Objective


The purpose of the following discussion and analysis is to provide material
information relevant to an assessment of our financial condition and results of
operations from management's perspective, including to describe and explain key
trends, events and other factors that impacted our reported results and that are
reasonably likely to impact our future performance.

As such, the following discussion and analysis of our financial condition and
results of operations should be read in conjunction with our condensed
consolidated financial statements and related notes appearing elsewhere in this
Quarterly Report on Form 10-Q and our Annual Report on Form 10-K for the year
ended December 31, 2021 filed with the Securities and Exchange Commission, or
SEC, on February 25, 2022. Some of the information contained in this discussion
and analysis or set forth elsewhere in this Quarterly Report on Form 10-Q,
including information with respect to our plans and strategy for our business,
includes forward-looking statements that involve risks and uncertainties. As a
result of many factors, including those factors set forth in the "Risk Factors"
section of this Quarterly Report on Form 10-Q, our actual results could differ
materially from the results described in, or implied by, the forward-looking
statements contained in the following discussion and analysis.

Overview



We are a clinical-stage biopharmaceutical company that is biologically
engineering red blood cells or RBCs, to develop an entirely new class of
cellular medicines called Red Cell Therapeutics, or RCTs, for the treatment of
cancer and autoimmune diseases. Based on our vision that human red blood cells
are the foundation of the next significant innovation in medicine, we have
developed a programmable and highly versatile platform, which we call the RED
PLATFORM, to biologically engineer and culture allogeneic cellular therapies
that enable multiple applications, or modalities. We believe that the advantage
of the platform is that once a modality is validated, as we have demonstrated
with our lead product candidate RTX-240 for the treatment of advanced cancers,
we increase the likelihood that all the programs within that modality will work,
underscoring the broad potential of the RED PLATFORM to help patients.

As part of the American Association for Cancer Research Annual Meeting in April
2022, we presented updated clinical data from our ongoing Phase 1 arm of RTX-240
in patients with locally advanced or relapsed/refractory solid tumors, which we
believe provides clinical validation for our RED PLATFORM and supports the
development of our entire oncology pipeline of RCTs. We continue to enroll
patients in our Phase 1 arm evaluating RTX-240 in combination with pembrolizumab
in patients with advanced solid tumors and have added an additional cohort to
evaluate the combination in patients with non-small cell lung cancer (NSCLC) and
renal cell carcinoma (RCC) to inform a Phase 2 clinical trial. We plan to report
initial clinical data for RTX-240 in combination with pembrolizumab in advanced
solid tumors and data from the initial NSCLC and RCC patients enrolled in the
expansion cohort during the second half of 2022. In January 2022, we began
dosing patients in the Phase 1/2 clinical trial of RTX-224, our second broad
immune agonist, for the treatment of patients with certain relapsed/refractory
or locally advanced solid tumors, including non-small cell lung cancer,
cutaneous melanoma, head and neck squamous cell carcinoma, urothelial (bladder)
carcinoma and triple-negative breast cancer. We expect to report initial
clinical results from the Phase 1 clinical trial of RTX-224 by year-end 2022 or
during the first quarter of 2023.

At our Investor Day in December 2021, we shared preclinical proof of concept
data, demonstrating tolerance induction and the potential for bystander
suppression in two stringent type 1 diabetes preclinical models. These findings
are potentially translatable beyond type 1 diabetes to multiple autoimmune
diseases, including our priority target indications such as multiple sclerosis
and celiac disease.

We continue to advance our manufacturing capabilities and achieved significant
manufacturing milestones, including successfully scaling our manufacturing to
200L bioreactors in support of a potential future pivotal trial for RTX-240 and
potential commercialization, as of April 2022. This results in a process at a
scale four times our previous 50L bioreactor process. We continue to provide
uninterrupted clinical supply for the RTX-240 clinical trial, the Phase 1
RTX-321 trial and the Phase 1 RTX-224 trial. We have the potential to
significantly expand our manufacturing capabilities and plan to

                                       14

Table of Contents

stage additional investments based on future needs and in preparation for potential pivotal trial and eventual commercialization.

