You should read the following discussion and analysis of our financial condition
and results of operations together with the condensed consolidated financial
statements and notes thereto included elsewhere in this report, and our audited
financial statements and related notes thereto included as part of our Annual
Report on Form 10­K for the year ended December 31, 2021. This discussion and
analysis, and other parts of this report, contain forward-looking statements,
including, but not limited to, statements related to the potential of
Gritstone's programs. Such forward-looking statements involve substantial risks
and uncertainties that could cause the outcome of Gritstone's programs, future
results, performance or achievements to differ significantly from those
expressed or implied by the forward-looking statements, including interim
results obtained may differ from those at completion of the studies and clinical
trials. Such risks and uncertainties include, among others, the uncertainties
inherent in the drug development process, including Gritstone's programs'
clinical development, the process of designing and conducting preclinical and
clinical trials, the regulatory approval processes, the timing of regulatory
filings, the challenges associated with manufacturing drug products, Gritstone's
ability to successfully establish, protect and defend its intellectual property
and other matters that could affect the sufficiency of existing cash to fund
Gritstone's operations. Our actual results could differ materially from those
discussed in these forward-looking statements. For a further description of the
risks and uncertainties that could cause actual results to differ from those
expressed in these forward-looking statements, as well as risks relating to our
business in general, see the section titled "Risk Factors". These
forward-looking statements speak only as of the date hereof. Except as required
by law, we assume no obligation to update or revise these forward-looking
statements for any reason.

Overview



We discover, develop, manufacture and deliver next generation cancer and
infectious disease vaccine candidates with the aim of improving patient outcomes
and eliminating disease. The immune system sits at the nexus of many diseases,
and manipulation of the immune system has enormous potential to drive
transformational therapeutic and preventative benefits. Our approach seeks to
generate a potent and durable therapeutic or protective immune response by
leveraging insights into the immune system's ability to recognize and destroy
diseased cells and eliminate virally-infected cells. Specifically, we focus on
the induction of T cells, critical but underexplored components in treatment and
prevention of disease, to generate differentiated immune responses that extend
and improve the quality of life.

Our programs are built on two key platforms. The first platform is our
proprietary EDGE™ epitope identification platform, which enables us to identify
antigens that can be recognized by the immune system on tumors or
virally-infected cells with a high degree of accuracy. The second platform is
our potent, flexible, vaccine platform, which we have engineered to deliver
immunogens to the immune system to drive the destruction of tumors or
virally-infected cells. Our vaccine platform leverages our two proprietary
vaccine vectors, self-amplifying mRNA (samRNA) and chimpanzee adenovirus (ChAd).
We utilize these "mix and match" vectors in a variety of ways, including as a
heterologous prime-boost (one vector followed by the other) or homologous
prime-boost (use the same vector twice). Together, these proprietary and
synergistic technologies enable us to build robust and distinct pipelines in
oncology and infectious disease. Additionally, our in-house manufacturing
capabilities enable us to drive down cost and production time, as well as
maintain control over intellectual property and product quality of our products.

Self-amplifying mRNA (samRNA)



Our samRNA vector is based on a synthetic RNA molecule derived from a wild-type
Venezuelan Equine Encephalitis Virus (VEEV) replicon with the goal of extending
the duration and magnitude of immunogen expression to drive potent and durable
immune responses. The samRNA is delivered in a lipid nanoparticle (LNP)
formulation. Like traditional mRNA vaccines, samRNA vaccines use the host cell's
transcription system to produce target antigens to stimulate adaptive immunity.
Unlike traditional mRNA, samRNA has an inherent ability to replicate by creating
copies of the original strand of RNA once it is in the cell. Potential benefits
of samRNA may include extended duration and magnitude of antigen expression,
strong and durable induction of neutralizing antibody and T cell immunity (CD4+
and CD8+), dose sparing, and a refrigerator stable product.

The samRNA platform is a key asset for Gritstone. Within our oncology programs,
we have presented clinical data supporting the identification and selection of
an optimal dosing regimen for our novel samRNA vector against

                                       32
--------------------------------------------------------------------------------


solid tumors. We also leverage our samRNA platform for infectious diseases, and
our preclinical and clinical studies to date demonstrate the potential overall
potency and dose sparing opportunity of samRNA in viral diseases.

The success of first-generation mRNA vaccines for SARS-CoV-2 (Comirnaty® and
Spikevax) has validated mRNA as a vaccine technology and we believe the samRNA
vector has the potential to offer key benefits over mRNA, including dose sparing
and more potent CD8+ T cell induction, within both oncology and viral diseases.

Chimpanzee Adenovirus (ChAd)

Chimpanzee Adenoviral (ChAd) vectors have been utilized in clinical studies in infectious disease and oncology over the last 20 years and have been demonstrated to be well tolerated and effective at generating rapid and substantial CD4+ and CD8+ T cell responses. Additionally, ChAd vectors can induce B cell immune responses, i.e., elicit nAbs.

In-house Manufacturing



We manufacture our product candidates at our own fully integrated current good
manufacturing practice (cGMP) biomanufacturing facilities. Our ability to
control the manufacturing of high-quality tumor-specific immunotherapy and
infectious disease vaccine candidates, and scale production, if early data are
positive, is critical for efficient clinical development of our vaccine
candidates and commercialization.

Our manufacturing know-how also contributes to our translational science and
optimization of our production candidates. Through our work, we gain insights
from "bench to manufacturing to bedside" and back. We translate such insights
across functions and systems to optimize antigen cassette design, dose and
vaccine regimen to induce differentiated immune response.

Clinical Programs



The table below summarizes key information about our ongoing clinical trials.

