Disclosures Regarding Forward-Looking Statements





The following should be read in conjunction with the unaudited condensed
consolidated financial statements and the related notes that appear elsewhere in
this report as well as in conjunction with the Risk Factors section in our
Annual Report on Form 10-K for the year ended September 30, 2019 as filed with
the United States Securities and Exchange Commission ("SEC") on January 10,
2020. This report and our Form 10-K include forward-looking statements made
based on current management expectations pursuant to the safe harbor provisions
of the Private Securities Litigation Reform Act of 1995, as amended.



This report includes "forward-looking statements" within the meaning of
Section 21E of the Exchange Act. Those statements include statements regarding
the intent, belief or current expectations of the Company and its subsidiaries
and our management team. Any such forward-looking statements are not guarantees
of future performance and involve risks and uncertainties, and actual results
may differ materially from those projected in the forward-looking statements.
These risks and uncertainties include but are not limited to those risks and
uncertainties set forth in Item 1A - Risk Factors of this Quarterly Report and
in Item 1A - Risk Factors of our Annual Report. In light of the significant
risks and uncertainties inherent in the forward-looking statements included in
this Quarterly Report on Form 10-Q and in our Annual Report on Form 10-K, the
inclusion of such statements should not be regarded as a representation by us or
any other person that our objectives and plans will be achieved. Further, these
forward-looking statements reflect our view only as of the date of this report.
Except as required by law, we undertake no obligations to update any
forward-looking statements and we disclaim any intent to update forward-looking
statements after the date of this report to reflect subsequent developments.
Accordingly, you should also carefully consider the factors set forth in other
reports or documents that we file from time to time with the SEC.



Overview


Restatement of Previously Issued Unaudited Financial Statements


We have restated certain previously reported financial information for the three
months ended December 31, 2018 in this Item 2, Management's Discussion and
Analysis of Financial Condition and Results of Operations, including but not
limited to information within the Results of Operations section.



See Note 2, Significant Accounting Policies-Restatement of Previously Issued
Unaudited Financial Statements, in Item 1, Financial Statements, for additional
information related to the restatement, including descriptions of the
misstatements and the impacts on our unaudited condensed consolidated financial
statements.



Description of the Company



We are a biotechnology company focused on developing next generation therapies
to treat rare genetic diseases and cancer caused by mutant genes. Given that
perhaps every human disease has a genetic component, we believe that our
differentiated platform technology has the potential for broad impact.



Mutated proteins resulting from errors in deoxyribonucleic acid ("DNA")
sequences cause many rare genetic diseases and cancer. DNA in each cell of the
body is transcribed into pre-mRNA, which is then processed (spliced) into mRNA
which is exported into the cytoplasm of the cell and translated into protein.
This is termed the "central dogma" of biology. Therefore, when errors in a DNA
sequence occur, they are propagated to RNAs and can become a damaging protein.



The type of therapies that we are developing are termed antisense
oligonucleotide ("ASO") therapies. ASOs are short single strands of nucleic
acids (traditionally thought of as single stranded RNA molecules) which will
bind to defective RNA targets in cells and inhibit their ability to be
translated into defective proteins. We believe we are a leader in the discovery
and development of the class of RNA-targeted ASO drugs called peptide nucleic
acids ("PNAs"). Our proprietary PATrOL™ platform allows for a more efficient
discovery of drug product candidates, potentially transforming the treatment
paradigm for people affected by rare genetic diseases and cancer.



The PATrOL™ platform allows for a potentially more efficient discovery of drug
product candidates because of manufacturing consistency and because we are not
constrained by folded regions of the target RNA molecule (secondary structures).
The peptide backbone of our ASOs is rigid, and once linked together to form a
series of backbone subunits, forms a single pre-organized structure.



