You should read the following discussion and analysis of our financial condition
and results of operations together with our financial statements and the related
notes appearing elsewhere in this report. In addition to historical information,
this discussion and analysis contains forward-looking statements that involve
risks, uncertainties and assumptions. Our actual results may differ materially
from those discussed below. Factors that could cause or contribute to such
differences include, but are not limited to, those identified below, and those
discussed in the section titled "Risk Factors" included elsewhere in this report
and our annual report on Form 10-K for the year ended December 31, 2021.

The following discussion highlights our results of operations and the principal
factors that have affected our financial condition as well as our liquidity and
capital resources for the periods described, and provides information that
management believes is relevant for an assessment and understanding of the
statements of financial condition and results of operations presented herein.
The following discussion and analysis are based on the unaudited financial
statements contained in this report, which we have prepared in accordance with
United States generally accepted accounting principles. You should read the
discussion and analysis together with such financial statements and the related
notes thereto.

Explanatory Note

On May 17, 2020, Neurotrope, Inc. (Neurotrope or Parent) announced plans for the
complete legal and structural separation of us from Neurotrope, also known as
the Spin-Off. Under the Separation and Distribution Agreement, Neurotrope
planned to distribute all of its equity interest in us to Neurotrope's
stockholders. Following the Spin-Off, Neurotrope would not own any equity
interest in us, and we would operate independently from Neurotrope. Neurotrope
Bioscience, Inc. was a wholly-owned subsidiary of Neurotrope prior to the
completion of the Spin-Off on December 7, 2020 (see below for description of
Spin-Off). Neurotrope Bioscience, Inc. represented substantially all the
business of Neurotrope.

On December 6, 2020, Neurotrope approved the final distribution ratio and
holders of record of Neurotrope common stock, Neurotrope preferred stock and
certain warrants as of November 30, 2020 received a pro rata distribution at the
rate of (i) one share of our Common Stock for every five shares of Neurotrope
common stock held, (ii) one share of our Common Stock for every five shares of
Neurotrope common stock issuable upon conversion of Neurotrope preferred stock
held and (iii) one share of our Common Stock for every five shares of Neurotrope
common stock issuable upon exercise of certain Neurotrope warrants held that
were entitled to participate in the Spin-Off pursuant to the terms thereof.

Basis of Presentation


The unaudited financial statements for the three and six months ended June
30,2022 and 2021 include a summary of our significant accounting policies and
should be read in conjunction with the discussion below and our financial
statements and related notes included elsewhere in this quarterly report. In the
opinion of management, all material adjustments necessary to present fairly the
results of operations for such periods have been included in the financial
statements. All such adjustments are of a normal recurring nature.

Overview


We are a biopharmaceutical company with product candidates in pre-clinical and
clinical development. We began operations in October 2012. We are principally
focused on developing a product platform based upon a drug candidate called
Bryostatin-1 for the treatment of Alzheimer's disease, which is in the clinical
testing stage. We are also evaluating Bryostatin-1 for other neurodegenerative
or cognitive diseases and dysfunctions, such as Fragile X syndrome, Multiple
Sclerosis, and Niemann-Pick Type C disease, which have undergone pre-clinical
testing.

                                       21

  Table of Contents
Neurotrope had been a party to a technology license and services agreement with
the original Blanchette Rockefeller Neurosciences Institute (which has been
known as Cognitive Research Enterprises, Inc. since October 2016), and its
affiliate NRV II, LLC, which we collectively refer to herein as "CRE," pursuant
to which we now have an exclusive non-transferable license to certain patents
and technologies required to develop our proposed products. We were formed for
the primary purpose of commercializing the technologies initially developed by
BRNI for therapeutic applications for AD or other cognitive dysfunctions. These
technologies have been under development by BRNI since 1999 and, until
March 2013, had been financed through funding from a variety of non-investor
sources (which include not-for-profit foundations, the NIH, which is part of the
U.S. Department of Health and Human Services, and individual philanthropists).
From March 2013 forward, development of the licensed technology has been funded
principally through us in collaboration with CRE.

