SAVARA INC. (NASDAQ:SVRA) Files An 8-K Other Events

0

SAVARA INC. (NASDAQ:SVRA) Files An 8-K Other Events

Item8.01

Other Events.

Clinical Development of Molgradex

Phase3

Savara is currently conducting a Phase 3 clinical study on
Molgradex in Europe and Japan in autoimmune pulmonary alveolar
proteinosis, or PAP, patients. Based on guidance received from
the U.S. Food and Drug Administration, or FDA, as well as
scientific advice received from the European Medicines Agency, or
EMA, Savara believes the study has the potential to be accepted
as the sole pivotal study in support of a BLA submission in the
United States, and a marketing authorization application in the
European Union. The aim of this randomized, double-blind,
placebo-controlled study is to compare efficacy and safety of
Molgradex with placebo in up to 90 PAP patients. Enrollment of
the study is expected to be completed by the first quarter of
2018, with top line data readout expected in the fourth quarter
of 2018.

The Phase 3 clinical study on Molgradex is enrolling patients
diagnosed with autoimmune PAP and fulfilling all other entry
criteria. These patients are randomized to receive double-blind
treatment for up to 24 weeks in one of three treatment arms: 1)
Molgradex 300 g administered once daily, 2) Molgradex 300 g and
matching placebo administered daily in 7-day intermittent cycles
of each, or 3) inhaled placebo administered once daily. The
placebo-controlled period is followed by an open label follow-up
period of 24-48 weeks. The study is being conducted at multiple
sites in the European Union, Russia, Israel and Japan, with plans
to open new study sites in certain other countries, including the
United States. The primary endpoint of the study is the absolute
change from baseline of arterial-alveolar oxygen gradient
((A-a)DO2) after 24 weeks of treatment. This endpoint is a
measure of the patients oxygenation status, and the endpoint
value is expected to decrease as the physical obstacle of gas
exchange is reduced by clearance of excess surfactant from the
lungs. In addition, based on FDA guidance recently received by
Savara, the FDA will focus its review on three key secondary
endpoints that will be assessed for improvement in clinical
symptoms and function, including six-minute walk distance (6MWD),
St. Georges respiratory questionnaire (SGRQ), and the time to
whole lung lavage.

Based on the sample size calculation for the study, 42 evaluable
patients (14 in each treatment group) are required to be
randomized to have 90% power to detect a difference of 10 mmHg in
(A-a)DO between the two active
arms combined and placebo, using a significance level of 0.01. To
account for the FDAs review of the key secondary endpoints, a
sample size of 30 patients per treatment group will be enrolled
to achieve approximately 90% power to detect a difference of 50
meters in the 6MWD between each active arm and placebo, and a
difference of ten points in the SGRQ between each active arm and
placebo, each active arm analyzed sequentially using a
significance level of approximately 0.02, after correction for
multiplicity with an appropriate method for correlated
endpoints.

A data safety monitoring
board, or DSMB, provides safety oversight in the Phase 3 study.
Following its first two meetings, no concerning safety issues
were identified, and the DSMB has endorsed continuation of the
study as planned.

1

Clinical
Development of AeroVanc

Phase
3

Savara intends to initiate a
Phase 3 clinical study designed to demonstrate the safety and
efficacy of AeroVanc in cystic fibrosis, or CF, patients with
persistent methicillin-resistant Staphylococcus aurens,
or MRSA, lung infection. The plan is to initiate this trial in
the third quarter of 2017. The study is planned to be conducted
primarily in the United States and Canada.

Savara has received detailed
guidance from the FDA on the design of the study, and believes
that the planned study is in accordance with the FDAs
requirements for a sole pivotal study to be used in an NDA
submission. The study has also been planned in consultation with
the Cystic Fibrosis Foundations Therapeutic Development Network.
The Phase 3 study is designed to detect whether the
administration of AeroVanc results in a significant improvement
in lung function. The study will assess a 32 mg dose administered
twice a day for three on/off cycles of 28 days. The planned
primary efficacy endpoint is absolute change from baseline in FEV
percent predicted at Week 4
and Week 20, a commonly used measure of lung function. Secondary
efficacy endpoints include the time to use of other antibiotics
for pulmonary infection, FEV improvement (relative change, number of
response cycles) and a respiratory symptoms
diary.

The planned Phase 3 study is a
randomized (1:1), double-blind, placebo-controlled study of
AeroVanc in approximately 200 CF patients with persistent MRSA
lung infection. The plan is to enrich the study with younger
patients, by enrolling 75% of the subjects between the ages of 6
and 21 years. This was the population most responsive to
treatment in the Phase 2 study, and will form the primary
analysis population of the study. The duration of the study drug
(AeroVanc or placebo) administration will be three cycles of 28
days on drug and 28 days off drug, during which time the primary
efficacy endpoint will be measured and assessed. Following the
efficacy study period, subjects will transition into another
three cycles (28 days on treatment, 28 days off treatment per
cycle) of open label AeroVanc use to provide more information on
long-term safety.

The planned primary efficacy
endpoint of the study is the mean absolute change from baseline
in FEV percent predicted. In
accordance with guidance from the FDA, the endpoint will be
analyzed sequentially at Week 4 (first treatment cycle), and at
Week 20 (third treatment cycle). Both time points will be tested
at a statistical significance level of p = 0.05 due to the
sequential nature of the analysis. Savara believes that a
statistically significant improvement at Week 20 would provide
support for a chronic treatment label, whereas improvement at
Week 4 only may result in a more restricted label. Approval in
any form is subject to the positive evaluation of the clinical
meaningfulness of the treatment effect, judged by the review of
all data, including safety data, and the outcome of key secondary
endpoints, such as time to use of other antibiotics.

2

In the single-cycle Phase 2
study, with missing data imputed using conservative rules adopted
by the FDA, a difference in the mean absolute change in
FEVpercent predicted of 4.3%
was observed between the treatment arms in subjects below 21
years of age. Based on the observed treatment effect size and
variability, a sample size of 45 subjects per arm would provide
90% power to detect a statistically significant difference at an
alpha level of 0.05. To account for a potential loss of power
caused by premature discontinuations in a three-cycle study, a
sample size of 100 subjects per arm will be enrolled.

Selection of the dose for the
study was made based on the Phase 2 study in CF patients. In that
study, administration of the 32 mg bid dose resulted in sputum
trough vancomycin concentrations that were on average more than
100-fold above the observed minimum inhibitory concentration
(MIC90) value, suggesting that the concentrations reached after
repeated administration of the 32 mg bid dose are likely to be
sufficient for effective management of MRSA infection. In terms
of safety and tolerability, the 32 mg AeroVanc dose did not
appear significantly different from placebo, and produced
encouraging trends of efficacy in all key endpoints in subjects
below 21 years of age. In contrast, the higher AeroVanc dose of
64 mg bid was not as well tolerated in the older subjects (above
21 years of age), resulting in an increased number of premature
discontinuations of the study drug treatment in this
subgroup.

After the completion of the
Phase 3 study, Savara intends to submit an NDA under the
505(b)(2) regulatory pathway. In addition to being designated an
Orphan Drug Product and QIDP, AeroVanc has been designated a Fast
Track development program by the FDA.

Clinical
Development
of Aironite
We have supported, or currently are supporting, four
investigator-sponsored Phase 2 clinical studies of Aironite (also
known as AIR001) in patients with heart failure with preserved
ejection fraction (HFpEF) being conducted at prominent research
institutions in the United States.

Phase 2
INDIE-HFpEF Study

In 2016, Aironite was selected
by the Heart Failure Clinical Research Network, or HFN, for
evaluation in a multicenter, randomized, double-blind,
placebo-controlled crossover Phase 2 study in approximately 100
patients with HFpEF known as the Inorganic Nitrite Delivery to
Improve Exercise Capacity in HFpEF study, or the INDIE-HFpEF
study. The study began in the third quarter of 2016 and patient
enrollment is ongoing. Results are expected in the first half of
2018. The study is being conducted with significant support from
a grant awarded by the National Heart, Lung, and Blood Institute,
part of the National Institutes of Health. We are providing test
materials (Aironite and placebo), drug delivery devices
(nebulizers), regulatory and technical support, and some
additional financial support.

The study is a randomized,
double-blind, placebo-controlled crossover study to evaluate the
effect of AIR001 on peak exercise capacity as assessed by
cardiopulmonary exercise testing, or CPET.Approximately 100
patients with a diagnosis of HFpEF will be enrolled across
approximately 20 clinical centers in the United States that are
part of the HFN.The primary efficacy endpoint is the peak oxygen
consumption (VO2) after four weeks of treatment with Aironite or
placebo as assessed by CPET performed at peak drug levels.
Secondary objectives include (i) submaximal activity tolerance
chronically, (ii) quality of life, (iii) chronic filling
pressures as assessed by echocardiography and natriuretic peptide
levels, and/or (iv) ventilator efficiency or submaximal exercise
capacity at peak drug levels, and evaluation of the safety and
tolerability of Aironite.

Phase 2
INABLE-TRAINING Study

Enrollment is ongoing in a
Phase 2 clinical study of Aironite in patients with HFpEF known
as the Inorganic Nitrite to Amplify the Benefits and Tolerability
of Exercise Training study, or the INABLE-TRAINING study. This
randomized, blinded, placebo-controlled, two-arm, parallel-group
study in approximately 68 patients with HFpEF is evaluating
Aironites potential to improve the clinical responses to and
tolerability of exercise training, or ET, in individuals with
HFpEF. The primary endpoint of the study is the change in
exercise capacity as measured by peak oxygen consumption
(baseline to 12 weeks).

All study subjects will
undergo 12 weeks of ET. Subjects will be randomized to receive
Aironite three times daily or nebulized inhaled placebo (sodium
chloride) three times daily during the study period and will wear
accelerometry devices to track daily physical activity at home.
After 12 weeks of ET as part of standard cardiac rehabilitation,
study subjects will repeat the assessment of cardiovascular
function and exercise capacity as performed at study entry to
assess efficacy at a final visit.