Program   Phase   Status          Indication(s)             Collaborator      Commercial Rights
GRANITE    1/2    Enrollment      Early stage & advanced    -                 Gritstone
                  Complete;       solid tumors
                  Treatment
                  Ongoing
GRANITE    2/3    Enrolling;      MSS-CRC* first line       -                 Gritstone
                  Treatment       maintenance
                  Ongoing
GRANITE     2     Terminated      MSS-colon cancer          -                 Gritstone
                                  adjuvant
SLATE      1/2    Complete        p53, KRAS Advanced        -                 Gritstone
                                  Solid Tumors
SLATE       2     Enrolling       KRASmut                   -                 Gritstone
CORAL       1     Enrollment      COVID-19 naïve &          NIAID, IDCRC      Gritstone
                  Complete        booster
CORAL       1     Enrollment      COVID-19 booster          -                 Gritstone
                  Complete
CORAL       1     Enrolling       COVID-19 in South         CEPI              Gritstone
                                  Africa (naïve,
                                  convalescent, HIV+)
HIV         1     Ongoing         HIV treatment/cure        Gilead Sciences   Gilead**


* MSS-CRC = microsatellite stable colorectal cancer ** Gilead is responsible for conducting a Phase 1 study

COVID-19 Update



Since the COVID-19 pandemic began, providers of healthcare services have had to
deal with significant strains on their operations. These strains have affected
all healthcare institutions, including those where we conduct our clinical
trials, with some institutions prohibiting or postponing the initiation of new
clinical trials, slowing or halting enrollment in existing trials and
restricting the on-site monitoring of clinical trials. Although our operations
have not been materially impacted by the COVID-19 pandemic, we have experienced
slowing of patient recruitment and sample collection in our ongoing clinical
trials. Additionally, as a result of the COVID-19 pandemic, competition for
potential patients in our trials may be further exaggerated as a result of
multiple clinical site closures. To date, the COVID-19 pandemic has not
materially affected our supply chain or production schedule, but further
escalation of the health

                                       33
--------------------------------------------------------------------------------

crisis has the potential to cause delays in our supply chain and manufacturing operations, which could materially adversely impact our business.



In response to the COVID-19 pandemic, we have implemented heightened health and
safety measures designed to comply with applicable federal, state and local
guidelines, and transitioned to a flexible work environment, where employees who
can work from home effectively are allowed to do so. We have implemented virtual
meeting and messaging technology and encourage employees to follow local health
authority guidance. As the pandemic and its impacts continue to evolve, we may
need to undertake additional actions that could impact our operations if
required by applicable laws or regulations or if we determine such actions to be
in the best interests of our employees.

Oncology Program Updates



We are developing a portfolio of vaccine-based cancer immunotherapy product
candidates using a heterologous prime (ChAd)/boost (samRNA) approach aimed at
the highly targeted activation of tumor-specific neoantigens (TSNA) in solid
tumors. Our two clinical-stage programs (GRANITE, which is "individualized" and
SLATE, which is "off-the-shelf") aim to induce a substantial neoantigen-specific
CD8+ T cell response using neoantigen-containing immunotherapies. GRANITE
patients receive a product candidate made specifically for them, based upon
their tumor DNA/RNA sequence. In contrast, SLATE patients receive an
off-the-shelf product candidate made for a subset of patients based on common
driver mutations.

GRANITE Individualized Vaccine Program for Solid Tumors



Our first oncology program, GRANITE, consists of individualized neoantigen-based
immunotherapy candidates for solid tumors. GRANITE was granted Fast Track
designation by the FDA for the treatment of microsatellite stable colorectal
cancer (MSS-CRC).

In an ongoing Phase 1/2 study evaluating GRANITE in combination with checkpoint
inhibitors for patients with MSS-CRC that have been treated with FOLFOX/FOLFIRI
therapy as well as in patients with gastro-esophageal (GEA) cancer that have
been treated with platinum-based chemotherapy, GRANITE has shown to be generally
well-tolerated, with no dose limiting toxicities, and demonstrated consistent
and potent immunogenicity (CD8+ neoantigen-specific T cell induction in all
subjects), in addition to tumor lesion size reductions and molecular responses
as measured by reduction in circulating tumor DNA (cDNA). Initial results from
this study were presented during the European Society of Medical Oncology (ESMO)
Congress in September 2021, and follow-up for patients in the study continues.
As of the August 5, 2021 data cutoff, 4 of 9 treated patients with MSS-CRC had a
molecular response. Patients who demonstrated molecular response had median
overall survival of >17 months (median not reached) whereas those without
molecular response exhibited a median overall survival of 7.8 months, consistent
with expected outcomes in 3rd line treatment of MSS-CRC. As of the next data
cutoff on May 9, 2022, the observed median overall survival in this group
exceeded 18 months with median OS not yet reached. All patients with MSS-CRC
assessed for molecular response and alive at the time of our ESMO 2021 data
presentation remained alive after an additional 35 weeks of follow-up. We
believe these data demonstrate a correlation between a decrease in ctDNA and
extended overall survival. Interim results from the Phase 1/2 study were
published in Nature Medicine in August 2022 ("Individualized, heterologous
chimpanzee adenovirus and self-amplifying mRNA neoantigen vaccine for advanced
metastatic solid tumors: phase 1 trial interim results").

In the first quarter of 2022, we initiated a randomized Phase 2/3 study
(GRANITE-CRC-1L, NCT05141721), evaluating GRANITE as a maintenance treatment in
patients with newly diagnosed, metastatic MSS-CRC who have completed
FOLFOX-bevacizumab induction therapy. This Phase 2/3 study has registrational
intent and has been discussed with the FDA. In support of this study, we entered
into a clinical trial collaboration and supply agreement with F. Hoffman-La
Roche Ltd to evaluate the safety and tolerability of GRANITE in combination with
TECENTRIQ (atezolizumab). Enrollment in GRANITE-CRC-1L study is ongoing, and the
first patient was treated in July 2022. Initial data from GRANITE-CRC-1L are
expected in the fourth quarter of 2023. In August 2022, we terminated the
GRANITE-ADJUVANT study (NCT05456165), a randomized Phase 2 trial evaluating
GRANITE in patients with stage II/III colon cancer who are circulating tumor DNA
(ctDNA)+ after definitive surgery, due to reprioritization of resources. No
patients had been enrolled at the time of study termination.

