 2006, the SEC declared effective a registration statement of
the
Company registering up to 4,453,601 shares of common stock for resale from
time
to time by the selling stockholders named in the prospectus contained in the
registration statement. The Company will not receive any proceeds from the
sale
of the underlying shares of common stock, although to the extent the selling
stockholders exercise warrants for the underlying shares of common stock, the
Company will receive the exercise price of those warrants. The registration
statement was filed to satisfy registration rights that the Company had
previously granted.
On
November 16, 2006 the Company issued 50,000 warrants to an outside consultant.
These warrants are immediately exercisable into common shares of the Company
and
have an exercise price of $6.00 per share and an expiration date of November
16,
2011.
On
February 14, 2007, the Company issued 99,500 stock options to various employees
and consultants of the Company under non-qualified stock option agreements.
These options allow for the purchase of 99,500 shares of common stock at a
price
of $9.14. These options have various vesting schedules from immediate vesting
to
three years and expire on February 14, 2017.
On
February 26, 2007, the Company issued 55,000 warrants at an exercise price
of
$9.19 per share, to a placement agent as incentive for work on the private
placement offering.
On
March
16, 2007, the Company entered into a Securities Purchase Agreement with various
accredited investors (the Buyers),
pursuant to which the Company agreed to sell to the Buyers Series B Convertible
Preferred Stock (Series
B
Preferred)
convertible into an aggregate of 4,288,712 shares of common stock and Series
B
Warrants that are exercisable for an aggregate of 2,144,356 shares of common
stock. The Series B Preferred have an initial conversion price of $7.00 per
share, and in the event of a conversion at such conversion price, one share
of
Series B Preferred would convert into one share of common stock. The Series
B
Warrants have an exercise price of $10.36 per share, the closing bid price
on
the day prior to the private placement. To the extent, however, that the
conversion price of the Series B Preferred or the exercise price of the Series
B
Warrants is reduced as a result of certain anti-dilution protections, the number
of shares of common stock into which the Series B Preferred are convertible
and
for which the Series B Warrants are exercisable may increase.
The
Company also issued to the placement agents in the private placement (the
Agents),
as
compensation for their services, Series B Preferred, Series B Warrants, and
Series C Warrants. The Agents collectively received Series B Preferred that
are
convertible into an aggregate of 290,298 shares of common stock, Series B
Warrants that are exercisable for an aggregate of 221,172 shares of the
Company’s common stock, and Series C Warrants that are exercisable for 267,074
shares of the Company’s common stock. The Series C Warrants have an exercise
price of $11.00 per share, and are also subject to anti-dilution protections
that could increase the number of shares of common stock for which they are
exercisable.
F-21
In
total,
the securities issued in the private placement will be convertible into, or
exercisable for, up to approximately 7,211,612 shares of common stock, which
amount is subject to adjustment in the event of certain corporate events such
as
stock splits or issuances of securities at a price below the conversion price
of
the Series B Preferred or exercise price of the warrants, as the case may be.
On
September 13, 2007, the Company paid $807,913 to the Series B Preferred
stockholders
for the
semiannual dividend.
On
March
19, 2007, the Company issued 20,000 stock options to members of the Scientific
Advisory Board of the Company under non-qualified stock option agreements.
These
options are immediately exercisable and allow for the purchase of 20,000 shares
of common stock at a price of $8.82. These options expire on March 18,
2017.
On
April
6, 2007, the Company issued 152,500 stock options to officers and consultants
under non-qualified stock option agreements. These options are immediately
exercisable and allow for the purchase of 152,500 shares of common stock at
a
price of $8.36. These options expire on April 5,
2017.
The Company also issued 115,000 shares of common stock to consultants under
the
Plan.
On
June
12, 2007, the Company issued 140,000 stock options to four independent members
of the Board of Directors of the Company under non-qualified stock option
agreements. These options are immediately exercisable and allow for the purchase
of 140,000 shares of common stock at a price of $9.40. These options expire
on
June 11,
2017.
On
June
15, 2007, the Company issued 110,000 stock options to various key employees
and
consultants under non-qualified stock option agreements. These options have
various vesting schedules including immediate vesting, up to three year vesting,
and vesting upon the company stock price obtaining certain levels. These options
allow for the purchase of 110,000 shares of common stock at a price ranging
from
$9.93 to $17.00. These options expire on June 14,
2017.
The Company also issued 30,000 shares of common stock to the same consultants
under the Plan.
On
June
21, 2007, the Company issued 3,000 stock options to a consultant under a
non-qualified stock option agreement. These options vest over a six month period
and allow for the purchase of 3,000 shares of common stock at a price of $10.84.
These options expire on June 20,
2017.
On
June
27, 2007, the Company issued 30,000 shares of common stock to various outside
consultants under the Plan.