The INABLE-TRAINING study has
2 aims. First, to determine whether treatment with Aironite in
addition to ET for 12 weeks improves exercise capacity and
hemodynamic reserve in HFpEF. Expired gas analysis, inert gas
(C2H2) rebreathe, and echocardiography will be performed during
rest and exercise to measure oxygen consumption (VO2), CO, and
hemodynamics before and after completion of 12 weeks of ET with
inhaled NO2- vs ET with inhaled placebo. Second, to determine
whether treatment with Aironite in addition to ET for 12 weeks
increases daily activity levels and quality of life, or QOL, and
reduces symptoms of effort intolerance during ET. Tolerability of
ET will be assessed by Borg perceived effort and dyspnea scores.
Large and small vessel endothelial function (brachial and digital
arteries) and QOL will also be assessed. Secondary endpoints
include cardiac output reserve, peak exercise workload, rest and
exercise hemodynamics assessed by echocardiography, Borg dyspnea
and fatigue scores recorded during ET, endothelial function
assessed by tonometry and brachial artery flow mediated dilation,
and QOL assessed by the Kansas City Cardiomyopathy
Questionnaire

Phase 1/2 Aironite
Study in Cystic Fibrosis Patients

An investigator-sponsored
open-label Phase 1/2 study of Aironite in adults with CF and
airway infection with Pseudomonas aeruginosa (P.
aeruginosa
) was initiated in January 2017 and is being
conducted at a prestigious research institution with funding from
a grant award from a non-profit organization. The study will
assess safety of Aironite administered in a dose escalation
manner. The study also aims to explore the effects of Aironite on
measures of lung function, exhaled airway nitric oxide, and
reduction in bacterial burden. Sodium nitrite has demonstrated
in vitro antimicrobial activity against P.
aeruginosa
and other airway pathogens, as well as the
ability to disperse biofilms. If supportive, we believe data from
this Phase 1/2 study would facilitate the design of a potential
Phase 2 program to establish Aironites utility as an
antimicrobial agent for P. aeruginosa and sensitizer to
standard antibiotic therapies in CF patients.

3

Government
Regulation

We believe our AeroVanc
product candidate will be regulated by the FDA as a drug and will
require the submission and approval of a New Drug Application, or
NDA, and our Molgradex product candidate will be regulated by the
FDA as a biologic and will require the submission and approval of
a Biologic License Application, or BLA.

The FDA and other regulatory
authorities at federal, state, and local levels, as well as in
foreign countries, extensively regulate, among other things, the
research, development, testing, manufacture, quality control,
import, export, safety, effectiveness, labeling, packaging,
storage, distribution, record keeping, approval, advertising,
promotion, marketing, post-approval monitoring, and post-approval
reporting of biologics and drugs, such as those Savara is
developing. Savara, along with third-party contractors, will be
required to navigate the various preclinical, clinical and
commercial approval requirements of the governing regulatory
agencies of the countries in which it wishes to conduct studies
or seek approval or licensure of its product candidates. The
process of obtaining regulatory approvals and the subsequent
compliance with appropriate federal, state, local, and foreign
statutes and regulations require the expenditure of substantial
time and financial resources.

BLA/NDA Approval
Process

The process required by the
FDA before a biologic or drug product candidate may be marketed
in the United States generally involves the following:

completion of preclinical laboratory tests and animal studies
performed in accordance with the FDAs current Good Laboratory
Practices, or GLP, regulation;
submission to the FDA of an IND, which must become effective
before clinical trials may begin and must be updated annually
or when significant changes are made;
approval by an independent Institutional Review Board, or
IRB, or ethics committee for each clinical site before a
clinical trial can begin;
performance of adequate and well-controlled human clinical
trials to establish the safety, purity and potency of the
proposed product candidate for its intended purpose;
preparation of and submission to the FDA of a Biologic
License Application, or BLA or New Drug Application, or NDA,
after completion of all required clinical trials;
a determination by the FDA within 60 days of its receipt of a
BLA or NDA to file the application for review;
satisfactory completion of an FDA Advisory Committee review,
if applicable;
satisfactory completion of an FDA pre-approval inspection of
the manufacturing facility or facilities at which the
proposed product is produced to assess compliance with
current Good Manufacturing Practices, or cGMP, and to assure
that the facilities, methods and controls are adequate to
preserve the products continued safety, purity and potency,
and of selected clinical investigational sites to assess
compliance with current Good Clinical Practices, or cGCPs;
and
FDA review and approval of the BLA or NDA to permit
commercial marketing of the product for particular
indications for use in the United States, which must be
updated annually and when significant changes are made.

The testing and approval
process requires substantial time, effort and financial
resources, and Savara cannot be certain that any approvals for
its product candidates will be granted on a timely basis, if at
all. Prior to beginning the first clinical trial with a product
candidate, Savara must submit an IND to the FDA. An IND is a
request for authorization from the FDA to administer an
investigational new drug product to humans. The central focus of
an IND submission is on the general investigational plan and the
protocol(s) for clinical studies. The IND also includes results
of animal and in vitro studies assessing the toxicology,
pharmacokinetics, pharmacology, and pharmacodynamic
characteristics of the product; chemistry, manufacturing, and
controls information; and any available human data or literature
to support the

4

use of the investigational
product. An IND must become effective before human clinical
trials may begin. The IND automatically becomes effective 30 days
after receipt by the FDA, unless the FDA, within the 30-day time
period, raises safety concerns or questions about the proposed
clinical trial. In such a case, the IND may be placed on clinical
hold and the IND sponsor and the FDA must resolve any outstanding
concerns or questions before the clinical trial can begin.
Submission of an IND therefore may or may not result in FDA
authorization to begin a clinical trial.

Clinical trials involve the
administration of the investigational product to human subjects
under the supervision of qualified investigators in accordance
with cGCPs, which include the requirement that all research
subjects provide their informed consent for their participation
in any clinical study. Clinical trials are conducted under
protocols detailing, among other things, the objectives of the
study, the parameters to be used in monitoring safety and the
effectiveness criteria to be evaluated. A separate submission to
the existing IND must be made for each successive clinical trial
conducted during product development and for any subsequent
protocol amendments. Furthermore, an independent Institutional
Review Board, or IRB, for each site proposing to conduct the
clinical trial must review and approve the plan for any clinical
trial and its informed consent form before the clinical trial
begins at that site, and must monitor the study until completed.
Regulatory authorities, the IRB or the sponsor may suspend a
clinical trial at any time on various grounds, including a
finding that the subjects are being exposed to an unacceptable
health risk or that the trial is unlikely to meet its stated
objectives. Some studies also include oversight by an independent
group of qualified experts organized by the clinical study
sponsor, known as a data safety monitoring board, which provides
authorization for whether or not a study may move forward at
designated check points based on access to certain data from the
study and may halt the clinical trial if it determines that there
is an unacceptable safety risk for subjects or other grounds,
such as no demonstration of efficacy. There are also requirements
governing the reporting of ongoing clinical studies and clinical
study results to public registries.

Clinical trials typically are
conducted in three or four sequential phases, but the phases my
overlap or be combined.

Phase 1. The drug product is initially introduced
into healthy human subjects and tested for safety. In the
case of some products for severe or life-threatening
diseases, the initial human testing is often conducted in
patients.
Phase 2. The drug product is evaluated in a limited
patient population to identify possible adverse effects and
safety risks, to preliminarily evaluate the efficacy of the
product for specific targeted diseases and to determine
dosage tolerance, optimal dosage and dosing schedule.
Phase 3. Clinical trials are undertaken to further
evaluate dosage, clinical efficacy, potency, and safety in an
expanded patient population at geographically dispersed
clinical trial sites. These clinical trials are intended to
establish the overall risk to benefit ratio of the product
and provide an adequate basis for product labeling.
Phase 4. In some cases, the FDA may require, or
companies may voluntarily pursue, additional clinical trials
after a product is approved to gain more information about
the product. These so-called Phase 4 studies
may be required as a condition to approval of the BLA or
NDA.

Phase 1, Phase 2 and Phase 3
testing may not be completed successfully within a specified
period, if at all, and there can be no assurance that the data
collected will support FDA approval or licensure of the product.
Concurrent with clinical trials, companies may complete
additional animal studies and develop additional information
about the drug characteristics of the product candidate, and must
finalize a process for manufacturing the product in commercial
quantities in accordance with cGMP requirements. The
manufacturing process must be capable of consistently producing
quality batches of the product candidate. Additionally,
appropriate packaging must be selected and tested and stability
studies must be conducted to demonstrate that the product
candidate does not undergo unacceptable deterioration over its
shelf life. Assuming successful completion of all required
testing in accordance with all applicable regulatory
requirements, the results of product development, nonclinical
studies and clinical trials are submitted to the FDA as part of a
BLA or NDA requesting approval to market the product for one or
more indications. The BLA or NDA must include all relevant data
available from pertinent preclinical and clinical studies,
including negative or ambiguous results as well as positive
findings, together with detailed information relating to the
products chemistry, manufacturing, controls, and proposed
labeling, among other things. Data can come from
company-sponsored clinical studies intended to test the safety
and effectiveness of a use of the product, or from a number of
alternative sources, including studies initiated by
investigators. The submission of a BLA or NDA requires payment of
a substantial User Fee to FDA, and the sponsor of an approved NDA
is also subject to annual product and establishment user fees.
These fees are typically increased annually. A waiver of user
fees may be obtained under certain limited circumstances.

5

Within 60 days following
submission of the application, the FDA reviews the BLA or NDA to
determine if it is substantially complete before the agency
accepts it for filing. The FDA may refuse to file any BLA or NDA
that it deems incomplete or not properly reviewable at the time
of submission and may request additional information. In this
event, the BLA or NDA must be resubmitted with the additional
information. Once a BLA or NDA has been filed, the FDAs goal is
to review the application within ten months after it accepts the
application for filing, or, if the application relates to an
unmet medical need in a serious or life-threatening indication,
six months after the FDA accepts the application for filing. The
review process is often significantly extended by FDA requests
for additional information or clarification. The FDA reviews a
BLA or NDA to determine, among other things, whether a product is
safe and effective for the indication being pursued, and the
facility in which it is manufactured, processed, packed, or held
meets standards designed to assure the products continued safety
and effectiveness. The FDA may convene an advisory committee to
provide clinical insight on application review questions. Before
approving a BLA or NDA, the FDA will typically inspect the
facility or facilities where the product is manufactured. The FDA
will not approve an application unless it determines that the
manufacturing processes and facilities are in compliance with
cGMP requirements and adequate to assure consistent production of
the product within required specifications. Additionally, before
approving a BLA or NDA, the FDA will typically inspect one or
more clinical sites to assure compliance with cGCP. If the FDA
determines that the application, manufacturing process or
manufacturing facilities are not acceptable, it will outline the
deficiencies in the submission and often will request additional
testing or information. Notwithstanding the submission of any
requested additional information, the FDA ultimately may decide
that the application does not satisfy the regulatory criteria for
approval.

The testing and approval
process requires substantial time, effort and financial
resources, and each may take several years to complete. The FDA
may not grant approval on a timely basis, or at all, and Savara
may encounter difficulties or unanticipated costs in its efforts
to secure necessary governmental approvals, which could delay or
preclude us from marketing its products. After the FDA evaluates
a BLA or NDA and conducts inspections of manufacturing facilities
where the investigational product and/or its drug substance will
be produced, the FDA may issue an approval letter or a Complete
Response Letter. An approval letter authorizes commercial
marketing of the product with specific prescribing information
for specific indications. A Complete Response Letter indicates
that the review cycle of the application is complete and the
application is not ready for approval. A Complete Response Letter
may request additional information or clarification. The FDA may
delay or refuse approval of a BLA or NDA if applicable regulatory
criteria are not satisfied, require additional testing or
information and/or require post-marketing testing and
surveillance to monitor safety or efficacy of a product.