SLATE "Off the shelf" Vaccine Program for Solid Tumors



Our second oncology program, SLATE, consists of "off-the-shelf", TSNA-directed
immunotherapy product candidates. SLATE contains a fixed cassette with TSNA that
are shared across a subset of cancer patients rather than a cassette unique to
an individual patient, which distinguishes it as a potential off-the-shelf
alternative candidate to

                                       34
--------------------------------------------------------------------------------

GRANITE. Our long-term vision for the SLATE program is to develop a suite of novel vaccine candidates that target common tumor antigens to broaden addressable patient population and drive multiple antigens per patient.



The first version of SLATE (SLATE v1) was studied in a Phase 1/2 study, in
collaboration with Bristol-Myers Squibb, in 26 patients with metastatic solid
tumors, most of whom had KRAS-mutant tumors largely focused on non-small cell
lung cancer (NSCLC), MSS-CRC, and pancreatic ductal adenocarcinoma. In the
initial part of this study, which was focused on KRAS and p53 mutations, SLATE
v1 demonstrated induction of CD8+ T cells against multiple KRAS driver
mutations, and greatest activity was observed in a subset of NSCLC patients with
the KRASmut G12C mutations. Although these initial outcomes were promising, we
believed our SLATE candidate could be further optimized to maximize potential
clinical benefit.

Subsequently, we developed a next-generation, optimized SLATE candidate,
SLATE-KRAS, (formerly referred to as SLATE v2) that exclusively includes
epitopes from mutated KRAS and exhibited immunogenic superiority over v1 in
human HLA-transgenic mice. SLATE-KRAS is now in Phase 2 testing under the same
IND and protocol as SLATE v1 in patients with advanced NSCLC and CRC. We
disclosed the initial results from the Phase 2 portion of the Phase 1/2 study,
including data from both SLATE v1 and SLATE-KRAS, at the 2022 AACR Annual
Meeting in April 2022 and at the 2022 ESMO Congress in September 2022. These
data demonstrate a favorable safety and tolerability profile, support the
potential of SLATE-KRAS to drive stronger CD8+ T cell responses to mutant KRAS
than SLATE v1 and provide early signals of efficacy as measured by reduction in
ctDNA (molecular response). Specifically, we saw a molecular response rate (MRR)
of 39% in evaluable patients with advanced NSCLC and CRC, a figure consistent
with the MRR seen in the Phase 1/2 studies of GRANITE (MRR of 44%). We believe
these initial data also demonstrate a correlation between molecular response and
overall survival (OS) among patients with NSCLC. Based on these data, we have
begun evaluating moving SLATE-KRAS into additional studies and earlier lines of
treatment.

Infectious Disease Program Updates



In early 2021, we initiated two programs in infectious diseases: CORAL, a
second-generation prophylactic program against COVID-19, and a collaboration
with Gilead Sciences to develop a therapeutic vaccine against HIV. Our
infectious disease programs aim to deliver vaccine candidates that induce both B
cell and T cell immunity with the potential to drive potent and durable immune
response that can be applied for either protective or therapeutic benefit. This
approach has demonstrated the ability to generate robust CD8+ T cells and
neutralizing antibodies against SARS-CoV-2 in multiple preclinical and clinical
studies and is being evaluated against multiple other pathogens in
Gritstone-owned and partnered studies. We believe that initially evaluating our
approach in SARS-CoV-2 can provide proof of concept for a number of infectious
diseases.

CORAL - Second Generation COVID-19 Vaccine Program



Our CORAL program is a second-generation SARS-CoV-2 vaccine platform delivering
spike and additional SARS-CoV-2 T cell epitopes. We believe this approach of
inducing both neutralizing antibodies and T cell responses could offer the
potential for more durable protection and broader immunity against SARS-CoV-2
variants than that provided by first-generation SARS-CoV-2 vaccines and serve as
a basis for developing a pan-coronavirus vaccine. Within our CORAL program, we
developed an optimized samRNA vaccine candidate that we believe is
differentiated from first-generation mRNA vaccines. The program is supported by
key relationships with the Gates Foundation, the National Institute of Allergy
and Infectious Disease (NIAID), the Coalition for Epidemic Preparedness
Innovations (CEPI), and through a license agreement with the La Jolla Institute
for Immunology (LJI).

We have conducted preclinical studies demonstrating that our SARS-CoV-2 vaccine
candidate induced significant and sustained levels of neutralizing antibodies
and T cells against the Spike protein, plus a broad T cell response against
epitopes from multiple viral genes outside of Spike. Results from one of these
studies, a non-human primate challenge study (NHP Challenge Study), were
published in Nature Communications in June 2022.

We are currently evaluating four distinct SARS-CoV-2 product candidates across
three different Phase 1 clinical trials containing Spike plus additional
non-Spike T cell epitope (TCE) sequences (and also full-length nucleocapsid).
These studies include homologous and heterologous prime-boost regimens. All of
these studies are ongoing and data from all are expected in the fourth quarter
of 2022.

In January 2022, we shared data from the first cohort of our Phase 1 CORAL-BOOST study which showed our samRNA vaccine candidate induced robust neutralizing antibody titers and elicited broad T cell responses when


                                       35
--------------------------------------------------------------------------------

administered at 10ug following two-dose administration of Vaxzervria. The neutralizing antibody titer levels against SARS-CoV-2 Spike protein shared at this time were consistent with published data from higher doses of first-generation mRNA vaccines in a similar clinical context.



In August 2022, we reported 6-month follow-up data from a subset of patients
within the first two cohorts of the CORAL-BOOST study who elected to receive
only a single 10µg or 30µg samRNA boost vaccination (n=7). The data demonstrated
the neutralizing antibody levels reported in January 2022 persisted after 6
months, and durable neutralizing antibodies against wild type Spike as well as
key Spike variants of concern (Beta, Delta and Omicron) were observed.
Additionally, T cell responses to Spike and non-Spike T cell epitopes (TCEs)
remained generally stable over the 6-month observation period.