Highlights of our clinical product candidates, RTX-240, RTX-321 and RTX-224 are described further below.

Broad Immune Stimulation for the Treatment of Cancer

RTX-240



In April 2022, we announced updated clinical data from the ongoing Phase 1/2
clinical trial of monotherapy RTX-240 in advanced solid tumors that we believe
provides clinical validation of the RED PLATFORM's potential ability to mimic
the human immune system and stimulate adaptive and innate immunity to generate
clinical responses in cancer patients with refractory disease. RTX-240 is an
allogeneic, off-the-shelf cellular therapy product candidate that is engineered
to simultaneously present hundreds of thousands of copies of the costimulatory
molecule 4-1BB ligand (4-1BBL) and IL-15TP (trans-presentation of IL-15 on
IL-15R?) in their native forms. RTX-240 is designed to broadly stimulate the
immune system by activating and expanding both NK and CD8+ memory T cells to
generate a potent anti-tumor response.

The data reported in April 2022 at the American Association for Cancer Research
Annual Meeting included initial safety (n=34) and efficacy (n=27) data from the
monotherapy RTX-240 Phase arm in relapsed/refractory or locally advanced solid
tumors. Nine dose cohorts were completed at the time of the data cutoff on March
4, 2022. Enrollment continues in the 5e10 Q3W dose cohort.

As of the cutoff date, disease control was observed in 10 patients (1 partial
response, 2 unconfirmed partial responses and 7 with stable disease), 9 of whom
had previously experienced disease progression on prior anti-PD-1/anti-PD-L1
therapy.

There were three best responses of partial response (PR) in NSCLC, anal cancer and uveal melanoma patients:

an unconfirmed PR (uPR) with 41% decrease of all target lesions and a notable ? decrease of an external protruding chest wall mass in a patient with NSCLC

whose disease had progressed on prior anti-PD-L1 therapy;

? a confirmed PR with a 54% reduction in the target lesions in a patient with

metastatic anal cancer whose disease had progressed on anti-PD-L1 therapy; and

a uPR with 100% decrease of the target hepatic lesion and resolution of ? multiple non-target hepatic lesions in a patient with metastatic uveal melanoma

whose disease had progressed on anti-PD-1 therapy.


Amount the patients with stable disease (SD), there were 3 with metastatic NSCLC
and 2 with RCC across the 3e10 cohorts, supporting the Company's decision to
expand the Phase 1 arm of RTX-240 plus pembrolizumab to NSCLC and RCC patients.
One patient each with NSCLC and RCC remained on monotherapy treatment with SD
greater than 6 months as of the cutoff date.

As of the cutoff date, RTX-240 has been shown to have been generally well tolerated with no treatment-related or investigator-identified immune-related Grade 3/4 adverse events (AEs) and no dose-limiting toxicities.


Based on the totality of clinical, tolerability and pharmacodynamic data, a
recommended monotherapy Phase 2 dose of 5e10 cells administered every 3 weeks
was selected. This dose will be further explored in the combination expansion
cohort of NSCLC and RCC patients. Enrollment continues in the monotherapy arm of
the trial at the recommended Phase 2 dose of 5e10 cells administered every 3
weeks.

In April 2022, we also announced final clinical results from the Phase 1 arm of
monotherapy RTX-240 in relapsed/refractory AML. As of the cutoff date of March
4, 2022, seventeen patients were enrolled across 4 dose levels. No DLTs were
observed and there were 3 treatment-related Grade 3/4 adverse events. There

were
no investigator-

                                       15

  Table of Contents

reported immune-related AEs. Five patients had SD greater than 3 months, and 1 patient had a significant blast count reduction (53% to 6%).