If regulatory approval of a
product is granted, such approval may entail limitations on the
indicated uses for which such product may be marketed. For
example, the FDA may approve the BLA or NDA with a Risk
Evaluation and Mitigation Strategy, or REMS, plan to mitigate
risks, which could include medication guides, physician
communication plans, or elements to assure safe use, such as
restricted distribution methods, patient registries and other
risk minimization tools. The FDA also may condition approval on,
among other things, changes to proposed labeling or the
development of adequate controls and specifications. Once
approved, the FDA may withdraw the product approval if compliance
with pre- and post-marketing regulatory standards is not
maintained or if problems occur after the product reaches the
marketplace. The FDA may require one or more Phase 4 post-market
studies and surveillance to further assess and monitor the
products safety and effectiveness after commercialization, and
may limit further marketing of the product based on the results
of these post-marketing studies. In addition, new government
requirements, including those resulting from new legislation, may
be established, or the FDAs policies may change, which could
delay or prevent regulatory approval of its products under
development.

A sponsor may seek approval of
its product candidate under programs designed to accelerate FDAs
review and approval of new drugs that meet certain criteria.
Specifically, new drug products are eligible for fast track
designation if they are intended to treat a serious or
life-threatening condition and demonstrate the potential to
address unmet medical needs for the condition. For a fast track
product, the FDA may consider sections of the BLA or NDA for
review on a rolling basis before the complete application is
submitted if relevant criteria are met. A fast track designated
product candidate may also qualify for priority review, under
which the FDA sets the target date for FDA action on the BLA
or

6

NDA at six months after the
FDA accepts the application for filing. Priority review is
granted when there is evidence that the proposed product would be
a significant improvement in the safety or effectiveness of the
treatment, diagnosis, or prevention of a serious condition. If
criteria are not met for priority review, the application is
subject to the standard FDA review period of 10 months after FDA
accepts the application for filing. Priority review designation
does not change the scientific/medical standard for approval or
the quality of evidence necessary to support approval.

Under the accelerated approval
program, the FDA may approve a BLA or NDA on the basis of either
a surrogate endpoint that is reasonably likely to predict
clinical benefit, or on a clinical endpoint that can be measured
earlier than irreversible morbidity or mortality, that is
reasonably likely to predict an effect on irreversible morbidity
or mortality or other clinical benefit, taking into account the
severity, rarity, or prevalence of the condition and the
availability or lack of alternative treatments. Post-marketing
studies or completion of ongoing studies after marketing approval
are generally required to verify the biologics or drugs clinical
benefit in relationship to the surrogate endpoint or ultimate
outcome in relationship to the clinical benefit. In addition, the
Food and Drug Administration Safety and Innovation Act, or
FDASIA, which was enacted and signed into law in 2012,
established breakthrough therapy designation. A sponsor may seek
FDA designation of its product candidate as a breakthrough
therapy if the product candidate is intended, alone or in
combination with one or more other drugs or biologics, to treat a
serious or life-threatening disease or condition and preliminary
clinical evidence indicates that the therapy may demonstrate
substantial improvement over existing therapies on one or more
clinically significant endpoints, such as substantial treatment
effects observed early in clinical development. Sponsors may
request the FDA to designate a breakthrough therapy at the time
of or any time after the submission of an IND, but ideally before
an end-of-phase 2
meeting with FDA. If the FDA designates a breakthrough therapy,
it may take actions appropriate to expedite the development and
review of the application, which may include holding meetings
with the sponsor and the review team throughout the development
of the therapy; providing timely advice to, and interactive
communication with, the sponsor regarding the development of the
biologic or drug to ensure that the development program to gather
the nonclinical and clinical data necessary for approval is as
efficient as practicable; involving senior managers and
experienced review staff, as appropriate, in a collaborative,
cross-disciplinary review; assigning a cross-disciplinary project
lead for the FDA review team to facilitate an efficient review of
the development program and to serve as a scientific liaison
between the review team and the sponsor; and considering
alternative clinical trial designs when scientifically
appropriate, which may result in smaller or more efficient
clinical trials that require less time to complete and may
minimize the number of patients exposed to a potentially less
efficacious treatment. Breakthrough designation also allows the
sponsor to file sections of the BLA or NDA for review on a
rolling basis. Savara may seek designation as a breakthrough
therapy for some or all of its product candidates.

Fast Track designation,
priority review and breakthrough therapy designation do not
change the standards for approval but may expedite the
development or approval process.

Orphan Drug
Status

Under the Orphan Drug Act, the
FDA may grant orphan drug designation to drug candidates intended
to treat a rare disease or condition, which is generally a
disease or condition that affects fewer than 200,000 individuals
in the United States, or more than 200,000 individuals in the
United States and for which there is no reasonable expectation
that costs of research and development of the drug for the
indication can be recovered by sales of the drug in the United
States. Orphan drug designation must be requested before
submitting a BLA or NDA. After the FDA grants orphan drug
designation, the generic identity of the therapeutic agent and
its potential orphan use are disclosed publicly by the FDA.
Although there may be some increased communication opportunities,
orphan drug designation does not convey any advantage in or
shorten the duration of the regulatory review and approval
process.

If a biologic or drug
candidate that has orphan drug designation subsequently receives
the first FDA approval for the disease for which it has such
designation, the product is entitled to orphan exclusivity, which
means that the FDA may not approve any other applications,
including a full BLA or NDA, to market the same drug for the same
indication for seven years, except in very limited circumstances,
such as if the second applicant demonstrates the clinical
superiority of its product or if FDA finds that the holder of the
orphan exclusivity has not shown that it can assure the
availability of sufficient quantities of the orphan drug to meet
the needs of patients with the disease or condition for which the
biologic or drug was designated. Orphan exclusivity does not
prevent the FDA from approving a different biologic or drug for
the same disease or condition, or the same biologic or drug for a
different disease or condition. Among the other benefits of
orphan designation are tax credits for certain research and a
waiver of the BLA or NDA application user fee.

7

Orphan exclusivity could block
the approval of Savaras biologic or drug candidates for seven
years if a competitor obtains approval of the same product as
defined by the FDA or if Savaras biologic or drug candidate is
determined to be contained within the competitors product for the
same indication or disease.

As in the United States,
designation as an orphan product for the treatment of a specific
indication in the European Union, must be made before the
application for marketing authorization is made. Orphan products
in Europe enjoy economic and marketing benefits, including up to
10 years of market exclusivity for the approved indication unless
another applicant can show that its product is safer, more
effective or otherwise clinically superior to the orphan
designated product.

The FDA and foreign regulators
expect holders of exclusivity for orphan biologics or drugs to
assure the availability of sufficient quantities of their orphan
biologic or drugs to meet the needs of patients. Failure to do so
could result in the withdrawal of marketing exclusivity for the
orphan biologic or drug.

GAIN Exclusivity
for Antibiotics

In 2012, Congress passed
legislation known as the Generating Antibiotic Incentives Now
Act, or GAIN Act. This legislation is designed to encourage the
development of antibacterial and antifungal drug products that
treat pathogens that cause serious and life-threatening
infections. To that end, the new law grants an additional five
years of exclusivity upon the approval of an NDA for a drug
product designated by FDA as a QIDP. Thus, for a QIDP with Orphan
Designation, the periods of five-year exclusivity and seven-year
orphan drug exclusivity, would become 12 years.

A QIDP is defined in the GAIN
Act to mean an antibacterial or antifungal drug for human use
intended to treat serious or life-threatening infections,
including those caused by (1)an antibacterial or antifungal
resistant pathogen, including novel or emerging infectious
pathogens or (2)certain qualifying pathogens. A qualifying
pathogen is a pathogen that has the potential to pose a serious
threat to public health (such as resistant Gram-positive
pathogens, multi-drug resistant Gram-negative bacteria,
multi-drug resistant tuberculosis, and C. difficile) and that is
included in a list established and maintained by FDA. A drug
sponsor may request the FDA to designate its product as a QIDP
any time before the submission of an NDA. The FDA must make a
QIDP determination within 60 days of the designation request. A
product designated as a QIDP will be granted priority review by
the FDA and can qualify for fast track status.

The additional five years of
exclusivity under the GAIN Act for drug products designated by
the FDA as QIDPs applies only to a drug that is first approved on
or after July9, 2012. Additionally, the five year exclusivity
extension does not apply to: a supplement to an application under
FDCA Section505(b) for any QIDP for which an extension is in
effect or has expired; a subsequent application filed with
respect to a product approved by the FDA for a change that
results in a new indication, route of administration, dosing
schedule, dosage form, delivery system, delivery device or
strength; or a product that does not meet the definition of a
QIDP under Section505(g) based upon its approved
uses.

Abbreviated
Licensure Pathway for Biological Products as Biosimilar or
Interchangeable

The Patient Protection and
Affordable Care Act, or the ACA, signed into law in March, 2010,
includes a subtitle called the Biologics Price Competition and
Innovation Act of 2009, or BPCIA, which amended the Public Health
Services Act and created an abbreviated approval pathway for
biological products shown to be highly similar to an FDA-licensed
reference biological product. The BPCIA attempts to minimize
duplicative testing, and thereby lower development costs and
increase patient access to affordable treatments.

The timing of final FDA
approval of a biosimilar for commercial distribution depends on a
variety of factors, including whether the manufacturer of the
branded product is entitled to one or more statutory exclusivity
periods, during which time the FDA is prohibited from approving
any products that are biosimilar to the branded product. The FDA
cannot approve a biosimilar application for twelve years from the
date of first licensure of the reference product. Additionally, a
biosimilar product sponsor may not submit an biosimilar
application for four years from the date of first

8

licensure of the reference
product. A reference product may also be entitled to exclusivity
under other statutory provisions. For example, a reference
product designated for a rare disease or condition (an orphan
drug) may be entitled to seven years of exclusivity, in which
case no product that is biosimilar to the reference product may
be approved until either the end of the twelve-year period
provided under the BPCIA or the end of the seven-year orphan
exclusivity period, whichever occurs later. In addition, the FDA
may under certain circumstances extend the exclusivity period for
the reference product by an additional six months if the FDA
requests, and the manufacturer undertakes, studies on the effect
of its product in children, a so-called pediatric
extension.

The first biological product
determined to be interchangeable with a branded product for any
condition of use is also entitled to a period of exclusivity,
during which time the FDA may not determine that another product
is interchangeable with the reference product for any condition
of use. This exclusivity period extends until the earlier of:
(1)on year after the first commercial marketing of the first
interchangeable product; (2) 18 months after resolution of a
patent infringement suit instituted against the applicant that
submitted the application for the first interchangeable product,
based on a final court decision regarding all of the patents in
the litigation or dismissal of the litigation with or without
prejudice; (3) 42 months after approval of the first
interchangeable product, if a patent infringement suit instituted
against the applicant that submitted the application for the
first interchangeable product is still ongoing; or (4) 18 months
after approval of the first interchangeable product if the
applicant that submitted the application for the first
interchangeable product has not been sued.