HIV Vaccine Collaboration with Gilead Sciences



In January 2021, we entered into a collaboration, option and license agreement
with Gilead Sciences, Inc. (Gilead) to research and develop a vaccine-based
immunotherapy for HIV. Together, we plan to develop an HIV-specific therapeutic
vaccine using our proprietary prime-boost vaccine platform, comprised of samRNA
and adenoviral vectors, with antigens developed by Gilead. The collaboration and
the program are progressing well and a Phase I trial is ongoing. If Gilead
decides to progress development beyond the Phase 1 study by exercising their
exclusive option, the Company will receive a $40.0 million non-refundable option
exercise fee.

Preclinical Research Updates



Beyond GRANITE, SLATE, CORAL and the collaboration with Gilead, we continue to
apply our broad set of capabilities in oncology and infectious diseases through
promising preclinical work and partnerships. These projects include a
pan-coronavirus program, a flu program, and a program aiming to develop an
optimal immunogen in the context of human papillomavirus (HPV), which is
supported by the Gates Foundation.

Components of Our Operating Results

Collaboration and Grant Revenue



We have no products approved for sale and have never generated any revenue from
product sales. For the three and nine months ended September 30, 2022,
respectively, we recognized $3.0 million and $15.7 million of revenue from the
2seventy Agreement, the Gilead Collaboration Agreement, and the grant agreements
with CEPI and the Gates Foundation. For the three and nine months ended
September 30, 2021, we recognized $2.6 million and $45.1 million for the three
and nine months ended September 30, 2021, respectively, of revenue from the
2seventy Agreement, the Gilead Collaboration Agreement, another small
collaboration agreement, and the grant agreement with CEPI. See Note 9 to our
condensed consolidated financial statements for additional information.

In the future, we expect to continue to recognize revenue from the 2seventy
Agreement and the Gilead Collaboration Agreement and may generate revenue from
product sales or other collaboration agreements, strategic alliances and
licensing arrangements. We expect our revenue to fluctuate on a quarterly and
annual basis due to the timing and amount of license fees, reimbursement of
costs incurred, milestone and other payments, as well as product sales, to the
extent that any are successfully commercialized. If we fail to complete the
development of our product candidates in a timely manner or obtain regulatory
approval for them, our ability to generate future revenue, and our results of
operations and financial position, would be materially adversely affected.

Operating Expenses

Research and Development Expenses



Since our inception, we have committed significant resources to our research and
development activities, including conducting preclinical studies, manufacturing
development efforts and related development activities for our product
candidates.

                                       36
--------------------------------------------------------------------------------


Research and development activities account for a significant portion of our
operating expenses. Research and development costs are expensed as incurred.
These costs include:

External research and development expenses, including:



o

expenses incurred under arrangements with third parties, including clinical research organizations, or CROs, preclinical testing organizations, CMOs, academic and non-profit institutions and consultants;



o

fees related to our license agreements;

Internal research and development expenses, including (i) headcount-related expenses, such as salaries, payroll taxes, benefits, non-cash stock-based compensation and travel, for employees contributing to research and (ii) development activities, including the costs associated with the development of our EDGE™ platform; and

Other expenses, which include direct and allocated expenses for laboratories, facilities and other costs.



Pursuant to our Arbutus License Agreement, Arbutus granted us a worldwide,
exclusive license to certain technology of Arbutus, including Arbutus' portfolio
of proprietary and clinically-validated LNP products and associated intellectual
property, as well as technology transfer of Arbutus' manufacturing know-how.
During the nine months ended September 30, 2022 and 2021, we had no research and
development expense under the agreement.

Pursuant to our 2020 Genevant License Agreement, Genevant granted us exclusive
license rights under certain intellectual property related to Genevant's LNP
technology for a single indication, and we agreed to pay Genevant an initial
payment of $2.0 million, and up to an aggregate of $71.0 million in specified
development, regulatory, and commercial milestones, and low to mid-single digit
royalties on net sales of licensed products. The upfront payment of $2.0 million
was included in research and development expenses during 2020. In March 2022, a
milestone in the amount of $1.0 million was met, which was included in research
and development expense for the nine months ended September 30, 2022.

Pursuant to our 2021 Genevant License Agreement, we obtained a nonexclusive
license to Genevant's LNP technology to develop and commercialize
self-amplifying RNA, or samRNA, vaccines against SARS-CoV-2, the virus that
causes COVID-19. Under the 2021 Genevant License Agreement, we made a $1.5
million upfront payment to Genevant, and Genevant is eligible to receive from us
up to $141.0 million in contingent milestone payments per product, plus certain
royalties on future product sales or licensing (or, in certain scenarios and
subject to certain conditions, in lieu of these milestones and royalties
Genevant would receive a percentage of amounts we receive from sublicenses). In
March 2021, a milestone was met following the initial patient treatment in the
Phase 1 clinical trial conducted through the NIAID-supported IDCRC. Both the
$1.5 million upfront and $1.0 million milestone payments were recorded as
research and development expense for the nine months ended September 30, 2021.
No research and development expense was recorded for the nine months ended
September 30, 2022.

We expect our research and development expenses to increase substantially in the
future as we continue to advance our product candidates into and through
clinical studies and pursue regulatory approval. Such activities are costly and
time-consuming and we expect our clinical studies to generally become larger and
more costly to conduct as they advance into later stages. The successful
development of our product candidates is highly uncertain. The actual
probability of success for our product candidates may be affected by a variety
of risks and uncertainties associated with drug development, including those
described in the section entitled "Risk Factors" included in Part II, Section 1A
and elsewhere in this report.

                                       37
--------------------------------------------------------------------------------

The following table summarizes our research and development expenses by program and category (in thousands):



                                            Three Months Ended September 30,           Nine Months Ended September 30,
                                               2022                  2021                2022                  2021

GRANITE program external expenses $ 3,088 $ 3,387 $ 9,232 $ 8,767 SLATE program external expenses

                       604                 1,024               2,076                 2,935
CORAL program external expenses                     2,802                 1,193               9,002                 2,828
Other program external research and
development expenses                                5,463                 5,996              17,702                19,533
Personnel-related expenses (1)                     10,159                 8,624              31,117                25,171
Other unallocated research and
development expenses                                4,320                 4,172              12,854                12,090

Total research and development expenses $ 26,436 $ 24,396 $ 81,983 $ 71,324

(1)

Personnel-related expenses include stock-based compensation expense of $1.6 million and $5.1 million, respectively, for the three and nine months ended September 30, 2022, and $1.7 million and $5.0 million, respectively, for the three and nine months ended September 30, 2021.