In this study, RTX-240 showed activation and expansion of NK and T cells with
favorable safety results, which continues to support the proposed mechanism of
action of RTX-240. Based on these data, we believe RTX-240 could improve
outcomes for AML patients when used as maintenance therapy for patients in
remission following high-dose chemotherapy and/or stem cell transplantation and
that we have established the necessary foundation to evaluate RTX-240 in the
maintenance setting for the treatment of AML. However, to focus our resources on
advancing RTX-240 in combination with pembrolizumab in NSCLC and RCC, we do not
plan to pursue a separate clinical trial in AML in the near-term.

In June 2021, we began dosing patients in the arm of our RTX-240 clinical trial
that is evaluating RTX-240 as a combination therapy with pembrolizumab for the
treatment of patients with relapsed/refractory or locally advanced solid tumors.
We believe that RTX-240, with its mechanism of action as a broad immune agonist,
may have synergy with immune checkpoint inhibition and could potentially
overcome resistance to PD-1 inhibition. Based on the updated clinical results
from the ongoing Phase 1 arm of monotherapy RTX-240 in advanced solid tumors, we
expanded the Phase 1 arm of RTX-240 in combination with pembrolizumab to
evaluate the combination in up to 20 patients with NSCLC and RCC to inform a
Phase 2 clinical trial. Patients with two or fewer prior treatment regimens in
the metastatic setting are eligible for the trial. If patients previously have
received a PD-1/PD-L1 regimen, a prior response of either SD ?6 months, PR or
complete response is required. We expect to report initial clinical results from
this arm of the ongoing Phase 1/2 clinical of RTX-240 in advanced solid tumors
and data from the initial enrolled NSCLC and RCC patients during the second

half
of 2022.

RTX-224

RTX-224 is an allogeneic cellular therapy that is engineered to express hundreds
of thousands of copies of 4-1BBL and interleukin-12, or IL-12, on the cell
surface. RTX-224 is designed as a broad immune agonist of both adaptive and
innate responses, designed to activate CD8+ and CD4+ T cells, activate and
expand NK cells, and promote antigen presentation. It is expected to produce a
broad and potent anti-tumor T cell response and an innate immune response and
have anti-tumor activity in those tumor types with known sensitivity to T cell
killing, including tumor types with high mutational burden, PD-L1 expression and
known responsiveness to checkpoint inhibitors. The combination of IL-12 and
4-1BBL has the potential to broadly induce an immune response in patients with
solid tumors and may serve as the bridge between the innate and adaptive immune
systems.

In January 2022, we began dosing patients in the Phase 1/2 clinical trial of
RTX-224 for the treatment of patients with certain relapsed/refractory or
locally advanced solid tumors, including non-small cell lung cancer, cutaneous
melanoma, head and neck squamous cell carcinoma, urothelial (bladder) carcinoma
and triple-negative breast cancer. We expect to report initial clinical results
from the Phase 1 trial by year-end 2022 or during the first quarter of 2023.

In November 2021, we presented preclinical data for RTX-224 at the Society for
Immunotherapy of Cancer's 36th Annual Meeting, showing that RTX-224 activated
immune cells in the spleen and blood, leading to their trafficking into the
tumor microenvironment to deliver an anti-tumor effect in our preclinical
models.

Antigen-Specific Immune Stimulation for the Treatment of Cancer

RTX-321



RTX-321 is an allogeneic, off-the-shelf artificial antigen presenting cells, or
aAPC, therapy product candidate that is engineered to induce a tumor-specific
immune response by expanding antigen-specific T cells. RTX-321 expresses
hundreds of thousands of copies of an HPV 16 peptide antigen bound to major
histocompatibility complex class I proteins, the costimulatory molecule 4-1BBL
and the cytokine IL-12 on the cell surface to mimic human T cell-APC
interactions.