505(b)(2) New Drug
Application

The provisions of section
505(b)(2) of the Food, Drug, and Cosmetic Act, or the FDCA, were
created, in part, to help avoid unnecessary duplication of
studies already performed on a previously approved (reference or
listed) drug; the section gives the FDA express permission to
rely on data not developed by the NDA applicant. An NDA filed
under section 505(b)(2) is one for which one or more of the
investigations relied upon by the applicant for approval were not
conducted by or for the applicant and for which the applicant has
not obtained a right of reference or use from the person by or
for whom the investigations were conducted. We are pursuing a
section 505(b)(2) regulatory strategy for AeroVanc.

The owner of an NDA for a
branded drug product may list with the FDA certain patents whose
claims allegedly cover the applicants branded product. Each of
the patents listed in the application for the drug is then
published in the Orange Book. Any applicant who files a 505(b)(2)
NDA application referencing a drug listed in the Orange Book must
certify to the FDA that: (1)no patent information on the drug
product that is the subject of the application has been submitted
to the FDA, referred to as a Paragraph I Certification; (2)such
patent has expired, referred to as a Paragraph II Certification;
(3)the date on which such patent expires, referred to ad a
Paragraph III Certification; or (4)such patent is invalid or will
not be infringed upon by the manufacture, use or sale of the drug
product for which the application is submitted, referred to as a
Paragraph IV Certification. The applicant may also elect to
submit a section viii statement certifying that its proposed
label does not contain, or carves out, any language regarding the
patented method-of use rather than certify to a listed
method-of-use patent. An applicant submitting a Paragraph IV
Certification must provide notice to each owner of the patent
that is the subject of the certification and to the holder of the
approved branded drug to which the 505(b)(2) application refers.
If the reference branded drug holder and patent owners assert a
patent challenge directed to one of the Orange Book listed
patents within 45 days of the receipt of the Paragraph IV
Certification notice, the FDA is prohibited from approving the
505(b)(2) application until the earlier of 30 months from the
receipt of the Paragraph IV Certification, expiration for the
patent, settlement of the lawsuit, or a decision in the
infringement case that is favorable to the
applicant.

Additionally, a 505(b)(2)
application also will not be approved until any applicable
non-patent exclusivity listed in the Orange Book for the NDA
branded reference drug has expired as described in further detail
below. Market and data exclusivity provisions under the FDCA can
delay the submission or the approval of certain applications for
competing products. In addition to patent exclusivities, the
holder of the NDA for a reference listed drug may be entitled to
a period of non-patent exclusivity, during which the FDA cannot
approve another drug application that relies on the listed drug.
For example, a pharmaceutical manufacturer may obtain five years
of non-patent
exclusivity upon NDA approval of a new chemical entity, or NCE,
which is a drug that contains an active moiety that has not been
approved by the FDA in any other NDA. An active moiety is defined
as the molecule or ion responsible for the drug substances
physiological

9

or pharmacologic action.
During the five-year exclusivity period, the FDA cannot accept
for filing any application for the same active moiety and that
relies on the FDAs findings regarding that drug; the FDA may
accept an application for filing after four years if the
follow-on applicant makes a Paragraph IV Certification. A drug
may obtain a three-year period of exclusivity for a particular
condition of approval, or change to a marketed product, such as a
new formulation for a previously approved product, if one or more
new clinical trials, other than bioavailability or bioequivalence
studies, was essential to the approval of the application and was
conducted or sponsored by the applicant. Should this occur, the
FDA would be precluded from approving any Abbreviated New Drug
Application, or ANDA, that references such product until after
that three-year exclusivity period has run. However, unlike NCE
exclusivity, the FDA can accept an application and begin the
review process during the entire exclusivity
period.

Post-Approval
Requirements

Any products manufactured or
distributed by us to FDA approvals are subject to pervasive and
continuing regulation by the FDA, including, among other things,
requirements relating to record-keeping, reporting of adverse
experiences, periodic reporting, distribution, and advertising
and promotion of the product. After approval, most changes to the
approved product, such as adding new indications or other
labeling claims, are subject to prior FDA review and approval.
There also are continuing, annual user fee requirements for any
marketed products and the establishments at which such products
are manufactured, as well as new application fees for
supplemental applications with clinical data. Drug manufacturers
and their subcontractors are required to register their
establishments with the FDA and certain state agencies, and are
subject to periodic unannounced inspections by the FDA and
certain state agencies for compliance with GMP, which impose
certain procedural and documentation requirements upon Savara and
its third-party manufacturers. Changes to the manufacturing
process are strictly regulated, and, depending on the
significance of the change, may require prior FDA approval before
being implemented. FDA regulations also require investigation and
correction of any deviations from cGMP and impose reporting
requirements upon us and any third-party manufacturers that
Savara may decide to use. Accordingly, manufacturers must
continue to expend time, money and effort in the area of
production and quality control to maintain compliance with cGMP
and other aspects of regulatory compliance. Savara cannot be
certain that it or its present or future suppliers will be able
to comply with the cGMP regulations and other FDA regulatory
requirements. If Savaras present or future suppliers are not able
to comply with these requirements, the FDA may, among other
things, halt its clinical trials, require them to recall a
product from distribution, or withdraw approval of the
NDA.

Future FDA and state
inspections may identify compliance issues at Savaras facilities
or at the facilities of its contract manufacturers that may
disrupt production or distribution, or require substantial
resources to correct. In addition, discovery of previously
unknown problems with a product or the failure to comply with
applicable requirements may result in restrictions on a product,
manufacturer or holder of an approved NDA, including withdrawal
or recall of the product from the market or other voluntary,
FDA-initiated or judicial action that could delay or prohibit
further marketing.

The FDA may withdraw approval
of an NDA if compliance with regulatory requirements and
standards is not maintained or if problems occur after the
product reaches the market. Later discovery of previously unknown
problems with a product, including adverse events of
unanticipated severity or frequency, or with manufacturing
processes, or failure to comply with regulatory requirements, may
result in revisions to the approved labeling to add new safety
information; imposition of post-market studies or clinical
studies to assess new safety risks; or imposition of distribution
restrictions or other restrictions under a REMS program. Other
potential consequences include, among other
things:

restrictions on the marketing or manufacturing of the
product, complete withdrawal of the product from the market
or product recalls;
fines, warning letters or holds on post-approval clinical
studies;
refusal of the FDA to approve pending applications or
supplements to approved applications, or suspension or
revocation of product license approvals;
product seizure or detention, or refusal to permit the import
or export of products; or
injunctions or the imposition of civil or criminal penalties.

10

The FDA closely regulates the
marketing, labeling, advertising and promotion of drugs and
biologics. A company can make only those claims relating to
safety and efficacy that are approved by the FDA and in
accordance with the provisions of the approved label. The FDA and
other agencies actively enforce the laws and regulations
prohibiting the promotion of off-label uses. Failure to comply
with these requirements can result in, among other things,
adverse publicity, warning letters, corrective advertising and
potential civil and criminal penalties. Physicians may prescribe
legally available products for uses that are not described in the
products labeling and that differ from those tested by Savara and
approved by the FDA. Such off-label uses are common across
medical specialties. Physicians may believe that such off-label
uses are the best treatment for many patients in varied
circumstances. The FDA does not regulate the behavior of
physicians in their choice of treatments. The FDA does, however,
restrict manufacturers communications on the subject of off-label
use of their products.

Government
Regulationof Combination
Products

Savaras products under
development will be regulated as combination products, which
means that they are comprised of two or more different components
that, if marketed individually, would be subject to different
regulatory paths and would require approval of independent
marketing applications by the FDA. A combination product,
however, is assigned to a Center with the FDA that will have
primary jurisdiction over its regulation on a determination of
the combination products primary mode of action, which is the
single mode of action that provides the most important
therapeutic action. Savara believes its product candidates
include both a drug and medical device component, and will be
regulated as a drug, subject to the review of the FDAs Center for
Drug Evaluation and Research, or CDER, which will have primary
jurisdiction over premarket development and approval. FDAs Center
for Devices and Radiological Health, or CDRH, will provide
support and review of the inhaler component of the product
candidate.

Other Healthcare
Laws and Compliance
Requirements

Savaras sales, promotion,
medical education, clinical research and other activities
following product approval will be subject to regulation by
numerous regulatory and law enforcement authorities in the United
States in addition to FDA, including potentially the Federal
Trade Commission, the Department of Justice, the Centers for
Medicare and Medicaid Services, or CMS, other divisions of the
U.S. Department of Health and Human Services and state and local
governments. Savaras promotional and scientific/educational
programs and interactions with healthcare professionals must
comply with the federal Anti-Kickback Statute, the civil False
Claims Act, physician payment transparency laws, privacy laws,
security laws, and additional federal and state laws similar to
the foregoing.

The federal Anti-Kickback
Statute prohibits, among other things, the knowing and willing,
direct or indirect offer, receipt, solicitation or payment of
remuneration in exchange for or to induce the referral of
patients, including the purchase, order or lease of any good,
facility, item or service that would be paid for in whole or part
by Medicare, Medicaid or other federal health care programs.
Remuneration has been broadly defined to include anything of
value, including cash, improper discounts, and free or reduced
price items and services. The federal Anti-Kickback Statute has
been interpreted to apply to arrangements between pharmaceutical
manufacturers on one hand and prescribers, purchasers, formulary
managers, and beneficiaries on the other. Although there are a
number of statutory exceptions and regulatory safe harbors
protecting some common activities from prosecution, the
exceptions and safe harbors are drawn narrowly. Practices that
involve remuneration that may be alleged to be intended to induce
prescribing, purchases or recommendations may be subject to
increased scrutiny and review if they do not qualify for an
exception or safe harbor. Failure to meet all of the requirements
of a particular applicable statutory exception or regulatory safe
harbor does not make the conduct per se illegal under the federal
Anti-Kickback Statute. Instead, the legality of the arrangement
will be evaluated on a case-by-case basis based on a cumulative
review of all its facts and circumstances. Several courts have
interpreted the statutes intent requirement to mean that if any
one purpose of an arrangement involving remuneration is to induce
referrals of federal healthcare covered business, the federal
Anti-Kickback Statute has been violated. The government has
enforced the federal Anti-Kickback Statute to reach large
settlements with healthcare companies based on sham research or
consulting and other financial arrangements with physicians.
Further, a person or entity does not need to have actual
knowledge of the statute or specific intent to violate it to have
committed a violation. In addition, the government may assert
that a claim including items or services resulting from a
violation of the federal Anti-Kickback Statute constitutes a
false or fraudulent claim for purposes of the False Claims Act.
Many states have similar laws that apply to their state health
care programs as well as private payers.