On
July
18, 2007, the Company issued 15,000 shares of common stock to an outside
consultant under the Plan. On that date, the Company also issued 18,000 stock
options to another consultant under a non-qualified stock option agreement.
These options are immediately exercisable and allow for the purchase of 18,000
shares of common stock at a price of $10.61. These options expire on December
31, 2012.
On
December 4, 2007, the Company issued 117,000 stock options to various key
employees and consultants under non-qualified stock option agreements. These
options have up to three year vesting. These options allow for the purchase
of
117,000 shares of common stock at an exercise price of $10.00 per share. These
options expire on or before December 3,
2017.
On
December 11, 2007, the SEC declared effective a registration statement of the
Company registering up to 5,514,999 shares of common stock for resale from
time
to time by the selling stockholders named in the prospectus contained in the
registration statement. This number represents 5,514,999 shares of common stock
issuable upon the conversion or exercise of the securities issued the Company’s
March 2007 private placement at the current conversion and exercise prices.
Of
these 5,514,999 shares of common stock, 3,717,515 shares are issuable upon
conversion of Series B Preferred and
1,797,484 shares are issuable upon exercise of the Series B Warrants. The
Company will not receive any proceeds from the sale of the underlying shares
of
common stock, although to the extent the selling stockholders exercise warrants
for the underlying shares of common stock, the Company will receive the exercise
price of those warrants. The registration statement was filed to satisfy
registration rights that the Company had previously granted. Subsequent to
the
effectiveness of the registration statement, 708,743 Series B Preferred were
converted and $61,418 in dividends earned were paid as of December 31, 2007.
At
December 31, 2007, $396,469 in dividends were accrued on the outstanding Series
B Preferred.
F-22
Note
5. Income Taxes
The
provisions for income taxes charged to continuing operations is $0 for the
years
ended December 31, 2007, 2006, and 2005, respectively.
Deferred
tax assets (liabilities) are comprised of the following at December
31:
2007
|
2006
|
2005
|
||||||||
Deferred
tax assets:
|
||||||||||
Operating
loss carryforwards
|
$
|
13,289,000
|
$
|
4,586,000
|
$
|
1,897,000
|
||||
Tax
credit carryforwards
|
737,000
|
-
|
-
|
|||||||
Deferred
compensation
|
2,765,000
|
345,000
|
135,000
|
|||||||
Other
|
-
|
2,000
|
7,000
|
|||||||
Total
deferred income tax assets
|
16,791,000
|
4,933,000
|
2,039,000
|
|||||||
Deferred
tax liabilities
|
||||||||||
Equipment
|
(61,000
|
)
|
(35,000
|
)
|
(17,000
|
)
|
||||
Net
deferred income tax asset
|
16,730,000
|
4,898,000
|
2,022,000
|
|||||||
Valuation
allowance
|
(16,730,000
|
)
|
(4,898,000
|
)
|
(2,022,000
|
)
|
||||
|
$ | - |
$
|
-
|
$
|
-
|
F-23
The
provision for income taxes differs from the amount of income tax determined
by
applying the applicable U.S. statutory federal income tax rate to the pretax
loss from continuing operations as a result of the following
differences:
2007
|
2006
|
2005
|
||||||||
Tax
at the U.S. statutory rate
|
$
|
(9,474,000
|
)
|
$
|
(2,456,000
|
)
|
$
|
(811,000
|
)
|
|
Stock
option exercises
|
(363,000
|
)
|
-
|
-
|
||||||
Tax
credits
|
(477,000
|
)
|
-
|
-
|
||||||
Valuation
allowance
|
10,308,000
|
2,456,000
|
811,000
|
|||||||
Other
|
6,000
|
-
|
-
|
|||||||
|
$ | - |
$
|
-
|
$
|
-
|
At
December 31, 2007, the Company has federal net operating loss carryforwards
of
approximately $34,262,000, which begin to expire if not utilized by 2023, and
approximately $584,000 of tax credit carryforwards that begin to expire if
not
utilized by 2024. The utilization of approximately $2,768,000 of the net
operating loss carryforwards and approximately $203,000 of the tax credits
carryforwards is limited through 2011 as a result of ownership changes. The
Company also has state net operating loss carryforwards of approximately
$22,176,000, which will expire if not utilized by 2027, and state tax credit
carryforwards of approximately $153,000, which begin to expire if not utilized
by 2010.
The
Company files a United States federal tax return, along with various state
and
local income tax returns. The federal, state and local tax returns for the
years
ended December 31, 2006, 2005 and 2004 are still open for
examination.
Effective
January 1, 2008, the Company adopted Financial Accounting Standards Board FIN
48, “Accounting for Uncertainty in Income Taxes”, which prescribes a minimum
recognition threshold and measurement methodology that a tax position taken
or
expected to be taken in a tax return is required to meet before being recognized
in the financial statements. There was no impact to the financial statements
upon the adoption of FIN 48.