Since our research and development employees and infrastructure resources are utilized across our development programs, we do not track internal related expenses on a program-by-program basis.

General and Administrative Expenses



Our general and administrative expenses consist primarily of salaries and
related costs, including, but not limited to, payroll taxes, benefits, non-cash
stock-based compensation and travel. Other general and administrative expenses
include legal costs of pursuing patent protection of our intellectual property
and professional service fees for auditing, tax and general legal services. We
expect our general and administrative expenses to continue to increase in the
future as we expand our operating activities and prepare for potential
commercialization of our current and future product candidates, increase our
headcount and support our operations as a public company, including increased
expenses related to legal, accounting, regulatory and tax-related services
associated with maintaining compliance with requirements of the Nasdaq Global
Select Market and the SEC, directors and officers liability insurance premiums
and investor relations activities. Allocated expenses consist of rent expenses
related to our office and research and development facilities, depreciation and
other allocated costs not otherwise included in research and development
expenses.

Interest Income

Interest income consists primarily of interest income and investment income earned on our cash, cash equivalents and marketable securities.

Interest Expense



Interest expense consists of interest expense related to our debt facility. A
portion of the interest expense is non-cash expense relating to the accretion of
the final payment fees and amortization of debt discount and debt issuance costs
associated with the Loan Agreement (as defined below).

                                       38
--------------------------------------------------------------------------------

Results of Operations

Comparison of the Three and Nine Months Ended September 30, 2022 and 2021



The following table sets forth the significant components of our results of
operations (in thousands):

                                        Three Months Ended September 30,
                                           2022                   2021            Change
Revenues:
Collaboration and license revenues   $            436       $          2,401     $ (1,965 )
Grant revenues                                  2,585                    213        2,372
Total revenues                                  3,021                  2,614          407
Operating expenses:
Research and development                       26,436                 24,396        2,040
General and administrative                      6,462                  6,373           89
Total operating expenses                       32,898                 30,769        2,129
Net loss from operations                      (29,877 )              (28,155 )     (1,722 )
Interest income                                   462                     37          425
Interest expense                                 (551 )                    -         (551 )
Net loss                             $        (29,966 )     $        (28,118 )   $ (1,848 )



                                                    Nine Months Ended September 30,
                                                      2022                   2021             Change
Revenues:
Collaboration and license revenues              $          7,942       $         44,937     $  (36,995 )
Grant revenues                                             7,741                    213          7,528
Total revenues                                            15,683                 45,150        (29,467 )
Operating expenses:
Research and development                                  81,983                 71,324         10,659
General and administrative                                22,209                 19,251          2,958
Total operating expenses                                 104,192                 90,575         13,617
Net loss from operations                                 (88,509 )              (45,425 )      (43,084 )
Interest income                                              663                    112            551
Interest expense                                            (551 )                    -           (551 )
Net loss                                        $        (88,397 )     $        (45,313 )   $  (43,084 )

Collaboration and License and Grant Revenues



Collaboration and license revenues from our collaboration arrangements and grant
revenues were $3.0 million and $15.7 million for the three and nine months ended
September 30, 2022, respectively. During the three months ended September 30,
2022, we recognized $0.2 million in collaboration revenue related to the
2seventy Agreement, $0.2 million in collaboration revenue related to the Gilead
Collaboration Agreement, $2.3 million in grant revenue from the CEPI Funding
Agreement, and $0.3 million in grant revenue from the Gates Foundation. During
the nine months ended September 30, 2022, we recognized $6.5 million in
collaboration revenue related to the 2seventy Agreement, $1.5 million in
collaboration revenue related to the Gilead Collaboration Agreement, $6.9
million in grant revenue from the CEPI Funding Agreement, and $0.8 million in
grant revenue from the Gates Foundation. The amount of collaboration revenue
recognized related to the 2seventy Agreement during the nine months ended
September 30, 2022 included cumulative catch-up adjustments increasing
contribution revenue by $5.5 million due to revisions to estimated costs to
complete the remaining performance obligation.

Collaboration and license revenues from our collaboration arrangements and grant
revenues were $2.6 million and $45.1 million for the three and nine months ended
September 30, 2021, respectively. During the three months ended September 30,
2021, we recorded $1.6 million in collaboration revenue related to the Gilead
Collaboration Agreement, $0.8 million in collaboration revenue related to the
2seventy Agreement, and $0.2 million in grant revenue related to the CEPI
agreement. During the nine months ended September 30, 2021, we recorded $38.6
million in license revenue and $4.1 million in collaboration revenue related to
the Gilead Collaboration Agreement, $2.1 million

                                       39
--------------------------------------------------------------------------------

in collaboration revenue related to the 2seventy Agreement, $0.1 million in collaboration revenue related to another small collaboration agreement, and $0.2 million in grant revenue related to the CEPI agreement.

See Note 9 to our condensed consolidated financial statements for additional information.

Research and Development Expenses



Research and development expenses were $26.4 million and $82.0 million for the
three and nine months ended September 30, 2022, respectively, and $24.4 million
and $71.3 million for the three and nine months ended September 30, 2021,
respectively.

The increase of $2.0 million for the three months ended September 30, 2022
compared to the three months ended September 30, 2021 was primarily due to
increases of $1.6 million in personnel-related expenses, $1.0 million in outside
services, consisting primarily of clinical trial and other chemistry,
manufacturing and controls ("CMC") related expenses, and $0.6 million in
facilities-related costs, offset by a decrease of $1.2 million in laboratory
supplies.

The increase of $10.7 million for the nine months ended September 30, 2022
compared to the nine months ended September 30, 2021 was primarily due to
increases of $6.4 million in personnel-related expenses, $7.1 million in outside
services, consisting primarily of clinical trial and other CMC related expenses,
and $1.7 million in facilities-related costs, offset by decreases of $2.7
million in laboratory supplies and $1.8 million in milestone and license
payments.