Reverse Stock Split



On April 7, 2021, our stockholders approved our proposal to effect one reverse
stock split of our outstanding shares of Common Stock, at any ratio between
1-for-1.5 and 1-for-20, at such time as the Board shall determine, in its sole
discretion, before December 31, 2022. On May 19, 2021, we effected a 1-for-4
reverse stock split of our shares of Common Stock. As a result of the reverse
stock split, every four (4) shares of our pre-reverse split Common Stock was
combined and reclassified into one share of Common Stock. All share and per
share information herein has been adjusted to retrospectively reflect this
reverse stock split.

Results of Most Recent Confirmatory Phase 2 Clinical Trial

On September 9, 2019, Neurotrope issued a press release announcing that the confirmatory Phase 2 study of Bryostatin-1 in moderate to severe AD did not achieve statistical significance on the primary endpoint, which was changed from baseline to Week 13 in the Severe Impairment Battery ("SIB") total score.



An average increase in SIB total score of 1.3 points and 2.1 points was observed
for the Bryostatin-1 and placebo groups, respectively, at Week 13. There were
multiple secondary outcome measures in this trial, including the changes from
baseline at Weeks 5, 9 and 15 in the SIB total score. No statistically
significant difference was observed in the change from baseline in SIB total
score between the Bryostatin -1 and placebo treatment groups.

The confirmatory Phase 2 multicenter trial was designed to assess the safety and
efficacy of Bryostatin-1 as a treatment for cognitive deficits in patients with
moderate to severe AD - defined as a MMSE-2 score of 4-15 - who are not
currently taking memantine. Patients were randomized 1:1 to be treated with
either Bryostatin-1 20?g or placebo, receiving 7 doses over 12 weeks. Patients
on memantine, an NMDA receptor antagonist, were excluded unless they had been
discontinued from memantine treatment for a 30-day washout period prior to study
enrollment. The primary efficacy endpoint was the change in the SIB score
between the baseline and week 13. Secondary endpoints included repeated SIB
changes from baseline SIB at weeks 5, 9, 13 and 15.

On January 22, 2020, we announced the completion of an additional analysis in
connection with the confirmatory Phase 2 study, which examined moderately severe
to severe AD patients treated with Bryostatin-1 in the absence of memantine. To
adjust for the baseline imbalance observed in the study, a post-hoc analysis was
conducted using paired data for individual patients, with each patient as
his/her own control. For the pre-specified moderate stratum (i.e., MMSE-2
baseline scores 10-15), the baseline value and the week 13 value were used,
resulting in pairs of observations for each patient. The changes from baseline
for each patient were calculated and a paired t-test was used to compare the
mean change from baseline to week 13 for each patient. A total of 65 patients
had both baseline and week 13 values, from which there were 32 patients in the
Bryostatin-1 treatment group and 33 patients in the placebo group. There was a
statistically significant improvement over baseline (4.8 points) in the mean SIB
at week 13 for subjects in the Bryostatin-1 treatment group (32 subjects),
paired t-test p < 0.0076, 2-tailed. In the placebo group (33 subjects), there
was also a statistically significant increase from baseline in the mean SIB at
week 13, for paired t-test p < 0.0144, consistent with the placebo effect seen
in the overall 203 study. Although there was a signal of Bryostatin-1's benefit
for the moderately severe stratum, the difference between the Bryostatin-1 and
placebo treatment groups was not statistically significant (p=0.2727). As a
further test of the robustness of this Moderate Stratum benefit signal, a
pre-specified trend analysis (measuring increase of SIB improvement as a
function of successive drug doses) was performed on the repeated SIB measures
over time (Weeks 0, 5, 9, and 13). These trend analyses showed a significant
positive slope of improvement for the treatment groups in the 203 study that was
significantly greater than for the placebo group (p<.01).

                                       22

Table of Contents


In connection with the additional analysis, we also announced the approval of a
$2.7 million award from the NIH to support an additional Phase 2 clinical study
focused on the moderate stratum for which we saw improvement in the 203 study.
The grant provides for funds in the first year of approximately $1.0 million and
funding in year two of approximately $1.7 million subject to satisfactory
progress of the project. We are planning to meet with the FDA to present the
totality of the clinical data for Bryostatin-1.