F-24
The
following presents a rollforward of the unrecognized tax benefits and associated
interest and penalties:
Unrecognized
Tax Benefits
|
Interest
and Penalties
|
||||||
Balance
at January 1, 2007
|
$
|
-
|
$
|
-
|
|||
Prior
year tax positions
|
-
|
-
|
|||||
Current
year tax positions
|
-
|
-
|
|||||
Deferred
tax positions
|
230,000
|
-
|
|||||
Settlements
with tax authorities
|
-
|
-
|
|||||
Expiration
of the statute of limitations
|
-
|
-
|
|||||
Balance
at December 31, 2007
|
$
|
230,000
|
$
|
-
|
Note
6. Other Balance Sheet Details
Available-For-Sale
Cash Equivalents and Marketable Securities
Available-for-sale
Marketable Securities consist of the following:
Cost
|
Accrued
Interest
|
Gross
Unrealized
Gains
|
Gross
Unrealized
Losses
|
Fair
Value
|
||||||||||||
December
31, 2007 - Current Marketable Securities
|
$
|
1,000,000
|
$
|
17,881
|
$
|
-
|
$
|
-
|
$
|
1,017,881
|
The
Company considers investments with a maturity date of more than three months
from the date of purchase to be short-term investments and has classified these
securities as available-for-sale. Such investments are carried at fair value,
with unrealized gains and losses included as accumulated other comprehensive
income (loss) in stockholders’ equity. The cost of available-for-sale securities
sold is determined based on the specific identification method.As
a
result of changes in market interest rates on investment, the Company recognized
unrealized gains/(losses) of $4,165 $13,645, and ($17,810) for the years ending
December 31, 2007, 2006, and 2005, respectively. In 2007, the Company recaptured
$4,165 of the previously recorded other comprehensive loss due to the maturity
of the short term investments.
F-25
Equipment
Equipment
consists of the following:
2007
|
2006
|
||||||
Laboratory
Equipment
|
$
|
966,517
|
$
|
347,944
|
|||
Computer
Equipment
|
258,089
|
132,572
|
|||||
Furniture
|
274,903
|
65,087
|
|||||
1,499,509
|
545,603
|
||||||
Less
accumulated depreciation
|
(313,489
|
)
|
(142,011
|
)
|
|||
$
|
1,186,020
|
$
|
403,592
|
Note
7. Commitments and Contingencies
The
Company has entered into various agreements with third
parties
in connection with the research and development activities of its existing
product candidates as well as discovery efforts on potential new product
candidates. These agreements include costs for research and development and
license agreements that represent the Company’s fixed obligations payable to
sponsor research and minimum royalty payments for licensed patents. These
amounts do not include any additional amounts that the Company may be required
to pay under its license agreements upon the achievement of scientific,
regulatory and commercial milestones that may become payable depending on the
progress of scientific development and regulatory approvals, including
milestones such as the submission of an investigational new drug application
to
the FDA, similar submissions to foreign regulatory authorities and the first
commercial sale of the Company’s products in various countries. These agreements
include costs related to manufacturing, clinical trials and preclinical studies
performed by third parties.
The
Company is also party to three agreements that require it to make milestone
payments, royalties on net sales of the Company’s products and payments on
sublicense income received by the Company. As of December 31, 2007, $300,000
of
milestone payments have been recognized and paid.
From
time
to time, the Company may have certain contingent liabilities that arise in
the
ordinary course of business. The Company accrues for liabilities when it is
probable that future expenditures will be made and such expenditures can be
reasonably estimated. For all periods presented, the Company is not a party
to
any pending material litigation or other material legal
proceedings.
The
Company currently has operating lease commitments in place for facilities in
Buffalo, New York and Rosemont,
Illinois as well as office equipment. The Company recognizes rent expense on
a
straight-line basis over the term of the related operating leases. The operating
lease expenses recognized were $218,635, $160,742, and $112,967 in 2007, 2006
and 2005, respectively.
F-26
Annual
future minimum lease payments under present lease commitments are as
follows:
Operating
Leases
|
||||
2008
|
$
|
333,566
|
||
2009
|
349,878
|
|||
2010
|
389,940
|
|||
2011
|
360,300
|
|||
2012
|
174,726
|
|||
$
|
1,608,410
|
The
Company has entered into stock option agreements with key employees, board
members and consultants with exercise prices ranging from $0.66 to $17.00.
These
awards were approved by the Company’s Board of Directors. The options expire ten
years from the date of grant, subject to the terms applicable in the
agreement.