11

Federal false claims and false
statement laws, including the federal civil False Claims Act, or
FCA, imposes liability on persons and/or entities that, among
other things, knowingly present or cause to be presented claims
that are false or fraudulent or not provided as claimed for
payment or approval by a federal health care program. The FCA has
been used to prosecute persons or entities that cause the
submission of claims for payment that are inaccurate or
fraudulent, by, for example, providing inaccurate billing or
coding information to customers, promoting a product off-label, submitting claims
for services not provided as claimed, or submitting claims for
services that were provided but not medically necessary. Actions
under the FCA may be brought by the Attorney General or as a qui
tam action by a private individual, or whistleblower, in the name
of the government. Violations of the FCA can result in
significant monetary penalties and treble damages. The federal
government is using the FCA, and the accompanying threat of
significant liability, in its investigation and prosecution of
pharmaceutical and biotechnology companies throughout the
country, for example, in connection with the promotion of
products for unapproved uses and other illegal sales and
marketing practices. The government has obtained multi-million
and multi-billion dollar settlements under the FCA in addition to
individual criminal convictions under applicable criminal
statutes. In addition, certain companies that were found to be in
violation of the FCA have been forced to implement extensive
corrective action plans, and have often become subject to consent
decrees or corporate integrity agreements, restricting the manner
in which they conduct their business.

The federal Health Insurance
Portability and Accountability Act of 1996, or HIPAA, created
additional federal criminal statutes that prohibit, among other
things, knowingly and willfully executing, or attempting to
execute, a scheme to defraud any healthcare benefit program,
including private third-party payers; knowingly and willfully
falsifying, concealing or covering up a material fact or making
any materially false, fictitious or fraudulent statement in
connection with the delivery of or payment for healthcare
benefits, items or services; and willfully obstructing a criminal
investigation of a healthcare offense. Like the federal
Anti-Kickback Statute, the Affordable Care Act amended the intent
standard for certain healthcare fraud statutes under HIPAA such
that a person or entity no longer needs to have actual knowledge
of the statute or specific intent to violate it in order to have
committed a violation.

Given the significant size of
actual and potential settlements, it is expected that the
government will continue to devote substantial resources to
investigating healthcare providers and manufacturers compliance
with applicable fraud and abuse laws. Also, many states have
similar fraud and abuse statutes or regulations that may be
broader in scope and may apply regardless of payer, in addition
to items and services reimbursed under Medicaid and other state
programs. Additionally, to the extent that Savaras products, once
commercialized, are sold in a foreign country, Savara may be
subject to similar foreign laws.

In addition, there has been a
recent trend of increased federal and state regulation of
payments made to physicians and other healthcare providers. The
Patient Protection and Affordable Care Act, as amended by the
Health Care and Education Reconciliation Act, or collectively,
the Affordable Care Act, among other things, imposed new
reporting requirements on certain manufacturers of drugs,
devices, biologics and medical supplies for which payment is
available under Medicare, Medicaid or the Childrens Health
Insurance Program, with specific exceptions, for payments or
other transfers of value made by them to physicians and teaching
hospitals, as well as ownership and investment interests held by
physicians and their immediate family members. Covered
manufacturers are required to collect and report detailed payment
data and submit legal attestation to the accuracy of such data to
the government each year. Failure to submit required information
may result in civil monetary penalties of up to an aggregate of
$150,000 per year (or up to an aggregate of $1million per year
for knowing failures), for all payments, transfers of value or
ownership or investment interests that are not timely, accurately
and completely reported in an annual submission. Additionally,
entities that do not comply with mandatory reporting requirements
may be subject to a corporate integrity agreement. Certain states
also mandate implementation of commercial compliance programs,
impose restrictions on covered manufacturers marketing practices
and/or require the tracking and reporting of gifts, compensation
and other remuneration to physicians and other healthcare
professionals.

Savara may also be subject to
data privacy and security regulation by both the federal
government and the states in which it conducts its business.
HIPAA, as amended by the Health Information Technology and
Clinical Health Act, or HITECH, and their respective implementing
regulations, imposes specified requirements on certain health
care providers, plans and clearinghouses (collectively, covered
entities) and their business associates, relating to the privacy,
security and transmission of individually identifiable health
information. Among other things, HITECH makes
HIPAAs

12

security standards directly
applicable to business associates, defined as independent
contractors or agents of covered entities that create, receive,
maintain or transmit protected health information in connection
with providing a service for or on behalf of a covered entity.
HITECH also increased the civil and criminal penalties that may
be imposed against covered entities, business associates and
possibly other persons, and gave state attorneys general new
authority to file civil actions for damages or injunctions in
federal courts to enforce HIPAA and seek attorneys fees and costs
associated with pursuing federal civil actions. In addition,
certain states have their own laws that govern the privacy and
security of health information in certain circumstances, many of
which differ from each other and/or HIPAA in significant ways and
may not have the same effect, thus complicating compliance
efforts.

If Savaras operations are
found to be in violation of any of such laws or any other
governmental regulations that apply to them, Savara may be
subject to penalties, including, without limitation, civil and
criminal penalties, damages, fines, disgorgement, the curtailment
or restructuring of its operations, exclusion from participation
in federal and state healthcare programs, imprisonment,
contractual damages, reputational harm, and diminished profits
and future earnings, any of which could adversely affect its
ability to operate its business and its financial
results.

In addition to the foregoing
health care laws, Savara is also subject to the U.S. Foreign
Corrupt Practices Act and similar worldwide anti-bribery laws,
which generally prohibit companies and their intermediaries from
making improper payments to government officials or
private-sector recipients for the purpose of obtaining or
retaining business. Savara has plans to adopt an anti-corruption
policy, which will become effective upon the completion of this
offering, and expect to prepare and implement procedures to
ensure compliance with such policy. The anti-corruption policy
mandates compliance with the FCPA and similar anti-bribery laws
applicable to its business throughout the world. However, Savara
cannot assure you that such a policy or procedures implemented to
enforce such a policy will protect them from intentional,
reckless or negligent acts committed by its employees,
distributors, partners, collaborators or agents. Violations of
these laws, or allegations of such violations, could result in
fines, penalties or prosecution and have a negative impact on its
business, results of operations and
reputation.

Coverage and
Reimbursement

Sales of pharmaceutical
products depend significantly on the extent to which coverage and
adequate reimbursement are provided by third-party payers.
Third-party payers include state and federal government health
care programs, managed care providers, private health insurers
and other organizations. Although Savara currently believes that
third-party payers will provide coverage and reimbursement for
its product candidates, if approved, Savara cannot be certain of
this. Third-party payers are increasingly challenging the price,
examining the cost-effectiveness, and reducing reimbursement for
medical products and services. In addition, significant
uncertainty exists as to the reimbursement status of newly
approved healthcare products. The U.S. government, state
legislatures and foreign governments have continued implementing
cost containment programs, including price controls, restrictions
on coverage and reimbursement and requirements for substitution
of generic products. Adoption of price controls and cost
containment measures, and adoption of more restrictive policies
in jurisdictions with existing controls and measures, could
further limit Savaras net revenue and results. Savara may need to
conduct expensive clinical studies to demonstrate the comparative
cost-effectiveness of its products. The product candidates that
Savara develops may not be considered cost-effective and thus may
not be covered or sufficiently reimbursed. It is time consuming
and expensive for them to seek coverage and reimbursement from
third-party payers, as each payer will make its own determination
as to whether to cover a product and at what level of
reimbursement. Thus, one payers decision to provide coverage and
adequate reimbursement for a product does not assure that another
payer will provide coverage or that the reimbursement levels will
be adequate. Moreover, a payers decision to provide coverage for
a drug product does not imply that an adequate reimbursement rate
will be approved. Reimbursement may not be available or
sufficient to allow them to sell its products on a competitive
and profitable basis.

Healthcare
Reform

The United States and some
foreign jurisdictions are considering or have enacted a number of
legislative and regulatory proposals to change the healthcare
system in ways that could materially affect Savaras ability to
sell its products profitably. Among policy makers and payers in
the United States and elsewhere, there is significant interest in
promoting changes in healthcare systems with the stated goals of
containing healthcare costs, improving quality and/or expanding
access. In the United States, the pharmaceutical industry has
been a particular focus of these efforts and has been
significantly affected by major legislative
initiatives.

13

By way of example, in
March2010, the Affordable Care Act was signed into law, intended
to broaden access to health insurance, reduce or constrain the
growth of healthcare spending, enhance remedies against fraud and
abuse, add new transparency requirements for the healthcare and
health insurance industries, impose new taxes and fees on the
health industry and impose additional health policy reforms.
Among the provisions of the Affordable Care Act of importance to
Savaras potential drug candidates are:

an annual, nondeductible fee on any entity that manufactures
or imports specified branded prescription drugs and biologic
agents, apportioned among these entities according to their
market share in certain government healthcare programs;
an increase in the statutory minimum rebates a manufacturer
must pay under the Medicaid Drug Rebate Program to 23.1% and
13.0% of the average manufacturer price for branded and
generic drugs, respectively;
a new methodology by which rebates owed by manufacturers
under the Medicaid Drug Rebate Program are calculated for
drugs that are inhaled, infused, instilled, implanted or
injected;
a new Medicare PartD coverage gap discount program, in which
manufacturers must agree to offer 50% point-of-sale discounts
off negotiated prices of applicable brand drugs to eligible
beneficiaries during their coverage gap period, as a
condition for a manufacturers outpatient drugs to be covered
under Medicare PartD;
extension of a manufacturers Medicaid rebate liability to
covered drugs dispensed to individuals who are enrolled in
Medicaid managed care organizations;
expansion of eligibility criteria for Medicaid programs by,
among other things, allowing states to offer Medicaid
coverage to additional individuals and by adding new
mandatory eligibility categories for certain individuals with
income at or below 133% of the federal poverty level, thereby
potentially increasing a manufacturers Medicaid rebate
liability;
expansion of the entities eligible for discounts under the
Public Health Service pharmaceutical pricing program; and
a new Patient-Centered Outcomes Research Institute to
oversee, identify priorities in, and conduct comparative
clinical effectiveness research, along with funding for such
research.

In addition, other legislative
changes have been proposed and adopted since the Affordable Care
Act was enacted. These changes include, among others, the Budget
Control Act of 2011, which mandates aggregate reductions to
Medicare payments to providers of up to 2% per fiscal year
effective April1, 2013, and, due to subsequent legislative
amendments, will remain in effect through 2024 unless additional
Congressional action is taken. In January2013, President Obama
signed into law the American Taxpayer Relief Act of 2012, which,
among other things, further reduced Medicare payments to several
providers, including hospitals and cancer treatment centers,
increased the statute of limitations period for the government to
recover overpayments to providers from three to five years. These
new laws may result in additional reductions in Medicare and
other healthcare funding, which could have a material adverse
effect on customers for Savaras product candidates, if approved,
and, accordingly, its financial
operations.