On July 23, 2020, we entered into the 2020 Services Agreement with WCT. The 2020
Services Agreement relates to services for our Phase 2 clinical study assessing
the safety, tolerability and long-term efficacy of Bryostatin-1 in the treatment
of moderately severe AD subjects not receiving memantine treatment. On January
22, 2022, the Company executed a change order with WCT to accelerate trial
subject recruitment totaling approximately $1.4 million. In addition, on
February 10, 2022, the Company signed an additional agreement with a third-party
vendor to assist with the increased trial recruitment retention totaling
approximately $1.0 million. The updated total estimated budget for the services,
including pass-through costs, is approximately $12.0 million. As previously
disclosed, on January 22, 2020, we were granted a $2.7 million award from the
NIH, which award is being used to support the 2020 Study, resulting in an
estimated net budgeted cost of the 2020 Study to us of $9.3 million. Of the $2.7
million grant, virtually all has been received as of February 22, 2022.

As of June 30, 2022, we incurred cumulative expenses of approximately $9.4
million associated with services provided by WCT and certain pass thru expenses
incurred by WCT, which was offset by NIH reimbursements recognized of $2.7
million and, for the three months ended June 30, 2022, we incurred expenses of
approximately $1.3 million associated with services provided by WCT and certain
pass thru expenses incurred by WCT.

Additional Phase 2 Clinical Trial


On May 12, 2022, the Company entered into the 2022 Services Agreement with WCT.
The 2022 Services Agreement relates to services for a Phase 2 "open label," dose
ranging study, clinical trial assessing the safety, tolerability and efficacy of
Bryostatin-1 administered via infusion in the treatment of moderately severe to
severe AD subjects not receiving memantine treatment.

Pursuant to the terms of the 2022 Services Agreement, WCT is providing services
to enroll approximately 12 2022 Study subjects, which enrollment is currently
underway. The first 2022 Study site was initiated during the third quarter of
2022. The total estimated budget for the services, including pass-through costs,
is currently approximately $2.0 million. During the three and six months ended
June 30, 2022, the Company incurred a total of approximately $450,000 for this
clinical trial and, as of June 30, 2022, recorded a payable of approximately
$80,000 on its balance sheet. During July 2022, the Company paid WCT 20%, or
approximately $400,000, as a prepayment for services, pass thru and site
expenses to be incurred for the 2022 Services Agreement. Either party may
terminate the 2022 Services Agreement without cause upon ninety days prior
written notice. Furthermore, in the event of a material breach by the other
party, which breach is not cured by the breaching party, the other party may
terminate the agreement upon 30 days' prior written notice.

Other Development Projects



To the extent resources permit, we may pursue development of selected technology
platforms with indications related to the treatment of various disorders,
including neurodegenerative disorders such as AD, based on our currently
licensed technology and/or technologies available from third party licensors or
collaborators.

Nemours Agreement

On September 5, 2018, we announced a collaboration with Nemours, a premier U.S.
children's hospital, to initiate a clinical trial in children with Fragile X. In
addition to the primary objective of safety and tolerability, measurements will
be made of working memory, language and other functional aspects such as
anxiety, repetitive behavior, executive functioning, and social behavior. On
August 5, 2021, the Company announced its memorandum of understanding with
Nemours A.I. DuPont Hospital ("Nemours") to initiate a clinical trial using
Bryostatin-1, under Orphan Drug Status, to treat Fragile X. The Company intends
to provide the Bryostatin-1 drug product candidate and obtain the
investigational new drug documentation ("IND") and Nemours intends to provide
the clinical site and attendant support for the trial. The Company and Nemours,
jointly, will develop the trial protocol. The Company estimates its total trial
and IND cost to be approximately $700,000. The Company plans to initiate a Phase
1 clinical trial during the second half of 2022.