The
following table summarizes the stock option activity for the years ended
December 31, 2007 and 2006:
Number of
Options
|
Weighted Average
Exercise Price
|
||||||
Outstanding
at December 31, 2005
|
324,240
|
$
|
0.82
|
||||
Granted
|
161,750
|
$
|
4.92
|
||||
Exercised
|
625
|
$
|
4.50
|
||||
Forfeited
|
1,875
|
$
|
4.50
|
||||
Outstanding
at December 31, 2006
|
483,490
|
$
|
2.17
|
||||
Granted
|
660,000
|
$
|
9.85
|
||||
Exercised
|
131,750
|
$
|
1.34
|
||||
Forfeited
|
-
|
n/a
|
|||||
Outstanding
at December 31, 2007
|
1,011,740
|
$
|
7.29
|
F-27
The
number of options and weighted average exercise price of options fully vested
and exercisable for the years ending December 31, 2007, 2006 and 2005 were
646,930, 243,183, and 83,560 options at $6.89, $2.27, and $0.88 respectively.
A
table showing the number of options outstanding and exercisable (fully vested)
at December 31, 2007 appears below:
|
Outstanding
|
Exercisable
|
|||||||||
Exercise Price
|
Number of
Options
|
Weighted
Average
Years to
Expiration
|
Number of
Options
|
||||||||
$
|
0.66
|
112,500
|
7.50
|
65,000
|
|||||||
$
|
0.67
|
77,740
|
7.50
|
51,680
|
|||||||
$
|
2.00
|
10,000
|
7.92
|
10,000
|
|||||||
$
|
4.50
|
111,500
|
8.17
|
54,375
|
|||||||
$
|
6.00
|
45,000
|
8.55
|
45,000
|
|||||||
$
|
8.36
|
152,500
|
9.26
|
152,500
|
|||||||
$
|
8.82
|
20,000
|
9.22
|
20,000
|
|||||||
$
|
9.14
|
94,500
|
9.12
|
24,375
|
|||||||
$
|
9.40
|
140,000
|
9.45
|
140,000
|
|||||||
$
|
9.93
|
|
30,000
|
9.46
|
15,000
|
||||||
$
|
10.00
|
117,000
|
9.93
|
48,000
|
|||||||
$
|
10.61
|
|
18,000
|
5.00
|
18,000
|
||||||
$
|
10.84
|
3,000
|
9.47
|
3,000
|
|||||||
$
|
11.00
|
25,000
|
9.46
|
-
|
|||||||
$
|
14.00
|
25,000
|
9.46
|
-
|
|||||||
$
|
17.00
|
30,000
|
9.46
|
-
|
|||||||
Total
|
1,011,740
|
8.80
|
646,930
|
F-28
The
Company has entered into warrant agreements with strategic partners, consultants
and investors with exercise prices ranging from $1.13 to $11.00. These awards
were approved by the Company’s Board of Directors. The warrants expire between
five and six years from the date of grant, subject to the terms applicable
in
the agreement. A list of the total warrants awarded and exercised appears
below:
Number of
Warrants
|
Weighted Average
Exercise Price
|
||||||
Outstanding
at December 31, 2005
|
594,424
|
$
|
1.61
|
||||
Granted
|
220,000
|
$
|
8.09
|
||||
Exercised
|
-
|
N/A
|
|||||
Forfeited
|
-
|
N/A
|
|||||
Outstanding
at December 31, 2006
|
814,424
|
$
|
3.36
|
||||
Granted
|
2,687,602
|
$
|
10.40
|
||||
Exercised
|
48,758
|
$
|
2.00
|
||||
Forfeited
|
-
|
N/A
|
|||||
Outstanding
at December 31, 2007
|
3,453,268
|
$
|
8.86
|
The
Company has entered into employment agreements with
three
key
executives who, if terminated by the Company without cause as described in
these
agreements, would be entitled to severance pay.
The
Company is not currently a party to any pending legal actions. From time to
time
in the ordinary course of business, the Company may be subject to claims brought
against it. It is not possible to state the ultimate liability, if any, in
these
matters.
Note
8. Subsequent Events
In
December 2007, the Company accrued $2,687,355 of non-cash, stock-based
compensation related to the stock options awarded under the 2007 Executive
Compensation Plan. This accrual was based on the closing price of $8.80 of
the
Company’s stock as listed on the NASDAQ Global Market at December 31, 2007. The
stock options were awarded by the Compensation Committee on February 4, 2008.
At
the time the options were awarded, the grant date fair value of the options
was
$1,137,345 based on the exercise price of $4.00 as determined by the 2006 Equity
Incentive Plan at the time of the award. This results in a recapture of
$1,550,010 of non-cash, stock-based compensation expense in the first quarter
of
2008.
F-29
Item 9.
Changes In and Disagreements with Accountants on Accounting and Financial
Disclosure
There
were no disagreements with accountants on accounting and financial disclosures
for the years ended, December 31, 2007 and 2006.
Item 9A.