                                       18





At a more detailed level, each subunit of the peptide backbone has only a single
chiral center - a point in the chemical structure where the conformation of the
backbone could fluctuate - and this chiral center is locked into one
conformation and thus pre-organized to form only a single conformation or
stereoisomer. A stereoisomer is a term used in the ASO therapeutics field to
mean a string of backbone subunits with nucleo-bases attached that are linked
together into a specific sequence that matches (complements) the target
sequence, but because of the nature of the backbone subunits used, the drug
assumes various conformations often with varying affinity for the target
sequence. These stereoisomers often require a manufacturing step to purify the
heterogeneous mixture of conformations into a more homogenous mixture or even a
single conformation of the drug in order to obtain the hoped-for therapeutic
effect. Our PNAs assume only a single conformation with any constellation of
nucleo-bases added to the backbone or any oligomer length. This backbone also
has a neutral charge, as opposed to the negatively charged backbones of DNA and
RNA. This neutral charge allows our ASO to open up RNAs which are folded upon
themselves and bind to their target sequence. This potentially accelerates
identification of drug candidates which have the desired activity.



In addition to the backbone conformational purity which allows for a more
efficient discovery of drug product candidates, we also have a kit of
proprietary bi-facial (also known as bi-specific) nucleotides (traditional
nucleotides only have a single binding face and thus are restricted to only
binding single-stranded RNA targets) which can be used in any combination to
access RNA secondary structures (double stranded RNA targets which are folded
upon themselves) such as hairpins. This allows us to potentially access regions
of the target transcript which may be unique in secondary structure to allow
enhanced selectivity for the target (mutant) RNA vs. the normal RNA. Enhanced
selectivity for mutant RNAs vs. normal RNAs is critical as normal RNAs are
likely required for effective functioning of the cell. These bi-specific
nucleotides can also target genomic loci and microRNAs in their double-stranded
form.


In addition to the backbone and modified nuclear bases, the platform toolkit also includes linker technology which, when added to both ends of the PNAs, allow cooperative binding between individual drug molecules once they are engaged with the target RNA to form longer and more tightly bound drugs.


The final component of the platform is a chemical moiety, which is used to
decorate the peptide backbone in a proprietary manner and allows the PNAs to
penetrate cell membranes and distribute throughout the body when administered
systemically.


This relatively simple toolkit of components forms the PATrOL™ platform and allows us to manufacture genome and transcript-specific PNAs quickly for screening.


We are currently focused on therapeutic areas in which we believe our drugs will
provide the greatest benefit with a significant market opportunity and intend to
utilize our technology to build out a pipeline of custom designed therapeutics
for additional high-value disease targets. We are developing several preclinical
programs using our PATrOL™ platform, including: NT0100 program, targeted at
Huntington's Disease ("HD"), a repeat expansion disorder, and the NT0200
program, targeted at myotonic dystrophy Type 1 ("DM1"). Preclinical studies are
being conducted to evaluate the PATrOL™ platform technology and program
candidates in the areas of pharmacokinetics and pharmacodynamics, and we expect
to report results from those studies in the first calendar quarter and the
second calendar quarter of 2020. In addition, the emerging pipeline of other
assets that target primary and secondary RNA structure and genomic DNA allows a
unique market advantage across a variety of rare diseases and oncology targets.



Using our PATrOL™ platform, we believe we can create ASOs that have distinct
potential advantages over other chemical entities currently in the market or in
development for gene silencing applications. These advantages include, among
others: a backbone that has only one chiral center and thus forms only one
stereoisomer; the ability of the PNA backbone to invade, open up secondary (RNA
folded upon itself) and tertiary structures (RNA molecules that interact with
other RNA molecules in the cell) and bind within these double-stranded RNA in a
highly selective manner; a proprietary set of engineered nucleo-bases that
increase selectivity to specific target sequences including secondary and
tertiary structures that has been licensed exclusively from Carnegie Mellon
University ("CMU"); technology to allow self-assembly of our small PNAs at the
RNA target to increase selectivity which has been licensed exclusively from CMU;
the ability to modulate cell permeability and be broadly distributed throughout
the body; the lack of innate or acquired immune responses of similar PNAs in
preclinical models; and potential minimal toxicity based on previous in-vivo
studies in rodent models. With these advantages, our PATrOL™ platform-enabled
therapies can potentially address a multitude of rare genetic diseases and
cancer, among other indications.