Savara expects that healthcare
reform measures that may be adopted in the future, including the
possible repeal and replacement of the Affordable Care Act which
the Trump administration has stated is a priority, are
unpredictable, and the potential impact on Savaras operations and
financial position are uncertain, but may result in more rigorous
coverage criteria and lower reimbursement, and place additional
downward pressure on the price that it receives for any approved
product. Any reduction in reimbursement from Medicare or other
government-funded programs may result in a similar reduction in
payments from private payers. The implementation of cost
containment measures or other healthcare reforms may prevent
Savara from being able to generate revenue, attain profitability
or commercialize their drugs.

14

Foreign
Regulation

In addition to regulations in
the United States, Savara will be subject to a variety of foreign
regulations governing clinical trials and commercial sales and
distribution of its products to the extent Savara chooses to
develop or sell any products outside of the United States. The
approval process varies from country to country and the time may
be longer or shorter than that required to obtain FDA approval.
The requirements governing the conduct of clinical trials,
product licensing, pricing and reimbursement vary greatly from
country to country.

There is
significant uncertainty regarding the regulatory approval process
for any investigational new drug, substantial further testing and
validation of Savaras product candidates and related
manufacturing processes may be required, and regulatory approval
may be conditioned, delayed or denied, any of which could delay
or prevent Savara from successfully marketing its product
candidates and substantially harm its
business.

Pharmaceutical products
generally are subject to rigorous nonclinical testing and
clinical studies and other approval procedures mandated by the
FDA and foreign regulatory authorities. Various federal and
foreign statutes and regulations also govern or materially
influence the manufacturing, safety, labeling, storage, record
keeping and marketing of pharmaceutical products. The process of
obtaining these approvals and the subsequent compliance with
appropriate U.S. and foreign statutes and regulations is
time-consuming and requires the expenditure of substantial
resources.

Savara is preparing AeroVanc
for a Phase 3 trial, the success of which will be needed for FDA
approval to market AeroVanc in the United States to treat
persistent MRSA lung infection in cystic fibrosis patients. While
significant communication with the FDA on the Phase 3 study
design has occurred, even if the Phase 3 clinical study meets all
of its statistical goals and protocol end points, the FDA may not
view the results as robust and convincing. They may require
additional clinical studies and/or other costly studies, which
could require Savara to expend substantial additional resources
and could significantly extend the timeline for clinical
development prior to market approval. Additionally, Savara is
required by the FDA to conduct a two-year nonclinical
carcinogenicity study on the AeroVanc powder. The results of this
study will not be known until a short time prior to potential
submission of an NDA for AeroVanc. If the carcinogenicity study
cannot be completed for technical or other reasons, or provides
results that the FDA determine to be concerning, this may cause a
delay or failure in obtaining approval for
AeroVanc.

Molgradex is currently
undergoing a Phase 3 clinical study in Europe and Japan.
Concurrently, Savara plans to explore formulation changes to
Molgradex that could simplify the composition of the drug product
by eliminating one or more potentially unnecessary or harmful
excipients. While this change is expected by Savara to improve
the product quality and possibly reduce other documentation
requirements, the regulatory agencies may require additional
clinical or nonclinical studies prior to approval of such
formulation changes. Savara currently plans not to make any
formulation changes prior to submitting applications to
regulatory authorities for regulatory approvals of Molgradex, but
instead, to qualify the excipients in its nonclinical and
clinical studies. However, regulatory agencies may request Savara
to attempt to make the aforementioned formulation changes prior
to approval of the product, and therefore, even if current
clinical studies are deemed successful, such formulation changes
could require Savara to expend substantial additional resources,
including conducting an additional Phase 3 clinical study that
would significantly extend the timeline for clinical development
of Molgradex in PAP.

Savara has recently received
guidance from the FDA on the requirements to initiate clinical
studies in the United States and on the clinical study
requirements to achieve BLA approval for Molgradex. Based on the
guidance, Savara plans to amend its ongoing Phase 3 clinical
study to include more patients, and to amend its endpoint
hierarchy and statistical analyses to be used for U.S. approval
purposes prior to submission of a U.S. IND. However, no agreement
has yet been reached on the specific details of the statistical
analysis plan, which Savara plans to submit for FDA review prior
to the data analysis. Furthermore, even if the clinical study
meets all of its statistical goals and protocol end points, the
FDA may not view the results to provide persuasive evidence of
efficacy across multiple clinical endpoints. Instead, they may
require additional clinical studies and/or other costly studies,
including an additional Phase 3 study, which would require Savara
to expend substantial additional resources and would
significantly extend the timeline for clinical development prior
to market approval, or in failure to complete the clinical
development of Molgradex.

15

Significant uncertainty exists
with respect to the regulatory approval process for any
investigational new drug, including AeroVanc and Molgradex.
Regardless of any guidance the FDA or foreign regulatory agencies
may provide a drugs sponsor during its development, the FDA or
foreign regulatory agencies retain complete discretion in
deciding whether to accept an NDA or BLA or the equivalent
foreign regulatory approval submission for filing or, if
accepted, approve an NDA or BLA. There are many components to an
NDA or BLA or marketing authorization application submission in
addition to clinical study data. For example, the FDA or foreign
regulatory agencies will review the sponsors internal systems and
processes, as well as those of its CROs, CMOs and other vendors,
related to development of its product candidates, including those
pertaining to its clinical studies and manufacturing processes.
Before accepting an NDA for review or before approving the NDA or
BLA, the FDA or foreign regulatory agencies may request that
Savara provide additional information that may require
significant resources and time to generate and there is no
guarantee that its product candidates will be approved for any
indication for which Savara may apply. The FDA or foreign
regulatory agencies may choose not to approve an NDA or BLA for
any of a variety of reasons, including a decision related to the
safety or efficacy data, manufacturing controls or systems, or
for any other issues that the agency may identify related to the
development of its product candidates. Even if one or more Phase
3 clinical studies are successful in providing statistically
significant evidence of the efficacy and safety of the
investigational drug, the FDA or foreign regulatory agencies may
not consider efficacy and safety data from the submitted studies
adequate scientific support for a conclusion of effectiveness
and/or safety and may require one or more additional Phase 3 or
other studies prior to granting marketing approval. If this were
to occur, the overall development cost for the product candidate
would be substantially greater and its competitors may bring
products to market before Savara, which could impair its ability
to generate revenues from the product candidates, or even seek
approval, if blocked by a competitors Orphan Drug exclusivity,
which would have a material adverse effect on Savaras business,
financial condition and results of
operations.

Further, development of
Savaras product candidates and/or regulatory approval may be
delayed for reasons beyond its control. For example, U.S. federal
government shut-down or budget sequestration, such as one that
occurred during 2013, may result in significant reductions to the
FDAs budget, employees and operations, which may lead to slower
response times and longer review periods, potentially affecting
Savaras ability to progress development of its product candidates
or obtain regulatory approval for its product
candidates.

Even if the FDA or foreign
regulatory agencies grant approvals for Savaras product
candidates, the conditions or scope of the approval(s) may limit
successful commercialization of the product candidates and impair
Savaras ability to generate substantial sales revenue. For
example, the FDA may approve label claims for AeroVanc with age
restrictions and/or treatment duration limitations, or Molgradex
with restrictions for use only by patients unresponsive to the
current standard of care. They may limit the label of AeroVanc or
Molgradex to a subset of patients based on a review of which
patient groups had the greatest efficacious response in clinical
studies. Such label restriction may be undesirable and may limit
successful commercialization. The FDA or foreign regulatory
agencies may also only grant marketing approval contingent on the
performance of costly post-approval nonclinical or clinical
studies, or subject to warnings or contraindications that limit
commercialization. Additionally, even after granting approval,
the manufacturing processes, labeling, packaging, distribution,
adverse event reporting, storage, advertising, promotion and
recordkeeping for its products will be subject to extensive and
ongoing regulatory requirements. These requirements include
submissions of safety and other post-marketing information and
reports, registration, and continued compliance with current good
manufacturing processes, or cGMP, good clinical practices,
international conference on harmonization regulations and good
laboratory practices, which are regulations and guidelines that
are enforced by the FDA or foreign regulatory agencies for all of
its clinical development and for any clinical studies that Savara
conducts

16

post-approval. The FDA or
foreign regulatory agencies may decide to withdraw approval, add
warnings or narrow the approved indications in the product label,
or establish risk management programs that could restrict
distribution of its products. These actions could result from,
among other things, safety concerns, including unexpected side
effects or drug-drug interaction problems, or concerns over
misuse of a product. If any of these actions were to occur
following approval, Savara may have to discontinue
commercialization of the product, limit its sales and marketing
efforts, implement risk minimization procedures, and/or conduct
post-approval studies, which in turn could result in significant
expense and delay or limit its ability to generate sales
revenues.

Regulations may be changed
prior to submission of a marketing application that require
higher hurdles than currently anticipated. These may occur as a
result of drug scandals, recalls, or a political environment
unrelated to Savaras products.

SPECIAL NOTE REGARDING
FORWARD-LOOKING STATEMENTS

This Current Report on Form
8-K contains forward-looking statements within the meaning of
Section 27A of the Securities Act of 1933 and Section 21E of the
Securities Exchange Act of 1934. All statements, other than
statements of historical fact, included herein regarding the
development of our products, financial position, strategy,
regulatory status, clinical and nonclinical studies,
collaborations, commercial prospects, internal growth,
competition, intellectual property, regulatory reforms, products
and objectives are forward-looking statements. Forward-looking
statements may include, but are not limited to, statements
about:

the anticipated timing, structure and results of the clinical
trials for our product candidates;
the anticipated timing and outcome of the regulatory review
process for our product candidates;
any statements of the plans, strategies and objectives for
our future operations;
any statements concerning proposed new products, services or
developments;
any statements regarding future economic conditions or
performance;
our ability to protect our intellectual property and operate
our business without infringing upon the intellectual
property rights of others; and
our estimates regarding the sufficiency of our cash resources
and our need for additional funding.

The words believe, anticipate,
estimate, plan, expect, intend, may, could, should, potential,
likely, projects, continue, will, and would and similar
expressions are intended to identify forward-looking statements,
although not all forward-looking statements contain these
identifying words. Forward-looking statements reflect our current
views with respect to future events, are based on assumptions and
are subject to risks and uncertainties. We cannot guarantee that
we actually will achieve the plans, intentions or expectations
expressed in our forward-looking statements and you should not
place undue reliance on these statements. There are a number of
important factors that could cause our actual results to differ
materially from those indicated or implied by our forward-looking
statements. These important factors include those discussed
herein and the risks and uncertainties described in Savaras
filings with the SEC including the Form 8-K filed on April 27,
2017, other filings on Form 8-K, the Annual Report on Form 10-K
for the fiscal year ended December 31, 2016, the Form 10-Q for
the quarter ended March 31, 2017 and the Registration Statement
on Form S-4, as amended, related to the Mast/Savara merger. These
factors and the other cautionary statements made herein and
therein should be read as being applicable to all related
forward-looking statements whenever they appear. Except as
required by law, we do not assume any obligation to update any
forward-looking statement. We disclaim any intention or
obligation to update or revise any forward-looking statement,
whether as a result of new information, future events or
otherwise.