Controls and Procedures
Effectiveness
of Disclosure
Our
management, with the participation of our Chief Executive Officer and Chief
Financial Officer, evaluated the effectiveness of our disclosure controls and
procedures as of December 31, 2007 as defined in Rules 13a-15(e) and
15d-15(e) under the Securities Exchange Act of 1934, as amended, or the Exchange
Act. Our management recognizes that any controls and procedures, no matter
how
well designed and operated, can provide only reasonable assurance of achieving
their objectives and management necessarily applies its judgment in evaluating
the cost-benefit relationship of possible controls and procedures. Based on
the
evaluation of our disclosure controls and procedures as of December 31, 2007,
our Chief Executive Officer and Chief Financial Officer concluded that, as
of
such date, our disclosure controls and procedures were effective to assure
that
information required to be declared by us in reports that we file or submit
under the Exchange Act is (1) recorded, processed, summarized, and reported
within the periods specified in the SEC’s rules and forms and (2) accumulated
and communicated to our management, including our Chief Executive Officer and
Chief Financial Officer, as appropriate, to allow timely decisions regarding
required disclosure.
Changes
in Internal Control over Financial Reporting
Our
management is responsible for establishing and maintaining adequate internal
control over financial reporting, as such term is defined in Exchange Act Rules
13a-15(f). Under the supervision and with the participation of management,
including our principal executive officer and principle financial officer,
we
conducted an evaluation of the effectiveness of our internal control over
financial reporting based on the framework in Internal
Control – Integrated Framework
issued
by the Committee of Sponsoring Organizations of the Treadway Commission. Based
on our evaluation under the framework in Internal
Control – Integrated Framework,
our
management concluded that our internal control over financial reporting was
effective as of December 31, 2007.
This
annual report does not include an attestation report of the Company’s registered
public accounting firm regarding internal control over financial reporting.
Management’s report was not subject to attestation by the Company’s registered
public accounting firm pursuant to temporary rules of the Securities and
Exchange Commission that permit the Company to provide only management’s report
in this annual report on Form 10-K.
There
was
no change in our internal control over financial reporting during our fourth
fiscal quarter ended December 31, 2007 that has materially affected, or is
reasonably likely to materially affect, our internal control over financial
reporting.
Item 9B.
Other Information
Not
applicable.
33
PART
III
Pursuant
to General Instruction G(3) of Form 10-K, Items 10 through 14, inclusive, have
not been restated or answered in this annual report on Form 10-K because the
Company intends to file within 120 days after the close of its fiscal year
with
the Securities and Exchange Commission a definitive proxy statement pursuant
to
Regulation 14A under the Securities Exchange Act of 1934, which proxy statement
involves the election of directors. The information required in these Items
10
through 14, inclusive, is incorporated by reference to that proxy statement.
PART
IV
Item 15.
Exhibits and Financial Statement Schedules
(a)
The
following financial statements and supplementary data are filed as a part of
this annual report on Form 10-K under Item 8.
Report
of
Independent Registered Public Accounting Firm
Balance
Sheets at December 31, 2007 and 2006
Statements
of Operations for years ended December 31, 2007, 2006, and 2005
Statements
of Stockholders’ Equity for period from January 1, 2005 to December 31,
2007
Statements
of Cash Flows for years ended December 31, 2007, 2006, and 2005
Notes
to
Financial Statements
(b)
The
following exhibits are incorporated herein by reference.
Exhibit
No.
|
|
Description
|
3.1
|
|
Certificate
of Incorporation filed with the Secretary of State of Delaware on
June 5,
2003***
|
|
|
|
3.2
|
|
Certificate
of Amendment of Certificate of Incorporation filed with the Secretary
of
State of Delaware on February 25, 2005***
|
|
|
|
3.3
|
|
Certificate
of Designation of Series A Participating Convertible Preferred Stock
filed
with the Secretary of State of Delaware on March 8,
2005***
|
|
|
|
3.4
|
|
Second
Certificate of Amendment of Certificate of Incorporation filed with
Secretary of State of Delaware on June 30, 2006***
|
|
|
|
3.5
|
|
Certificate
of Designations, Preferences and Rights of Series B Convertible Preferred
Stock, dated March 16, 2007******
|
|
|
|
3.6
|
|
Second
Amended and Restated By-Laws*******
|
|
|
|
4.1
|
|
Form
of Specimen Common Stock
Certificate*
|
|
|
|
4.2
|
|
Form
of Warrants issues to designees of Sunrise Securities Corp., dated
March
2005*
|
34
4.3
|
|
Form
of Warrants issued to underwriters***
|
|
|
|
4.4
|
|
Warrant
to Purchase Common Stock issued to ChemBridge Corporation, dated
April 27,
2004*
|
|
|
|
4.5
|
|
Form
of Series B Warrant ******
|
|
|
|
4.6
|
|
Form
of Series C Warrant ******
|
|
|
|
10.1
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Michael Fonstein,
dated as of July 5, 2003*
|
|
|
|
10.2
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Yakov Kogan,
dated as
of July 5, 2003*
|
|
|
|
10.3
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Andrei Gudkov,
dated
as of July 5, 2003*
|
|
|
|
10.4
|
|
Library
Access Agreement by and between ChemBridge Corporation and Cleveland
BioLabs, Inc., effective as of April 27, 2004*
|
|
|
|
10.5
|
|
Restricted
Stock and Investor Rights Agreement between Cleveland BioLabs, Inc.