General and Administrative Expenses



General and administrative expenses were $6.5 million for the three months ended
September 30, 2022 compared to $6.4 million for the three months ended September
30, 2021. The increase of $0.1 million was primarily attributable to an increase
of $0.7 million in personnel-related expenses, offset by decreases of $0.4
million in outside services and $0.2 million in facilities-related costs.

General and administrative expenses were $22.2 million for the nine months ended
September 30, 2022 compared to $19.2 million for the nine months ended September
30, 2021. The increase of $3.0 million was primarily attributable to increases
of $3.7 million in personnel-related expenses, offset by decreases of $0.4
million in outside services and $0.3 million in facilities-related costs.

Interest Income



Interest income was $0.5 million and $0.7 million for the three and nine months
ended September 30, 2022, respectively. Interest income was immaterial and $0.1
million for the three and nine months ended September 30, 2021, respectively.
The income for both periods represent interest and investment income from cash,
cash equivalents and marketable securities.

Interest Expense



Interest expense was $0.6 million for the three and nine months ended September
30, 2022. There was no interest expense for the three and nine months ended
September 30, 2021. The interest expense is comprised of the contractual coupon
interest expense, the amortization of the debt discount and issuance costs and
the accretion of the final payment fee associated with the Loan Agreement (as
defined below).

Liquidity and Capital Resources

Sources of Liquidity



Since our inception, we have funded our operations primarily through sales of
our convertible preferred stock, sales of our common stock in public offerings
and under our "at-the-market" offering programs, private placements of our
common stock and pre-funded warrants, proceeds from the Loan Agreement (as
defined below), and our collaborations, including with the receipt of proceeds
under the 2seventy Agreement and the Gilead Collaboration Agreement, and
non-dilutive grants from various nonprofit organizations. As of September 30,
2022, we had cash, cash equivalents, and marketable securities of $139.8 million
and an accumulated deficit of $489.8 million, compared to cash, cash
equivalents, and marketable securities of $206.3 million and an accumulated
deficit of $401.4 million as of December 31, 2021. We expect that our cash, cash
equivalents, and marketable securities as of September 30, 2022

                                       40
--------------------------------------------------------------------------------


will enable us to fund our current and planned operating expenses and capital
expenditures for at least the next 12 months from the date of the filing of this
report.

In October 2019, we filed the 2019 Shelf Registration Statement, covering the
offering of up to $250.0 million of various equity and debt securities,
including the sale and issuance of up to $75.0 million worth of shares of our
common stock under the 2019 ATM Offering Program. Through September 30, 2022, we
have received aggregate proceeds from our 2019 ATM Offering Program of $50.0
million, net of commissions and offering costs, pursuant to the issuance of
5,642,712 shares.

In March 2022, we filed the 2022 Shelf Registration Statement, covering the
offering of up to $250.0 million of various equity and debt securities,
including the sale and issuance of up to $100.0 million worth of shares of our
common stock under the 2022 ATM Offering Program. As of September 30, 2022, we
have received $0.2 million in gross proceeds from our 2022 ATM Offering Program
and have $99.8 million available thereunder.

In April 2022, we received the second tranche payment of $2.7 million under the CEPI Funding Agreement.



In July 2022, we entered into a loan and security agreement (the "Loan
Agreement") with Hercules Capital, Inc. ("Hercules") and Silicon Valley Bank
("SVB"), which provides us with a 60-month term loan facility for the Company up
to $80.0 million in borrowing capacity across five potential tranches. At the
closing of the Loan Agreement, we drew $20.0 million from the first tranche, and
we can draw up to an additional $10.0 million through March 2023. The remaining
tranches provide up to $50.0 million borrowing capacity and become available if
and when we meet certain milestones set forth in the Loan Agreement. The term
loan is secured by substantially all of our assets, other than intellectual
property. There are no warrants associated with the Loan Agreement.

Borrowings under the Loan Agreement bear interest (i) at an annual cash rate
equal to the greater of (x) the lesser of (1) the prime rate (as customarily
defined) and (2) 5.50%, in either case, plus 3.15%, and (y) 7.15% and (ii) at an
annual payment-in-kind rate, which may equal 2.00%. We are required to make
monthly interest-only payments prior to the amortization date of January 1,
2025, subject to a potential six-month and one-year extension upon satisfaction
of certain conditions. We will also be required to pay a facility charge equal
to 0.50% of the principal amount of any borrowings made pursuant to the last
four tranches.

All unpaid principal and accrued and unpaid interest with respect to each term
loan is due and payable in full on July 19, 2027. At our option, we may prepay
all or any portion of the outstanding borrowings, plus accrued and unpaid
interest thereon and fees and expenses, subject to a prepayment premium ranging
from zero to 2.5%, during the first three years after closing, depending on the
year of such prepayment. Upon repayment of the term loan, we will be required to
make a final payment fee to the lenders equal to 5.75% of the aggregate original
principal amount of the loan.

Beginning on April 1, 2023, so long as our market capitalization is equal to or
less than $400.0 million, we are subject to a minimum liquidity requirement
equal to the then outstanding balance under the Loan Agreement multiplied by
0.55 or 0.45, which multiplier depends on whether we achieve certain performance
milestones. Our obligations under the Loan Agreement are subject to acceleration
upon the occurrence of customary events of default, including payment default,
insolvency and the occurrence of certain events having a material adverse
effect, including (but not limited to) material adverse effects upon the
business, operations, properties, assets or financial condition of us and our
subsidiaries, taken as a whole.

Future Funding Requirements



We do not expect positive cash flows from operations in the foreseeable future.
Historically, we have incurred operating losses as a result of ongoing efforts
to develop our cancer immunotherapy candidates, including conducting ongoing
research and development, clinical and preclinical studies and providing general
and administrative support for these operations. We do not have any products
approved for sale, and we do not expect to generate any meaningful revenue
unless and until we obtain regulatory approval of and commercialize any of our
current and future product candidates and/or enter into additional significant
collaboration or grant agreements with third parties, and we do not know when,
or if, either will occur. We expect to continue to incur net operating losses
for at least the next several years and we expect the losses to increase as we
advance our CORAL, GRANITE, and SLATE programs, as well as any future product
candidates, through clinical development, seek regulatory approval, prepare for
and, if approved, proceed to commercialization, continue our research and
development efforts and invest in our manufacturing facility. We are subject to
all the risks typically related to the development of new product candidates,
and we may encounter unforeseen expenses, difficulties, complications, delays
and other unknown factors that may adversely affect our

                                       41
--------------------------------------------------------------------------------


business. Moreover, we incur substantial costs associated with operating as a
public company. We anticipate that we will need substantial additional funding
in connection with our continuing operations.