Item9.01 Financial Statements and Exhibits.

(b) Pro Forma Financial
Information

17

UNAUDITED PRO FORMA
CONDENSED COMBINED FINANCIAL
STATEMENTS

The following unaudited pro
forma condensed combined financial statements give effect to the
merger between Savara Inc. (formerly known as Mast Therapeutics,
Inc.) and Aravas Inc. (formerly known as Savara Inc.) and Aravass
previously consummated acquisition of Serendex A/S (Serendex) as
discussed below. The merger was structured as a reverse merger
and Aravas was determined to be the accounting acquirer based
upon the terms of the merger and other factors including:
(i)Aravas security holders owned approximately 77% of the
combined company immediately following the closing of the merger,
(ii)Aravas directors hold the majority (5 out of 7) of board
seats in the combined company, and (iii)Aravas management holds
all key positions in the management of the combined company. The
transaction will be accounted for under the acquisition method of
accounting under accounting principles generally accepted in the
United States (US GAAP). Under the acquisition method of
accounting for the purpose of these unaudited pro forma condensed
combined financial statements, management of Savara and Aravas
have determined a preliminary estimated purchase price,
calculated as described in Note 2 to these unaudited pro forma
condensed combined financial statements. The net tangible and
intangible assets acquired and liabilities assumed in connection
with the transaction are recorded at their estimated acquisition
date fair values. Any excess of purchase price over fair value of
identified assets acquired and liabilities assumed will be
recognized as goodwill. A final determination of these estimated
fair values will be based on the actual net tangible and
intangible assets of Savara that exist as of the date of
completion of the transaction.

Previously Consummated
Serendex Acquisition

On July15, 2016, Aravas
completed its acquisition of Serendex for total purchase
consideration of $12.4million. The purchase consideration
consisted primarily of $2.9million in common stock and $9.5 of
contingent consideration. The acquisition of Serendex is
reflected in Aravass historical consolidated balance sheet at
March31, 2017.

Pro Forma
Information

The unaudited pro forma
condensed combined balance sheet as of March31, 2017 and the
unaudited pro forma condensed combined statements of operations
for the three months ended March31, 2017 and for the year ended
December31, 2016 are based on (i)the historical consolidated
results of operations of Aravas and its subsidiary (which include
the results of Serendex subsequent to Aravass July15, 2016
acquisition of Serendex); (ii) the historical consolidated
results of operations of Savara; (iii)and the historical results
of operations of Serendex for the period January1, 2016 to
July14, 2016.

The unaudited pro forma
condensed combined balance sheet as of March31, 2017 assumes that
the merger took place on March31, 2017 and combines the
historical balance sheets of Savara and Aravas as of March31,
2017. The unaudited pro forma condensed combined statements of
operations for the three months ended March31, 2017 and for the
year ended December31, 2016 assumes that both the merger and the
acquisition of Serendex took place as of January1, 2016, and
combines the historical results of Savara and Aravas and the
pre-acquisition historical results of Serendex. The historical
financial statements of Savara, Aravas and Serendex (for the
interim period through June30, 2016), have been adjusted to give
pro forma effect to events that are (i)directly attributable to
the mergers, (ii)factually supportable, and (iii)with respect to
the statements of operations, expected to have a continuing
impact on the combined results.

The unaudited pro forma
condensed combined financial statements are based on the
assumptions and adjustments that are described in the
accompanying notes. The unaudited pro forma condensed combined
financial statements and pro forma adjustments have been prepared
based on preliminary estimates of fair value of assets acquired
and liabilities assumed. Differences between these preliminary
estimates and the final acquisition accounting will occur and
these differences could have a material impact on the
accompanying unaudited pro forma condensed combined financial
statements and the combined companys future results of operations
and financial position.

The unaudited pro forma
condensed combined financial statements do not give effect to the
potential impact of current financial conditions, regulatory
matters, operating efficiencies or other savings or expenses that
may be associated with the acquisition. The unaudited pro forma
condensed combined financial statements have been prepared for
illustrative purposes only and are not necessarily indicative of
the financial position or results of operations in future periods
or the results that actually would have been realized had Savara,
Aravas and Serendex been a combined company during the specified
periods. The unaudited pro forma condensed combined financial
statements, including the notes thereto, should be read in
conjunction with the Savara, Aravas and Serendex historical
audited financial statements for the year ended December31, 2016
and the unaudited condensed financial statements of Savara and
Aravas for the three months ended March31, 2017, and Serendex for
the six months ended June 30, 2016.

18

Unaudited Pro Forma
Condensed Combined Balance
Sheet

March31,
2017

(in
thousands)

Savara Aravas Pro Forma Merger Adjustments ProForma Combined

Assets

Current assets:

Cash and cash equivalents

$ 7,771 $ 10,464 $ 3,987 K $ 22,222

Investment securities

Grant and awards receivable

Prepaid expenses and other assets

1,038 1,426

Total current assets

8,159 11,502 3,987 23,648

Property, plant, and equipment, net

In-process research and development

2,500 10,609 18,803 G 31,912

Goodwill

3,007 3,089 18,394 H 24,490

Deposits and other non-current assets

Total assets

$ 13,885 $ 25,962 $ 41,184 $ 81,031

Liabilities, redeemable convertible preferred stock
and stockholders deficit

Current liabilities:

Accounts payable

$ $ $ $ 1,198

Accrued expenses and other liabilities

2,241 3,672 D 8,119
1,850 E

Accrued compensation and payroll taxes

2,270 2,270

Debt facility

1,580 (1,580 ) K

Capital lease obligation, current portion

Total current liabilities

6,593 4,810 12,029

Noncurrent liabilities:

Accrued interest on convertible promissory notes

(238 ) F

Debt facility, net of current portion

1,933 5,567 K 7,500

Deferred income tax liability

2,334 7,526 I 10,855

Convertible promissory notes

3,597 (3,597 ) F

Put option liability

1,055 (1,055 ) F

Contingent consideration

9,808 9,808

Capital lease obligation, net of current portion

Other long-term liabilities

Total liabilities

9,535 22,726 8,829 41,090

Redeemable convertible preferred stock:

Convertible preferred stock

43,885 (43,885 ) C

Stockholders equity:

Common stock

B
C
F

Additional paid-in-capital

321,037 3,174 (287,271 ) A 86,684
(3 ) B
43,878 C
5,869 F

Accumulated other comprehensive income/(loss)

(448 ) (448 )

Accumulated earnings/(deficit)

(316,942 ) (43,380 ) 316,942 A (46,566 )
(356 ) D
(1,850 ) E
(980 ) F

Total stockholders equity/(deficit)

4,350 (40,649 ) 76,240 39,941

Total liabilities and stockholders equity

$ 13,885 $ 25,962 $ 41,184 $ 81,031

See accompanying notes to the
unaudited pro forma condensed combined financial
statements.

19

Unaudited Pro Forma
Condensed Combined Statement of
Operations

(in thousands, except
share and per share data)

For the Three Months Ended March31, 2017

Savara Aravas Pro Forma Merger Adjustment Pro Forma Combined

Grant revenue

$ $ $ $

Operating expenses:

Product development

1,443 2,948 4,391

General and administrative

1,585 2,329

Impairment of IPRD

Depreciation and amortization

Transaction related costs

2,752 (3,744 ) J

Total operating expenses

5,791 4,774 (3,744 ) 6,821

Loss from operations

(5,697 ) (4,774 ) 3,744 (6,727 )

Interest and other income (expense), net

(172 ) (437 ) K (588 )

Loss before income taxes

(5,869 ) (5,211 ) 3,765 (7,315 )

Income taxes

Net loss

$ (5,869 ) $ (4,974 ) $ 3,765 $ (7,078 )

Accretion of redeemable convertible preferred stock

(24 ) (24 )

Net loss attributable to common stockholders

$ (5,869 ) $ (4,998 ) $ 3,765 $ (7,102 )

Basic and diluted net loss per share

$ (1.61 ) $ (0.97 ) $ $ (0.46 )

Weighted average common share outstanding- basic and
diluted

3,639,242 5,169,323 6,571,593 B 15,380,158

See accompanying notes to the
unaudited pro forma condensed combined financial
statements.

20

Unaudited Pro Forma
Condensed Combined Statement of
Operations

(in thousands, except
share and per share data)

For the Year Ended December31, 2016

Savara Aravas Serendex Pro Forma Merger Adjustment Pro Forma Combined
(seenote4)

Grant revenue

$ $ $ $ $

Operating expenses:

Product development

20,793 8,182 4,102 33,077

General and administrative

9,342 2,503 2,665 14,510

Impairment of IPRD

6,049 6,049

Depreciation and amortization

Transaction related costs

(618 ) J

Total operating expenses

36,584 11,348 6,767 (618 ) 54,081

Loss from operations

(36,456 ) (10,948 ) (6,767 ) (53,553 )

Interest and other income (expense), net

(2,053 ) (332 ) (58 ) 1,424 K (1,019 )

Loss before income taxes

(38,509 ) (11,280 ) (6,825 ) 2,042 (54,572 )

Income taxes

2,409 2,766

Net loss

$ (36,100 ) $ (10,923 ) $ (6,825 ) $ 2,042 $ (51,806 )

Accretion of redeemable convertible preferred stock

(94 ) (94 )

Net loss attributable to common stockholders

$ (36,100 ) $ (11,017 ) $ (6,825 ) $ 2,042 $ (51,900 )

Basic and diluted net loss per share

$ (12.12 ) $ (3.29 ) $ $ $ (4.12 )

Weighted average common share outstanding- basic and
diluted

2,978,348 3,348,647 6,260,093 B 12,587,088

See accompanying notes to the
unaudited pro forma condensed combined financial
statements.

21

NOTES TO THE UNAUDITED
PRO FORMA

CONDENSED COMBINED
FINANCIAL INFORMATION

1. Description of
Transaction and Basis of
Presentation

Description of
Transaction

On January6, 2017, Aravas
entered into the Merger Agreement with Savara, to which, among
other things, subject to the satisfaction or waiver of the
conditions set forth in the Merger Agreement, that a wholly-owned
subsidiary of Savara will merge with and into Aravas, with Aravas
becoming a wholly-owned subsidiary of Savara and the surviving
corporation of the merger. At the closing of the merger, each
outstanding share of Aravass common stock was converted into the
right to receive approximately .5860 of a share of common stock
of Savara, as well as the payment of cash in lieu of fractional
shares. Immediately following the effective time of the merger,
Savara equity holders owned approximately 23% of the outstanding
capital stock of the combined company, with Aravass preexisting
equity holders owning approximately
77%.