and
ChemBridge Corporation, dated as of April 27, 2004*
|
|
|
|
10.6
|
|
Common
Stockholders Agreement by and among Cleveland BioLabs, Inc. and the
stockholders named therein, dated as of July 1, 2004*
|
|
|
|
10.7
|
|
Exclusive
License Agreement by and between The Cleveland Clinic Foundation
and
Cleveland BioLabs, Inc., effective as of July 1, 2004*
|
|
|
|
10.8
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Michael
Fonstein,
dated August 1, 2004*
|
|
|
|
10.9
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Yakov Kogan,
dated August 1, 2004*
|
|
|
|
10.10
|
|
Consulting
Agreement between Cleveland BioLabs, Inc. and Dr. Andrei Gudkov,
dated
August 1, 2004*
|
|
|
|
10.11
|
|
Cooperative
Research and Development Agreement by and between the Uniformed Services
University of the Health Sciences, the Henry M. Jackson Foundation
for the
Advancement of Military Medicine, Inc., the Cleveland Clinic Foundation,
and Cleveland BioLabs, Inc., dated as of August 1,
2004**
|
|
|
|
10.12
|
|
Form
of Stock Purchase Agreement between Cleveland BioLabs, Inc. and the
Purchasers party thereto, dated as of March 15,
2005*
|
35
10.13
|
|
Form
of Series A Rights Agreement by and among Cleveland BioLabs, Inc.
and the
parties thereto, dated as of March 15, 2005*
|
|
|
|
10.14
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Farrel Fort,
dated June 1, 2005*
|
|
|
|
10.15
|
|
Amendment
to Employment Agreement by and between Cleveland BioLabs, Inc. and
Dr.
Farrel Fort, dated September 30, 2005*
|
|
|
|
10.16
|
|
Amendment
to Consulting Agreement between Cleveland BioLabs, Inc. and Dr. Andrei
Gudkov, dated as of January 23, 2006*
|
|
|
|
10.17
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Michael
Fonstein, dated as of January 23, 2006*
|
|
|
|
10.18
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Yakov
Kogan, dated as of January 23, 2006*
|
|
|
|
10.19
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Andrei
Gudkov, dated as of January 23, 2006*
|
|
|
|
10.20
|
|
Amendment
to Common Stockholders Agreement by and among Cleveland BioLabs,
Inc. and
the parties thereto, dated as of January 26, 2006*
|
|
|
|
10.21
|
|
Form
of Amendment to Series A Rights Agreement by and among Cleveland
BioLabs,
Inc. and the parties thereto, dated as of February 17,
2006*
|
|
|
|
10.22
|
|
Cleveland
BioLabs, Inc. 2006 Equity Incentive Plan***
|
|
|
|
10.23
|
|
Process
Development and Manufacturing Agreement between Cleveland BioLabs,
Inc.
and SynCo Bio Partners B.V., effective as of August 31,
2006****
|
|
|
|
10.24
|
|
Sponsored
Research Agreement between Cleveland BioLabs, Inc. and Roswell Park
Cancer
Institute Corporation, effective as of January 12,
2007*****
|
|
|
|
10.25
|
|
Securities
Purchase Agreement, dated March 16, 2007******
|
|
|
|
10.26
|
|
Registration
Rights Agreement, dated March 16, 2007******
|
|
|
|
23.1
|
|
Consent
of Meaden & Moore, Ltd.
|
|
|
|
31.1
|
|
Rule
13a-14(a)/15d-14(a) Certification of Michael
Fonstein
|
36
31.2
|
|
Rule
13a-14(a)/15d-14(a) Certification of John A. Marhofer,
Jr.
|
|
|
|
32.1
|
|
Section
1350 Certification.
|
*
|
Incorporated
by reference to Amendment No. 1 to Registration Statement on Form
SB-2 as
filed on April 25, 2006 (File No. 333-131918).
|
|
|
**
|
Incorporated
by reference to Amendment No. 2 to Registration Statement on Form
SB-2 as
filed on May 31, 2006 (File No. 333-131918).
|
|
|
***
|
Incorporated
by reference to Amendment No. 3 to Registration Statement on Form
SB-2 as
filed on July 10, 2006 (File No. 333-131918).
|
|
|
****
|
Incorporated
by reference to Form 8-K as filed on October 25, 2006.
|
|
|
*****
|
Incorporated
by reference to Form 8-K as filed on January 12, 2007.
|
|
|
******
|
Incorporated
by reference to Form 8-K as filed on March 19, 2007.
|
*******
|
Incorporated
by reference to Form 8-K as filed on December 5,
2007.
|
(c)
Not
applicable.