Product Pipeline



Huntington's Disease



HD is a devastating rare neurodegenerative disorder. After onset, symptoms such
as uncontrolled movements, cognitive impairments and emotional disturbances
worsen over time. HD is caused by toxic aggregation of mutant huntingtin
protein, leading to progressive neuron loss in the striatum and cortex of the
brain. The wildtype huntingtin gene (HTT) has a region in which a three-base DNA
sequence, CAG, is repeated many times. When the DNA sequence CAG is repeated 26
or fewer times in this region, the resulting protein behaves normally. While the
wildtype function of HTT is largely uncharacterized, the protein is known to be
essential for normal brain development. When the DNA sequence CAG is repeated 40
times or more in this region, the resulting protein becomes toxic and causes HD.
Every person has two copies, or alleles, of the HTT. Only one of the alleles
(the "mutant" allele) needs to bear at least 40 CAG repeats for HD to occur. HD
is one of many known repeat expansion disorders, which are a set of genetic
disorders caused by a mutation that leads to a repeat of nucleotides exceeding
the normal threshold. Current therapies for patients with HD can only manage
individual symptoms. There is no approved therapy that has been shown to delay
or halt disease progression. There are approximately 30,000 symptomatic patients
in the U.S. and more than 200,000 at-risk of inheriting the disease.



                                       19




NT0100 Program - PATrOL™ Enabled PNA for Huntington's Disease

The PATrOL™ platform has the potential to address many dominantly inherited genetic diseases. We will be initially focused on HD, a fatal rare genetic repeat expansion disorder with no viable treatment options.


One especially important advantage of the PATrOL™ platform that makes it
promising for the treatment of repeat expansion disorders like HD is the ability
of our small ASOs to potentially self-assemble within an RNA hairpin. As the
number of repeats increases, the PATrOL™ oligonucleotides bind more tightly to
each other and the mutant RNA. This allows our therapies to potentially
inactivate mutant HTT mRNA before it can be translated into harmful protein via
selective binding to the expanded CAG repeats while leaving the normal HTT mRNA
largely unbound to drug and producing functional protein. Achieving mutant
allele selectivity would be a key advantage for any RNA-based approach aiming to
treat HD. The PATrOL™-enabled NT0100 program is currently in preclinical
development for the treatment of HD.



NT0200 Program - PATrOL™ Enabled PNA for Myotonic Dystrophy Type 1





Our pipeline also contains a second near-term, potentially transformative
medicine, which we believe has significant potential for a different severe and
rare trinucleotide repeat disease, DM1. Myotonic dystrophy type 1 (DM1) is a
multisystem disorder that primarily affects skeletal and smooth muscle. DM1 is
caused by expansion of a CTG trinucleotide repeat in the noncoding region of the
DMPK gene, which captures and sequesters splice proteins. Sequestered splice
proteins cannot then fulfill their normal functions. The diagnosis of DM1 is
suspected in individuals with characteristic muscle weakness and is confirmed by
molecular genetic testing of DMPK. CTG repeat length exceeding 34 repeats is
abnormal. Molecular genetic testing detects pathogenic variants in nearly 100%
of affected individuals. It is estimated that the global prevalence of DM1 is
1:20,000 individuals. The clinical candidates in development target the DM1
expanded allele with PATrOL™-enabled drug candidates to disrupt and/or open the
mutant hairpin and allow release of sequestered splice proteins.



Additional Indications



In addition, we are in the process of building an early stage pipeline of other
therapies that focus on the unique advantages of our technology across a variety
of rare diseases.


Critical Accounting Estimates and Policies





The preparation of financial statements in accordance with United States
generally accepted accounting principles ("U.S. GAAP") requires management to
make estimates and assumptions that affect the amounts reported in our unaudited
condensed consolidated financial statements and accompanying notes. Management
bases its estimates on historical experience, market and other conditions, and
various other assumptions it believes to be reasonable. Although these estimates
are based on management's best knowledge of current events and actions that may
impact us in the future, the estimation process is, by its nature, uncertain
given that estimates depend on events over which we may not have control. If
market and other conditions change from those that we anticipate, our unaudited
condensed consolidated financial statements may be materially affected. In
addition, if our assumptions change, we may need to revise our estimates, or
take other corrective actions, either of which may also have a material effect
in our unaudited condensed consolidated financial statements. We review our
estimates, judgments, and assumptions used in our accounting practices
periodically and reflect the effects of revisions in the period in which they
are deemed to be necessary. We believe that these estimates are reasonable;
however, our actual results may differ from these estimates.