Basis of
Presentation

The unaudited pro forma
condensed combined financial statements were prepared in
accordance with the regulations of the Securities and Exchange
Commission (SEC). The unaudited pro forma condensed combined
balance sheet as of March31, 2017 is presented as if the merger
had been completed on March31, 2017. The unaudited pro forma
condensed combined statement of operations for the three months
ended March31, 2017 and for the year ended December31, 2016
assumes that both the merger and Aravass acquisition of Serendex
took place as of January1, 2016, and combines the historical
results of Mast and Savara and the pre-acquisition historical
results of Serendex.

Based on the terms of the
merger, Aravas is deemed to be the acquiring company for
accounting purposes and the merger will be accounted for under
the acquisition method of accounting in accordance with the
provisions of Accounting Standards Codification 805, Business
Combinations. Accordingly, assets and liabilities of Aravas will
be recorded as of the merger closing date at their respective
carrying value and assets and liabilities of Savara will be
recorded as of the merger closing date at their respective fair
values. Under the acquisition method of accounting for the
purpose of these unaudited pro forma financial statements,
management of Aravas and Savara have determined a preliminary
estimated purchase price, calculated as described in Note 2 to
these unaudited pro forma condensed combined financial
statements. The net tangible assets acquired and liabilities
assumed in connection with the transaction are at their estimated
acquisition date fair values. A final determination of these
estimated fair values will be based on the actual net tangible
assets of Savara that exist as of the date of completion of the
transaction.

To the extent there are
significant changes to the business following completion of the
merger, the assumptions and estimates set forth in the unaudited
pro forma condensed combined financial statements could change
significantly. Accordingly, the pro forma purchase price
adjustments are subject to further adjustments as additional
information becomes available and as additional analyses are
conducted following the completion of the merger. There can be no
assurances that these additional analyses will not result in
material changes to the estimates of fair
value.

2. Preliminary
Purchase Price

The preliminary estimated
purchase price of the merger is $34.0million using Savaras share
price for its common stock and its common shares outstanding as
of the close of business on April27, 2017. Note that in a reverse
merger, the purchase consideration determined under USGAAP will
be based on the market capitalization of Savara on the date of
the merger. The estimated fair value of the net assets acquired,
excluding goodwill is $12.6million.

22

Management of Aravas has
preliminarily concluded the proposed merger is a business
combination and will apply the acquisition method of accounting.
Under the acquisition method of accounting, the total purchase
price is allocated to the acquired tangible and intangible assets
and assumed liabilities of Savara based on their estimated fair
values as of the proposed merger closing date. The excess of the
purchase price over the fair value of assets acquired and
liabilities assumed, if any, is allocated to goodwill. To the
extent the actual purchase price varies from the estimated
purchase price used in these unaudited pro forma condensed
combined financial information, the impact will be an increase or
decrease in goodwill.

The preliminary allocation of
the estimated total purchase price of the proposed merger is as
follows (in thousands):

Fair value of Savara net assets to carry over to merged
company

$ 12,634

Goodwill

21,401

Total purchase consideration

$ 34,035

The preliminary estimated fair
values of the acquired assets and assumed liabilities of Savara
as of March31, 2017 is as follows (in
thousands):

Net tangible assets (liabilities)

$ (148 )

In-process research and development intangible asset, net
of deferred tax liability

12,782

Estimated fair value of net assets acquired

$ 12,634

The allocation of the
estimated purchase price is preliminary. The purchase price
allocation will remain preliminary until Aravass management
determines the fair values of assets acquired and liabilities
assumed. The final determination of the purchase price allocation
is anticipated to be completed as soon as practicable and will be
based on the fair values of the assets acquired and liabilities
assumed as of the merger closing date. The final amounts
allocated to assets acquired and liabilities assumed could differ
significantly from the amounts presented in the unaudited pro
forma condensed combined financial
statements.

3. Pro Forma
Adjustments

Pro forma adjustments are
necessary to reflect the acquisition consideration exchanged and
to adjust amounts related to the tangible assets and liabilities
of Savara to reflect the preliminary estimate of their fair
values, and to reflect the impact on the statements of operations
of the merger as if the companies had been combined during the
periods presented therein. The pro forma adjustments included in
the unaudited pro forma condensed combined financial statements
are as follows:

A. To reflect the elimination of Savaras historical stockholders
equity balances, including accumulated deficit, and to
reflect the adjustments to the fair value of Savaras net
assets recorded in the preliminary allocation of the
estimated total purchase price, at the close of the merger
referred to in Note 2 above.

Elimination of Savaras accumulated deficit

$ (316,942 )

Fair value adjustment to intangible assets (see G below)

18,803

Fair value adjustment to goodwill (see H below)

18,394

Adjustment to deferred tax liability (see I below)

(7,526 )

Total

$ (287,271 )

23

B. To reflect the reclassification Aravass par value of common
stock and additional paid-in capital in connection with the
exchange of Aravass common stock for Savaras common stock.
C. To reflect the conversion of Aravass redeemable convertible
preferred stock to Aravas common stock and then into shares
of Savara common stock.
D. To record $.4million of estimated transaction costs that were
not incurred as of March31, 2017.
E. To record $1.9million of severance liabilities in relation to
termination of employees of Savara upon consummation of the
merger.
F. To reflect the conversion of $4.3million in aggregate
principal of, and accrued interest on, Aravass convertible
notes into approximately 1.1million shares of Aravas common
stock and then into shares of Savara common stock and to
reflect the elimination of the put option (redemption
feature) on Aravass convertible notes.
G. To record intangible assets acquired in the merger and
eliminate Savaras historical intangible assets.

To record intangible assets acquired in the merger

$ 21,303

To eliminate historical Savara intangible assets

(2,500 )

Total

$ 18,803
H. To record goodwill as a result of the merger and eliminate
Savaras historical goodwill.

To record goodwill acquired in the merger

$ 21,401

To eliminate historical Savara goodwill

(3,007 )

Total

$ 18,394
I. To eliminate Savaras deferred tax liability related to prior
acquisitions that arose from amortizing, for tax purposes,
intangible assets from business combination transactions
prior to this merger and record deferred tax liability
related to the merger (assumes a 40% tax rate applied to
intangible assets acquired).

To record net deferred tax liability related to the merger

$ 8,521

To eliminate deferred tax liabilities related to Savaras
intangible assets from prior acquisitions

(995 )

Total

$ 7,526
J. To eliminate nonrecurring transaction costs of $3.7million
and $0.6million incurred during the three months ended
March31, 2017 and the year ended December31, 2016,
respectively, that are directly related to the merger.
K. The net increase to debt reflects the new debt of $7.5million
incurred to finance the acquisition of Savara, less the
effects of extinguishing Savaras outstanding debt of
$3.5million upon consummation of the acquisition.

To record the decrease for extinguishment of existing
Savara debt

$ (3,513 )

To record the increase for issuance of new debt

7,500

Total

$ 3,987

Represents the net increase to
interest expense resulting from interest on the new debt to
finance the merger and extinguish its existing debt as
follows:

Threemonths endedMarch31, 2017 Year ended December31, 2016

To eliminate interest expense and amortization of debt
issuance costs on outstanding Savara debt

$ (178 ) $ (2,053 )

To record the interest expense on new WSJ prime 4.25% debt

Total

$ (21 ) $ (1,424 )

24

4. Serendexs
Historical Financial
Statements

Schedule
1

Serendex

Statements of
Operations

For the Period from
January1, 2016 to July14,
2016

January1,2016 toJune30,2016 (1) (2) July1,2016 toJuly14,2016 (1) USGAAP Adjustments As Converted to USGAAP
DKK DKK DKK DKK USD

Grant revenue

(704 ) (a) $

Operating expenses

Product development

6,034 3,336 18,014 (a)(b) 27,384 4,102

General and administrative

12,954 4,834 17,788 2,665

Total operating expenses

18,988 8,170 18,014 45,172 6,767

Loss from operations

(18,284 ) (8,170 ) (18,718 ) (45,172 ) (6,767 )

Interest and other income (expense), net

(328 ) (59 ) (387 ) (58 )

Net loss

(18,612 ) (8,229 ) (18,718 ) (45,559 ) $ (6,825 )
(1) Amounts derived from Serendexs accounting records in
accordance with the IFRS as issued by the IASB and have been
reclassified to be consistent with the manner in which items
are classified in Aravas consolidated statement of operations
and comprehensive loss.
(2) Amounts derived from Serendexs historical unaudited condensed
financial statements for the six months ended June30, 2016.

USGAAP Adjustments to
Serendexs Historical Financial
Statements

On July15, 2016, Aravas
completed the acquisition of Serendex through its wholly-owned
subsidiary, Savara ApS. Serendex prepared its financial
statements in accordance with the IFRS as issued by the IASB.
Included in Schedule 1 above are the USGAAP adjustments to
Serendexs historical financial statements for the period from
January1, 2016 to July14, 2016.

(a) Revenue
recognition

i. In accordance with the IFRS as issued by the IASB, revenue
generated from sales of active pharmaceutical ingredient
(API) to vendors associated with clinical trial studies is
recognized as net revenue on the financial statements.
ii. Under USGAAP, revenue generated from sales of API to vendors
associated with clinical trial studies would be considered
contra-RD expenses as those revenues were not generated due
to commercialized sales to customers.

(b) Research and
development costs-
capitalization

i. In accordance with the IFRS as issued by the IASB, research
and development costs directly and indirectly attributable to
development of new products are capitalized as in-process RD.
ii. Under USGAAP, research and development costs are expensed as
incurred.

25

Translation of
Serendexs Historical Financial Statements to US
Dollars

The unaudited pro forma
condensed combined financial information is presented in US
dollars unless otherwise stated, and accordingly, the financial
information of Serendex used to prepare the unaudited pro forma
condensed combined financial information was translated from
Danish Krone to US dollars (Schedule 1) using the following
exchange rates, which correspond with the exchange rates for the
periods being presented:

Statement of operations for
the period from January1, 2016 to July14, 2016 (pre-acquisition
period): Average for Period 1 =
US$.1498

26


About SAVARA INC. (NASDAQ:SVRA)

Savara Inc., formerly Mast Therapeutics, Inc., is a clinical-stage pharmaceutical company. The Company is focused on the development and commercialization of novel therapies for the treatment of patients with rare respiratory diseases. Its pipeline includes AeroVanc, Molgradex and AIR001. AeroVanc is an inhaled formulation of vancomycin, which the Company is developing for the treatment of persistent methicillin-resistant Staphylococcus aureus, lung infection in cystic fibrosis patients. Molgradex is an inhaled formulation of recombinant human granulocyte-macrophage colony-stimulating factor. It is developing Molgradex for the treatment of autoimmune pulmonary alveolar proteinosis, a rare lung disease. AIR001 is a sodium nitrite solution for inhalation via nebulization. AIR001 is in Phase II clinical development for the treatment of heart failure with preserved ejection fraction, also known as diastolic heart failure or heart failure with preserved systolic function.

SAVARA INC. (NASDAQ:SVRA) Recent Trading Information

SAVARA INC. (NASDAQ:SVRA) closed its last trading session up +0.45 at 5.16 with 74,350 shares trading hands.