SIGNATURES
Pursuant
to the
requirements of Section 13 or 15(d) of the Securities Exchange Act of 1934,
the
Registrant has caused this report to be signed on its behalf by the undersigned
thereunto duly authorized.
|
CLEVELAND
BIOLABS, INC.
|
|
Dated:
March 21, 2008
|
By:
|
/s/
MICHAEL FONSTEIN
|
|
Michael
Fonstein
|
|
|
Chief
Executive Officer
|
|
|
(Principal
Executive Officer)
|
|
CLEVELAND
BIOLABS, INC.
|
||
Dated:
March 21, 2008
|
By:
|
/s/
JOHN A. MARHOFER, JR.
|
John
A. Marhofer, Jr.
|
||
Chief
Financial Officer
|
||
(Principal
Financial and Accounting Officer)
|
37
Pursuant
to the requirements of the Securities Exchange Act of 1934, this report has
been
signed by the following persons in the capacities and on the dates
indicated.
Signature
|
Title
|
Date
|
||
/
S
/ Michael Fonstein
Michael
Fonstein
|
Chief
Executive Officer, President, and
Director (Principal Executive
Officer)
|
March
21, 2008
|
||
/
S
/ John A. Marhofer, Jr.
John
A. Marhofer, Jr.
|
Chief
Financial Officer (Principal
Financial and Accounting Officer)
|
March
21, 2008
|
||
/
S
/ James Antal
James
Antal
|
Director
|
March
21, 2008
|
||
/
S
/ Paul DiCorleto
Paul
DiCorleto
|
Director
|
March
21, 2008
|
||
/
S
/ Andrei Gudkov
Andrei
Gudkov
|
Chief
Scientific Officer, and Director
|
March
21, 2008
|
||
/
S
/ Bernard L. Kasten
Bernard
L. Kasten
|
Director
|
March
21, 2008
|
||
/
S
/ Yakov Kogan
Yakov
Kogan
|
Chief
Operating Officer, Secretary, and
Director
|
March
21, 2008
|
||
/
S
/ H. Daniel Perez
H.
Daniel Perez
|
Director
|
March
21, 2008
|
38
EXHIBIT
INDEX
Exhibit
No.
|
|
Description
|
3.1
|
|
Certificate
of Incorporation filed with the Secretary of State of Delaware on
June 5,
2003***
|
|
|
|
3.2
|
|
Certificate
of Amendment of Certificate of Incorporation filed with the Secretary
of
State of Delaware on February 25, 2005***
|
|
|
|
3.3
|
|
Certificate
of Designation of Series A Participating Convertible Preferred Stock
filed
with the Secretary of State of Delaware on March 8,
2005***
|
|
|
|
3.4
|
|
Second
Certificate of Amendment of Certificate of Incorporation filed with
Secretary of State of Delaware on June 30, 2006***
|
|
|
|
3.5
|
|
Certificate
of Designations, Preferences and Rights of Series B Convertible Preferred
Stock, dated March 16, 2007******
|
|
|
|
3.6
|
|
Second
Amended and Restated By-Laws*******
|
|
|
|
4.1
|
|
Form
of Specimen Common Stock Certificate*
|
|
|
|
4.2
|
|
Form
of Warrants issues to designees of Sunrise Securities Corp., dated
March
2005*
|
|
|
|
4.3
|
|
Form
of Warrants issued to underwriters***
|
|
|
|
4.4
|
|
Warrant
to Purchase Common Stock issued to ChemBridge Corporation, dated
April 27,
2004*
|
|
|
|
4.5
|
|
Form
of Series B Warrant ******
|
|
|
|
4.6
|
|
Form
of Series C Warrant ******
|
|
|
|
10.1
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Michael Fonstein,
dated as of July 5, 2003*
|
|
|
|
10.2
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Yakov Kogan,
dated as
of July 5, 2003*
|
|
|
|
10.3
|
|
Restricted
Stock Agreement between Cleveland BioLabs, Inc. and Andrei Gudkov,
dated
as of July 5, 2003*
|
|
|
|
10.4
|
|
Library
Access Agreement by and between ChemBridge Corporation and Cleveland
BioLabs, Inc., effective as of April 27,
2004*
|
39
10.5
|
|
Restricted
Stock and Investor Rights Agreement between Cleveland BioLabs, Inc.