                                       23

  Table of Contents

Cleveland Clinic

On February 23, 2022, the Company announced its collaboration with the Cleveland
Clinic to pursue possible treatments for Multiple Sclerosis. The collaboration
entails filing an IND and conducting initial clinical trials using Bryostatin-1.
Future development work will be conducted pursuant to statements of work to

be
determined.

Impact of COVID-19

We face the ongoing risk that the coronavirus pandemic may slow the conduct of
our current trials. In order to prioritize patient health and that of the
investigators at clinical trial sites, we will monitor enrollment of new
patients in our Phase 2 clinical trials of Bryostatin-1 for the treatment of
patients with Alzheimer's disease. In addition, some patients may be unwilling
to enroll in our trials or be unable to comply with clinical trial protocols if
quarantines or travel restrictions impede patient movement or interrupt
healthcare services. These and other factors outside of our control could delay
our ability to conduct clinical trials or release clinical trial results. In
addition, the effects of the ongoing coronavirus pandemic may also increase
non-trial costs such as insurance premiums, increase the demand for and cost of
capital, increase loss of work time from key personnel, and negatively impact
our key clinical trial vendors and supplier of API.

In light of the COVID-19 outbreak, the FDA has issued a number of new guidance
documents. Specifically, as a result of the potential effect of the COVID-19
outbreak on many clinical trial programs in the US and globally, the FDA issued
guidance concerning potential impacts on clinical trial programs, changes that
may be necessary to such programs if they proceed, considerations regarding
trial suspensions and discontinuations, the potential need to consult with or
make submissions to relevant ethics committees, Institutional Review Board
("IRBs"), and the FDA, the use of alternative drug delivery methods, and
considerations with respect the outbreak's impacts on endpoints, data
collection, study procedures, and analysis. Such developments may result in
delays in our development of Bryostatin-1.

Results of Operations

Comparison of the six months ended June 30, 2022 and 2021

The following table summarizes our results of operations for the six months ended June 30, 2022 and 2021:



                                                       Six Months ended
                                                           June 30,               Dollar
                                                     2022           2021          Change       % Change
Revenue                                           $         -    $         -    $         -           0 %
Operating Expenses:
Research and development expenses                 $ 3,427,175    $ 2,138,060    $ 1,289,115        60.3 %
General and administrative expenses               $ 3,651,748    $ 3,312,306    $   339,442        10.2 %
Other income, net                                 $    49,097    $     2,147    $    46,950     2,186.8 %
Net loss                                          $ 7,029,826    $ 5,448,219    $ 1,581,607        29.0 %


Revenues

We did not generate any revenues for the six months ended June 30, 2022 and 2021.



Operating Expenses

Overview

Total operating expenses for the six months ended June 30, 2022 were $7,078,923
as compared to $5,450,366 for the six months ended June 30, 2021, an increase of
approximately 29.9%. The increase in total operating expenses is due to the
increase in research and development activities and general and administrative
expenses.

                                       24

  Table of Contents

Research and Development Expenses


For the six months ended June 30, 2022, we incurred $3,427,175 in research and
development expenses as compared to $2,138,060 for the six months ended June 30,
2021, an increase of approximately 60.3%. These expenses were incurred primarily
pursuant to developing the potential AD therapeutic product, specifically
expenses relating to our ongoing Phase 2 clinical trial for AD. Of these
expenses, for the six months ended June 30, 2022, $2,943,402 was incurred
principally relating to our current confirmatory clinical trial and related
storage of drug product, $152,658 for clinical consulting services, $14,931 of
amortization of prepaid licensing fees relating to the Stanford License
Agreement and Mount Sinai Agreement, $23,481 for development of alternative drug
supply with Stanford University and $292,703 of non-cash stock options
compensation expense; comparatively, for the six months ended June 30, 2021,
$1,732,669 was incurred principally relating to our confirmatory clinical trial
and related storage of drug product, $156,853 for clinical consulting services,
$14,876 of amortization of prepaid licensing fees relating to the Stanford
License Agreement and Mount Sinai Agreement, $24,197 for development of
alternative drug supply with Stanford University and $209,465 of non-cash stock
options compensation expense.