Our critical accounting policies and estimates are discussed in our Annual
Report on Form 10-K for the fiscal year ended September 30, 2019 and there have
been no material changes to such policies or estimates during the three months
ended December 31, 2019.


Recent Accounting Pronouncements

Please refer to Note 2, Significant Accounting Policies-Recent Accounting Pronouncements, in Item 1, Financial Statements for a discussion of recent accounting pronouncements.





                                       20





Results of Operations


Results of operations for the quarter ended December 31, 2019 reflect the following changes from the quarter ended December 31, 2018:





                                                           Three Months Ended December 31,
                                                           2019                  2018             Change
                                                                            (As Restated)
OPERATING EXPENSES

General and administrative expenses                  $      2,554,680       $      422,010     $  2,132,670
Research and development expenses                           1,227,686                4,876        1,222,810
Research and development expense- license acquired                  -            1,046,965       (1,046,965 )
TOTAL OPERATING EXPENSES                                    3,782,366      

1,473,851 2,308,515


LOSS FROM OPERATIONS                                       (3,782,366 )    

    (1,473,851 )     (2,308,515 )

OTHER EXPENSE
Interest expense                                               (1,311 )            (14,637 )         13,326

Change in fair value of warrant liabilities                  (694,134 )                  -         (694,134 )
Loss on disposal of fixed asset                                (3,230 )                  -           (3,230 )
Equity in losses on equity method investment                  (24,509 )    

             -          (24,509 )

Total other expenses                                         (723,184 )            (14,637 )       (708,547 )

NET LOSS                                             $     (4,505,550 )
$   (1,488,488 )   $ (3,017,062 )




During the quarter ended December 31, 2019, operating loss increased by $2.3
million compared to the quarter ended December 31, 2018. Our net loss increased
by $3.0 million for the quarter ended December 31, 2019, as compared to the
quarter ended December 31, 2018. Until we are able to generate revenue from
product sales, our management expects to continue to incur net losses.



General and Administrative Expenses





General and administrative expenses consist primarily of legal and professional
fees, wages and stock-based compensation. General and administrative expenses
increased by $2.1 million for the quarter ended December 31, 2019, as compared
to the quarter ended December 31, 2018, primarily due to an increase in
stock-based compensation expense, employee head count and need for legal and
professional services.


Research and Development Expenses





Research and development expenses consist primarily of professional fees,
manufacturing expenses, wages and stock-based compensation. Research and
development expenses increased by $1.2 million for the quarter ended December
31, 2019, as compared to the quarter ended December 31, 2018, primarily due to
an increase in stock-based compensation, employee head count and the ramp up of
research and development activities.



Research and Development Expense- licenses acquired





Research and development expense- licenses acquired during the quarter ended
December 31, 2018 consists of the license acquired from CMU. Research and
development expense- licenses acquired decreased by $1.0 million, for the
quarter ended December 31, 2019, as compared to the quarter ended December 31,
2018, due to our acquisition of license rights in the 2018 period.



                                       21





Interest Expense



Interest expense consists primarily of interest on convertible notes and notes
payable. Interest expense decreased by $0.01 million for the quarter ended
December 31, 2019, as compared to the quarter ended December 31, 2018, primarily
due to the outstanding convertible notes in the quarter ended December 31, 2019,
which were converted prior to the quarter ended December 31, 2019.



Change in fair value of warrant liabilities





Change in fair value of warrant liabilities reflects the changes in the fair
value of outstanding warrants which is primarily driven by changes in our stock
price. Change in fair value of warrant liabilities was $0.7 million for the
quarter ended December 31, 2019, as compared to the quarter ended December 31,
2018, due to the change in valuation of warrants acquired in the Merger with
Ohr, which did not exist in the comparative prior.