and
ChemBridge Corporation, dated as of April 27, 2004*
|
|
|
|
10.6
|
|
Common
Stockholders Agreement by and among Cleveland BioLabs, Inc. and the
stockholders named therein, dated as of July 1, 2004*
|
|
|
|
10.7
|
|
Exclusive
License Agreement by and between The Cleveland Clinic Foundation
and
Cleveland BioLabs, Inc., effective as of July 1, 2004*
|
|
|
|
10.8
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Michael
Fonstein,
dated August 1, 2004*
|
|
|
|
10.9
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Yakov Kogan,
dated August 1, 2004*
|
|
|
|
10.10
|
|
Consulting
Agreement between Cleveland BioLabs, Inc. and Dr. Andrei Gudkov,
dated
August 1, 2004*
|
|
|
|
10.11
|
|
Cooperative
Research and Development Agreement by and between the Uniformed Services
University of the Health Sciences, the Henry M. Jackson Foundation
for the
Advancement of Military Medicine, Inc., the Cleveland Clinic Foundation,
and Cleveland BioLabs, Inc., dated as of August 1,
2004**
|
|
|
|
10.12
|
|
Form
of Stock Purchase Agreement between Cleveland BioLabs, Inc. and the
Purchasers party thereto, dated as of March 15, 2005*
|
|
|
|
10.13
|
|
Form
of Series A Rights Agreement by and among Cleveland BioLabs, Inc.
and the
parties thereto, dated as of March 15, 2005*
|
|
|
|
10.14
|
|
Employment
Agreement by and between Cleveland BioLabs, Inc. and Dr. Farrel Fort,
dated June 1, 2005*
|
|
|
|
10.15
|
|
Amendment
to Employment Agreement by and between Cleveland BioLabs, Inc. and Dr.
Farrel Fort, dated September 30, 2005*
|
|
|
|
10.16
|
|
Amendment
to Consulting Agreement between Cleveland BioLabs, Inc. and Dr. Andrei
Gudkov, dated as of January 23, 2006*
|
|
|
|
10.17
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Michael
Fonstein, dated as of January 23, 2006*
|
|
|
|
10.18
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Yakov
Kogan, dated as of January 23,
2006*
|
40
10.19
|
|
Amendment
to Restricted Stock Agreement between Cleveland BioLabs, Inc. and
Andrei
Gudkov, dated as of January 23, 2006*
|
|
|
|
10.20
|
|
Amendment
to Common Stockholders Agreement by and among Cleveland BioLabs,
Inc. and
the parties thereto, dated as of January 26, 2006*
|
|
|
|
10.21
|
|
Form
of Amendment to Series A Rights Agreement by and among Cleveland
BioLabs,
Inc. and the parties thereto, dated as of February 17,
2006*
|
|
|
|
10.22
|
|
Cleveland
BioLabs, Inc. 2006 Equity Incentive Plan***
|
|
|
|
10.23
|
|
Process
Development and Manufacturing Agreement between Cleveland BioLabs,
Inc.
and SynCo Bio Partners B.V., effective as of August 31,
2006****
|
|
|
|
10.24
|
|
Sponsored
Research Agreement between Cleveland BioLabs, Inc. and Roswell Park
Cancer
Institute Corporation, effective as of January 12,
2007*****
|
|
|
|
10.25
|
|
Securities
Purchase Agreement, dated March 16, 2007******
|
|
|
|
10.26
|
|
Registration
Rights Agreement, dated March 16, 2007******
|
|
|
|
23.1
|
|
Consent
of Meaden & Moore, Ltd.
|
|
|
|
31.1
|
|
Rule
13a-14(a)/15d-14(a) Certification of Michael Fonstein
|
|
|
|
31.2
|
|
Rule
13a-14(a)/15d-14(a) Certification of John A. Marhofer,
Jr.
|
|
|
|
32.1
|
|
Section
1350 Certification.
|
*
|
Incorporated
by reference to Amendment No. 1 to Registration Statement on Form
SB-2 as
filed on April 25, 2006 (File No. 333-131918).
|
|
|
**
|
Incorporated
by reference to Amendment No. 2 to Registration Statement on Form
SB-2 as
filed on May 31, 2006 (File No. 333-131918).
|
|
|
***
|
Incorporated
by reference to Amendment No. 3 to Registration Statement on Form
SB-2 as
filed on July 10, 2006 (File No. 333-131918).
|
|
|
****
|
Incorporated
by reference to Form 8-K as filed on October 25, 2006.
|
|
|
*****
|
Incorporated
by reference to Form 8-K as filed on January 12, 2007.
|
|
|
******
|
Incorporated
by reference to Form 8-K as filed on March 19, 2007.
|
*******
|
Incorporated
by reference to Form 8-K as filed on December 5,
2007.
|
41