We expect our research and development expenses to level off as we expect that
our current Phase 2 clinical trial for AD will be materially concluded by the
end of 2022 while, to a lesser extent, our Phase 2 dose ranging study activities
increase. Other development expenses might increase, as our resources permit, in
order to advance our potential products. We are continuing to determine how to
proceed with respect to our other current development programs for Bryostatin-1.

General and Administrative Expenses



We incurred $3,651,748 and $3,312,306 of general and administrative expenses for
the six months ended June 30, 2022 and 2021, respectively, an increase of
approximately 10.2%. During the six months ended June 30, 2022, $601,507 was
incurred primarily for wages, bonuses, vacation pay, severance, taxes and
insurance, versus $667,552 for the six months ended June 30, 2021. The decrease
in wages is primarily attributable to the reduction of bonus accruals for our
former Chief Executive Officer; $207,019 was incurred for legal expenses versus
$468,406 for the 2021 comparable period. The decrease in legal fees for 2022 is
based upon prior year's increased fees for one-time work for fundraising
planning in 2021; $589,454 was incurred for outside operations consulting
services during the six months ended June 30, 2022, versus $956,530 for the
comparable period in 2021 as, during such period in 2021, we incurred additional
non-cash expenses associated with warrant issuances for investment banking
consulting services; $39,131 was incurred for travel expenses during the six
months ended June 30, 2022, versus $28,167 for the comparable period in 2021;
$133,569 was incurred for investor relations services during the six months
ended June 30, 2022, versus $156,528 for the comparable period in 2021; $109,725
was incurred for professional fees associated with auditing, financial,
accounting and tax advisory services during the six months ended June 30, 2022,
versus $88,163 for the comparable period in 2021; $365,544 was incurred for
insurance during the six months ended June 30, 2022, versus $327,545 for the
comparable period in 2021, which increase is primarily attributable to an
increase in premiums; $170,254 was incurred for utilities, supplies, license
fees, filing costs, rent, advertising and other during the six months ended June
30, 2022, versus $109,089 for the comparable period in 2021, with the increase
for 2022 primary attributable to stock market up-listing fees to Nasdaq and rent
expenses associated with our new Maryland laboratory facility; and $1,435,545
was recorded as non-cash stock options compensation expense during the six
months ended June 30, 2022, versus $510,326 for the comparable period in 2021,
which increase is primarily attributable to equity awards granted after June 30,
2021 expensed in 2022.

Other Income / Expense

We earned $49,097 of net interest income for the six months ended June 30, 2022
as compared to $2,147 for the six months ended June 30, 2021 on funds deposited
in interest bearing money market accounts. The increase is primarily
attributable to the increase in money market interest income rates and higher
average cash balances in 2022 compared to the corresponding period in 2021.

Net loss



We incurred losses of $7,029,826 and $5,448,219 for the six months ended June
30, 2022 and 2021, respectively. The increased loss was primarily attributable
to the increase in net research and development expenses associated with our
current Phase 2 confirmatory clinical trial and an increase in general and

administrative expenses.

                                       25

  Table of Contents

Comparison of the three months ended June 30, 2022 and 2021

The following table summarizes our results of operations for the three months ended June 30, 2022 and 2021:



                                                Three Months ended
                                                     June 30,                Dollar
                                                2022          2021           Change       % Change
Revenue                                      $         -   $         -    $          -           0 %
Operating Expenses:

Research and development expenses            $ 1,941,101   $   986,280    $    954,821        96.8 %
General and administrative expenses          $ 1,855,290   $ 1,308,893    $

   546,397        41.7 %
Other income, net                            $    44,487   $     1,331    $     43,156     3,242.4 %
Net loss                                     $ 3,751,904   $ 2,293,842    $  1,458,062        63.6 %


Revenues

We did not generate any revenues for the three months ended June 30, 2022 and 2021.



Operating Expenses

Overview

Total operating expenses for the three months ended June 30, 2022 were
$3,796,391 as compared to $2,295,173 for the three months ended June 30, 2021,
an increase of approximately 65.4%. The increase in total operating expenses is
due to the increase in research and development and general and administrative
expenses.