Equity in losses on equity method investment


The Company accounts for its investment in DepYmed common shares using the
equity method of accounting and records its proportionate share of DepYmed's net
income and losses. Equity in losses for the three months ended December 31, 2019
was approximately $0.02 million.



Liquidity, Capital Resources and Financial Condition

We have limited working capital reserves with which to fund our continuing operations. We are reliant, at present, upon our capital reserves for ongoing operations and have no product revenue.


Net working capital decreased from September 30, 2019 to the quarter ended
December 31, 2019 by $2.8 million (to $5.7 million from $8.5 million) primarily
due to development of our PATrOL™ platform technology and lead programs. Our
quarterly cash burn has increased significantly compared to prior periods due to
increased research and development activities. We anticipate that our cash needs
in the future will increase relative to prior periods as we proceed with our
research and development objectives. We believe that our current cash balance
will provide sufficient capital to continue operations to the end of fiscal
2020. We are closely monitoring ongoing developments in connection with the
COVID-19 pandemic, which may negatively impact our commercial prospects and
projected cash position in fiscal 2020. We will continue to assess our cash and
cash equivalents and, if circumstances warrant, we will make appropriate
adjustments to our operating plan. At present, however, we have no bank line of
credit or other fixed source of capital reserves. Should we need additional
capital in the future, we will be primarily reliant upon private or public
placement of our equity or debt securities, or a strategic transaction, for
which there can be no warranty or assurance that we may be successful in such
efforts. If the Company is unable to maintain sufficient financial resources,
its business, financial condition and results of operations will be materially
and adversely affected. This could affect future development and business
activities and potential future clinical studies and/or other future ventures.
Failure to obtain additional equity or debt financing will have a material,
adverse impact on the Company's business operations. There can be no assurance
that the Company will be able to obtain the needed financing on acceptable terms
or at all. Additionally, equity or debt financings will likely have a dilutive
effect on the holdings of the Company's existing stockholders. Accordingly,
there are material risks and uncertainties that raise substantial doubt about
the Company's ability to continue as a going concern.





Cash Flow Summary



The following table summarizes selected items in our condensed consolidated
statements of cash flows:



                                                   Three Months Ended
                                                      December 31,
                                                2019              2018
                                                              (As Restated)

Net cash used in operating activities $ (2,441,486 ) $ (14,933 ) Net cash used in investing activities

            (68,400 )           (97,463 )
Net used in financing activities                 (73,426 )               

(14 ) Net decrease in cash and cash equivalents $ (2,583,312 ) $ (112,410 )






Operating Activities



Net cash used in operating activities was approximately $2.4 million for the
quarter ended December 31, 2019, as compared to approximately $0.02 million for
the quarter ended December 31, 2018. Net cash used in operating activities in
the quarter ended December 31, 2019 was primarily the result of our net loss,
offset by our stock-based compensation expense and the change in fair value of
warrant liabilities. Net cash used in operating activities in the quarter ended
December 31, 2018 was primarily the result of our net loss, offset by research
and development expense-licenses acquired.



Investing Activities



Net cash used in investing activities was approximately $0.07 million for the
quarter ended December 31, 2019, as compared to $0.1 million for the quarter
ended December 31, 2018. Net cash used in investing activities in the quarter
ended December 31, 2019 was primarily the result of purchases of laboratory
equipment. Net cash used in investing activities in the quarter ended December
31, 2018 was primarily the result of costs paid in connection with the
acquisition of the CMU License.



                                       22





Financing Activities



Net cash used in financing activities was approximately $0.07 million for the
quarter ended December 31, 2019, as compared to approximately $14 for the
quarter ended December 31, 2018. Net cash used in financing activities for the
quarter ended December 31, 2019 reflects the principal payments of financed
insurance. Net cash used in financing activities for the quarter ended December
31, 2018 reflects the repurchase of common stock.



Off-Balance Sheet Arrangements

As of December 31, 2019, we did not have any off-balance sheet arrangements as defined in Item 303(a)(4)(ii) of Regulation S-K.

© Edgar Online, source Glimpses