Until we can generate a sufficient amount of revenue from the commercialization
of immunotherapy product candidates or from additional significant collaboration
or license agreements with third parties, if ever, we expect to finance our
future cash needs through private and public equity offerings, including our
"at-the-market" offering programs, debt financings, and potential future
collaboration, license and development agreements. Additional capital may not be
available on reasonable terms, if at all. If we are unable to raise additional
capital in sufficient amounts or on terms acceptable to us, we may have to
significantly delay, scale back or discontinue the development or
commercialization of one or more of our current or future product candidates. If
we raise additional funds by issuing equity or convertible debt securities, it
could result in dilution to our existing stockholders and increased fixed
payment obligations. In addition, as a condition to providing additional funds
to us, future investors may demand, and may be granted, rights superior to those
of existing stockholders. If we incur indebtedness, we could become subject to
covenants that would restrict our operations and potentially impair our
competitiveness, such as limitations on our ability to incur additional debt,
limitations on our ability to acquire, sell or license intellectual property
rights and other operating restrictions that could adversely impact our ability
to conduct our business. Additionally, any future collaborations we enter into
with third parties may provide capital in the near term, but we may have to
relinquish valuable rights to our product candidates or grant licenses on terms
that are not favorable to us. Any of the foregoing could significantly harm our
business, financial condition and prospects.

Since our inception, we have incurred significant losses and negative cash flows
from operations. We have an accumulated deficit of $489.8 million through
September 30, 2022. We expect to incur substantial additional losses in the
future as we conduct and expand our research and development activities. We
believe that our existing cash, cash equivalents and marketable securities will
be sufficient to enable us to fund our projected operations through at least the
next twelve (12) months from the date of this Quarterly Report on Form 10-Q. We
have based our projections of operating capital requirements on assumptions that
may prove to be incorrect and we may use all our available capital resources
sooner than we expect. Because of the numerous risks and uncertainties
associated with research, development and commercialization of product
candidates, we are unable to estimate the exact amount of our operating capital
requirements. Our future capital requirements depend on many factors, including:


the scope, progress, results and costs of developing our product candidates, and
of conducting preclinical studies and clinical trials, including our clinical
trials for GRANITE, SLATE and CORAL;


the timing of, and the costs involved in, obtaining regulatory approvals for our
oncology and infectious disease immunotherapy product candidates; in particular,
any costs incurred in connection with any future regulatory requirements that
may be imposed by the FDA or foreign regulatory bodies;

the number and characteristics of any additional product candidates we develop or acquire;

the timing and amount of any milestone, royalty or other payments we are required to make pursuant to any current or future collaboration or license agreements;

potential delays in our ongoing clinical trials as a result of the COVID-19 pandemic;

the cost of manufacturing our product candidates we successfully commercialize, including the cost of scaling up our internal manufacturing operations;

the cost of building a sales force in anticipation of product commercialization;

the cost of commercialization activities, including building a commercial infrastructure, marketing, sales and distribution costs;


our ability to maintain existing, and establish new, strategic collaborations,
licensing or other arrangements and the financial terms of any such agreements,
including the timing and amount of any future milestone, royalty or other
payments due under any such agreement;

any product liability or other lawsuits related to our products;

the costs to attract, hire and retain skilled personnel;

the costs associated with being a public company;


                                       42
--------------------------------------------------------------------------------

the costs involved in preparing, filing, prosecuting, maintaining, defending and enforcing our intellectual property portfolio; and

the timing, receipt and amount of sales of any future approved products, if any.



A change in the outcome of any of these or other variables with respect to the
development of any of our current and future product candidates could
significantly change the costs and timing associated with the development of
that product candidate. Furthermore, our operating plans may change in the
future, and we will need additional funds to meet operational needs and capital
requirements associated with such operating plans.

Cash Flows

The following table sets forth a summary of the primary sources and uses of cash for each of the periods presented below (in thousands):



                                                            Nine Months 

Ended September 30,


                                                              2022          

2021


Cash used in operating activities                       $        (85,572 )     $        (26,899 )
Cash provided by (used in) investing activities                   33,188                (71,927 )
Cash provided by financing activities                             18,733                 75,883
Net decrease in cash and cash equivalents               $        (33,651 )

$ (22,943 )

Cash Used in Operating Activities



During the nine months ended September 30, 2022, cash used in operating
activities was $85.6 million, which consisted of net loss of $88.4 million,
adjusted by non-cash charges of $21.6 million and net changes in our operating
assets and liabilities of $18.8 million. The non-cash charges consisted
primarily of depreciation and amortization expense of $4.8 million, stock-based
compensation of $9.5 million, non-cash operating lease expense of $6.9 million
and net amortization of premiums, discounts on marketable securities of $0.3
million, and amortization of debt discount and issuance costs of $0.1 million.
The change in our operating assets and liabilities was primarily due to
decreases of $11.6 million in deferred revenue, $0.2 million in accrued
compensation, $6.5 million in lease liability, $0.9 million in accounts payable,
and $3.2 million in deposits and other long term assets, offset by increases of
$1.5 million in accrued and other non-current liabilities, $1.3 million in
accrued research and development expenses, and $0.8 million in prepaid expenses
and other current assets.