Research and Development Expenses



For the three months ended June 30, 2022, we incurred $1,832,792 in research and
development expenses as compared to $986,280 for the three months ended June 30,
2021, an increase of approximately 85.8%. These expenses were incurred primarily
pursuant to developing the potential AD therapeutic product, specifically
expenses relating to our ongoing Phase 2 clinical trial for AD. Of these
expenses, for the three months ended June 30, 2022, $1,680,918 was incurred
principally relating to our current confirmatory clinical trial and related
storage of drug product, $73,113 for clinical consulting services, $7,479 of
amortization of prepaid licensing fees relating to the Stanford License
Agreement and Mount Sinai Agreement, $7,938 for development of alternative drug
supply with Stanford University and $171,653 of non-cash stock options
compensation expense as compared to, for the three months ended June 30, 2021,
$834,080 was incurred principally relating to our confirmatory clinical trial
and related storage of drug product, $93,203 for clinical consulting services,
$7,479 of amortization of prepaid licensing fees relating to the Stanford and
Mount Sinai license agreements, $15,755 for development of alternative drug
supply with Stanford University and $35,763 of non-cash stock options
compensation expense.

We expect our research and development expenses to level off as our current
Phase 2 clinical trial for AD will be materially concluded by the end of 2022
while, to a lesser extent, our Phase 2 dose ranging study activities increases.
Other development expenses might increase, as our resources permit, in order to
advance our potential products. We are continuing to determine how to proceed
with respect to our other current development programs for Bryostatin-1.

                                       26

Table of Contents

General and Administrative Expenses



We incurred $1,855,290 and $1,308,893 of general and administrative expenses for
the three months ended June 30, 2022 and 2021, respectively, an increase of
approximately 41.7%. Of the amounts for the three months ended June 30, 2022, as
compared to the comparable 2021 period: $307,603 was incurred primarily for
wages, bonuses, vacation pay, severance, taxes and insurance, versus $237,337
for the 2021 comparable period. The increase is primarily attributable to the
reclassification of our former Chief Executive Officer's compensation to
consulting fees for the comparable 2021 period; $77,804 was incurred for legal
expenses versus $233,822 for the 2021 comparable period. The decrease for 2022
is based upon prior year's increased fees for one-time work for fundraising
planning in 2021; $193,250 was incurred for outside operations consulting
services during the three months ended June 30, 2022, versus $363,854 for the
comparable period in 2021 as, during such period in 2021, we incurred additional
non-cash expenses associated with warrant issuances for investment banking
consulting services; $24,479 was incurred for travel expenses during the three
months ended June 30, 2022, versus $17,417 for the comparable period in 2021;
$61,355 was incurred for investor relations services during the three months
ended June 30, 2022, versus $89,459 for the comparable period in 2021; $87,677
was incurred for professional fees associated with auditing, financial,
accounting and tax advisory services during the three months ended June 30,
2022, versus $38,268 for the comparable period in 2021. The increase for 2022 is
attributable to expensing the 2021 audit in the second quarter of 2022 versus
during the first quarter in 2021; $181,236 was incurred for insurance during the
three months ended June 30, 2022, versus $165,437 for the comparable period in
2021, which increase is primarily attributable to an increase in premiums;
$76,849 was incurred for utilities, supplies, license fees, filing costs, rent,
advertising and other during the three months ended June 30, 2022, versus
$47,510 for the comparable period in 2021, with the increase for 2022 primary
attributable to stock market up-listing fees to Nasdaq and rent expenses
associated with our new Maryland laboratory facility; and $845,037 was recorded
as non-cash stock options compensation expense during the three months ended
June 30, 2022, versus $115,789 for the comparable period in 2021, which increase
is primarily attributable to equity awards granted after June 30, 2021 expensed
in 2022.

Other Income / Expense

We earned $44,487 of net interest income for the three months ended June 30,
2022 as compared to $1,331 for the three months ended June 30, 2021 on funds
deposited in interest bearing money market accounts. The increase is primarily
attributable to the increase in money market interest income rates and higher
average cash balances in 2022 compared to the corresponding period in 2021.

Net loss


We incurred losses of $3,751,904 and $2,293,842 for the three months ended June
30, 2022 and 2021, respectively. The increased loss was primarily attributable
to the increase in net research and development expenses associated with our
current Phase 2 confirmatory clinical trial and the increase in general and
administrative expenses.

Financial Condition, Liquidity and Capital Resources

Cash and Working Capital



Since inception, we have incurred negative cash flows from operations. As of
June 30, 2022, we had working capital of $28,932,496 as compared to working
capital of $33,509,303 as of December 31, 2021. The $4,576,807 decrease in
working capital was primarily attributable to approximately $7.1 million of
operating expenses partially offset by non-cash expenses of approximately $1.8
million and cash proceeds from warrant exercises of approximately $553,000.

We expect that our current cash and cash equivalents of approximately $27.4
million will be sufficient to support our projected operating requirements for
at least the next 12 months from the Form 10-Q filing date, which would include
the continuing development and current Phase 2 clinical trial, of Bryostatin-1,
our novel drug candidate targeting the activation of PKC epsilon.

                                       27

Table of Contents



We expect to require additional capital in order to initiate, pursue and
complete all potential AD clinical trials and obtain regulatory approval of one
or more therapeutic candidates. However, additional future funding may not be
available to us on acceptable terms, or at all. If we are unable to access
additional funds when needed, we may not be able to initiate, pursue and
complete all planned clinical trials or continue the development of our product
candidates or we could be required to delay, scale back or eliminate some or all
of our development programs and operations. Any additional equity financing, if
available, may not be available on favorable terms, would most likely be
significantly dilutive to our current stockholders and debt financing, if
available, and may involve restrictive covenants. If we are able to access funds
through collaborative or licensing arrangements, we may be required to
relinquish rights to some of our technologies or product candidates that we
would otherwise seek to develop or commercialize on our own, on terms that are
not favorable to us. Our ability to access capital when needed is not assured
and, if not achieved on a timely basis, would likely materially harm our
business and financial condition.

Sources and Uses of Liquidity



Since inception, we have satisfied our operating cash requirements from
transfers of cash from Neurotrope, which was raised by Neurotrope through the
private placement of equity securities sold principally to outside investors and
through the Company's two private placements. We expect to continue to incur
expenses, resulting in losses and negative cash flows from operations, over at
least the next several years as we continue to develop AD and other therapeutic
products. We anticipate that this development may include clinical trials in
addition to our current ongoing clinical trial and additional research and
development expenditures.

                                           Six Months Ended June 30,
                                             2022             2021

Cash used in operating activities $ 6,009,489 $ 5,585,322 Cash used in investing activities

                3,029               -

Cash provided by financing activities 553,150 31,492,353

Net Cash Used in Operating Activities



Cash used in operating activities was $6,009,489 for the six months ended June
30, 2022, compared to $5,585,322 for the six months ended June 30, 2021. The
$424,167 increase primarily resulted from the increase in net loss of
approximately $1.6 million and the decrease in accounts payable of approximately
$403,000 partially offset by a decrease in net of prepaid and accrued expenses
of approximately $795,000 and an increase in non-cash stock-based compensation
of approximately $765,000 for the six months ended June 30, 2022.

Net Cash Used in Investing Activities



Net cash used in investing activities was $3,029 for the six months ended June
30, 2022 compared to $0 for the six months ended June 30, 2021. The cash used in
investing activities for the six months ended June 30, 2022 was for capital
expenditures.

Net Cash Provided by Financing Activities



Net cash provided by financing activities was $553,150 for the six months ended
June 30, 2022 compared to cash provided by financing activities of $31,492,353
for the six months ended June 30, 2021. The net cash provided by financing
activities for 2022 resulted from the exercise of investor warrants issued in
2021. The net cash provided by financing activities in 2021 was primarily
attributable to $23.7 million of net proceeds from our private placement
offerings, $7.6 million from warrant exercises and $127,000 from NIH grant
funding proceeds

© Edgar Online, source Glimpses