During the nine months ended September 30, 2021, cash used in operating
activities was $26.9 million, which consisted of net loss of $45.3 million,
adjusted by non-cash charges of $18.8 million and net changes in our operating
assets and liabilities of $0.4 million. The non-cash charges consisted primarily
of depreciation and amortization expense of $4.8 million, stock-based
compensation of $7.8 million, non-cash operating lease expense of $5.7 million
and net amortization of premiums and discounts on marketable securities of $0.5
million. The change in our operating assets and liabilities was primarily due to
decreases of $5.9 million in lease liability, $0.9 million in accounts payable
and $0.2 million in accrued compensation, and increases of $3.5 million in
prepaid expenses and other current assets and $0.3 million in deposits and other
long-term assets, offset by increases $8.9 million in deferred revenue, $1.5
million in accrued research and development and $0.1 million in accrued and
other non-current liabilities.

Cash Provided by (Used in) Investing Activities



During the nine months ended September 30, 2022, cash provided by investing
activities was $33.2 million which consisted of $102.2 million in proceeds from
the maturity of marketable securities, offset by $64.6 million in purchases of
marketable securities and $4.4 million of capital expenditures to purchase
property and equipment.

During the nine months ended September 30, 2021, cash used in investing
activities was $71.9 million, which consisted of $133.4 million in purchases of
marketable securities and $4.1 million of capital expenditures to purchase
property and equipment, offset by $59.4 million in proceeds from the maturity of
marketable securities and $6.2 million from sales of marketable securities.

                                       43
--------------------------------------------------------------------------------

Cash Provided by Financing Activities



During the nine months ended September 30, 2022, cash provided by financing
activities was $18.7 million, which primarily consisted of $19.2 million in
proceeds from long-term debt, $0.2 million in proceeds from the 2022 ATM
Offering Program, $0.2 million in proceeds from the issuance of common stock
from option and warrant exercises and $0.3 million in proceeds from issuance of
common stock under the employee stock purchase plan, offset by $0.9 million in
tax withholding on vesting of restricted stock units, $0.1 million in payment of
financing costs, and $0.2 million in payment of financing lease.

During the nine months ended September 30, 2021, cash provided by financing
activities was $75.9 million, which primarily consisted of $21.2 million in
proceeds from the issuance of common stock pursuant to the Gilead Stock Purchase
Agreement, $55.0 million in proceeds from the Second PIPE Financing, $0.3
million in proceeds from the issuance of common stock under the employee stock
purchase plan, $2.3 million in proceeds from the issuance of common stock
through the 2019 ATM Offering Program and $3.1 million in proceeds from the
exercise of stock options, warrants and other, offset by $6.0 million in
financing and offering costs.

Off-Balance Sheet Arrangements

We have not entered into any off-balance sheet arrangements, as defined under SEC rules.

Contractual Obligations and Commitments



We lease office, laboratory and storage space in facilities at several locations
in California and Massachusetts. The terms of our lease agreements have
expiration dates between 2023 to 2033. The total future minimum lease payments
under the agreements are $108.1 million, of which $2.5 million of the payments
are due in the fourth quarter of 2022. See Note 6 to our condensed consolidated
financial statements.

We are party to license agreements pursuant to which we have in-licensed various
intellectual property rights. These license agreements obligate us to make
certain milestone payments related to achievement of specified events, as well
as royalties in the low-single digits based on sales of licensed products.
During the nine months ended September 30, 2022 and 2021, no royalties were due
from the sales of licensed products. The table above does not include any
milestone or royalty payments to the counterparties to these agreements as the
amounts, timing and likelihood of such payments are not known. See Note 9 to our
condensed consolidated financial statements for additional information.

In September 2017, we entered into a contract research and development agreement
with a third party CRO to provide research, analysis and antibody samples to
further the development of our antibody drug candidates. In June 2022, we
notified the CRO of our intent to terminate the agreement effective in August
2022. During the three and nine months ended September 30, 2022, we had no
research and development expense under this agreement. During the three and nine
months ended September 30, 2021, we had immaterial research and development
expense under this agreement. We are also obligated to pay the CRO certain
milestone payments of up to $36.4 million on achievement of specified events.
None of these events had occurred as of September 30, 2022. However, we are
unable to estimate the timing or likelihood of achieving the milestones and,
therefore, any related payments are not included in the table above.

In May 2019, we entered into a contract research and testing agreement with
another third-party CRO to provide antibody discovery related services. In March
2022, we notified such CRO of our intent to terminate the agreement effective as
of May 17, 2022. Under the agreement, we are obligated to pay such CRO certain
milestone payments of up to $34.8 million on achievement of specified events.
None of these events had occurred as of September 30, 2022. No research and
development expense was recorded under the agreement during the three and nine
months ended September 30, 2022 and 2021.

From time to time, in the normal course of business, we enter into contracts
with CROs for clinical trials, CMOs for clinical supply manufacturing and with
vendors for preclinical research studies and other services and products for
operating purposes, which generally provide for termination within 30 days of
notice. Therefore, all such contracts are cancelable contracts and not included
in the table above.

                                       44
--------------------------------------------------------------------------------

Critical Accounting Policies and Use of Estimates



This discussion and analysis of financial condition and results of operation is
based on our unaudited condensed consolidated financial statements, which have
been prepared in accordance with accounting principles generally accepted in the
United States, or U.S. GAAP. The preparation of financial statements requires
management to make estimates and judgments that affect the reported amounts of
assets and liabilities and disclosures of contingent assets and liabilities as
of the date of the financial statements and the reported amounts of expenses
during the reporting period. On an ongoing basis, management evaluates its
estimates, including those related to preclinical study trial accruals, fair
value of assets and liabilities, and the fair value of common stock and
stock-based compensation. Management bases its estimates on historical
experience and on various other market-specific and relevant assumptions that
management believes to be reasonable under the circumstances. Actual results
could differ from those estimates.

There have been no changes to our critical accounting policies since we filed
our Annual Report on Form 10-K for the year ended December 31, 2021 with the SEC
on March 10, 2022. For a description of our critical accounting policies, please
refer to our Annual Report on Form 10-K we filed with the SEC on March 10, 2022.

Recent Accounting Pronouncements



Refer to "Note 2. Summary of Significant Accounting Policies" in the notes to
our unaudited interim condensed consolidated financial statements in Part I,
Item 1 of this report, for a discussion of recent accounting pronouncements.

© Edgar Online, source Glimpses