                                       16

  Table of Contents

In May 2022, we announced that given the additional investment required to dose
escalate and the eventual need for a companion diagnostic for patient selection
for RTX-321, we are focusing our resources at this time on advancing our broad
agonism approach with RTX-240 and RTX-224. We believe RTX-321 has shown
promising pharmacodynamic effects with dramatic expansion of CD4+ T cells
observed, which is one of the key cells involved in the mechanism by which IL-12
stimulates a broad anti-tumor response. Importantly, RTX-321 was generally well
tolerated with no dose-limiting toxicities and no treatment-related AE's, giving
us confidence that we may be able to safely exploit IL-12's potent anti-tumor
activity with RTX-224, which expresses higher copy numbers of IL-12 on the cell
surface than does RTX-321.

Three dose cohorts were completed (n=9) with one patient with anal squamous cell
carcinoma with SD ongoing at 16 weeks at the highest dose cohort to date of 1e10
administered every three weeks. Prior to enrollment, the patient experienced
disease progression on anti-PD-1 therapy. Consistent with the combined mechanism
of action of IL-12 and 4-1BBL, increases in activated CD4+ T cells, activated
CD8+ T cells and activated NK cells were observed at the higher dose levels.

Antigen-Specific Immune Tolerance for the Treatment of Autoimmune Diseases

RTX-T1D (Type 1 Diabetes)



In December 2021, we shared preclinical proof of concept data, demonstrating
tolerance induction and the potential for bystander suppression in two stringent
type 1 diabetes preclinical models. Specifically, we established efficacy in the
BDC2.5 adoptive transfer model with data showing that an RTX-T1D surrogate
reversed established inflammation and induced two types of regulatory T cells,
resulting in protection against re-challenge. Moreover, the data showed that
repeated dosing could extend duration of disease protection to 5 months (the
endpoint of the study). We also showed early efficacy in the non-obese diabetic
mouse, or NOD, preclinical model. Results at 25 weeks showed that a mouse
surrogate of RTX-T1D that delivered only two antigens delayed disease. As
disease in NOD mice is caused by many autoantigens, these results demonstrate
the potential for bystander suppression. These findings are potentially
translatable beyond type 1 diabetes to multiple autoimmune diseases, including
other Rubius' high priority target indications such as multiple sclerosis and
celiac disease.

We intend to present these results in a peer-reviewed setting and expect to provide details of our development timeline later in 2022.

Manufacturing

Using our RED PLATFORM, we are utilizing our universal engineering and manufacturing processes to advance a broad pipeline of RCT product candidates into clinical trials in cancer and autoimmune diseases. Common design and manufacturing elements of our RCTs should enable us to achieve significant advantages in product development.


To more efficiently develop new RCTs designed to treat different diseases, we
modify one of our initial manufacturing steps in which we add a gene or genes of
interest that encode biotherapeutic proteins within the cell or on the cell
surface. Using this approach, we have expressed more than 1,000 different
therapeutic proteins since platform inception. This programmable process allows
for the repeated generation of product candidates and enables us to leverage
common CMC and toxicology data packages across our therapies.

Recognizing the importance of controlling our own manufacturing capabilities to
produce consistent and reproducible product at greater scale, we acquired,
renovated and operationalized a manufacturing facility in Smithfield, RI, which
is currently providing cGMP supply for our ongoing Phase 1 clinical trials:
RTX-240 in advanced solid tumors, RTX-240 in combination with pembrolizumab and
RTX-224 in advanced solid tumors.

We have industrialized the production of RCTs by developing and scaling up a
manufacturing process by which hematopoietic progenitor cells are expanded, then
biologically engineered and subsequently differentiated into erythroid cells
(RCTs) that express biotherapeutic proteins within the cell or on the cell
surface. The RED PLATFORM allows us to generate a wide variety of allogeneic,
ready-to-use RCT product candidates with a universal and proprietary process.

                                       17

  Table of Contents

We have recently achieved the following manufacturing milestones:

successfully scaled our manufacturing to 200L bioreactors in support of a ? potential future pivotal trial for RTX-240 and potential commercialization.

This results in a process at a scale four times our previous 50L bioreactor

process;

increased cells produced per batch in 50L bioreactors by four times that of ? 2020, enabling uninterrupted clinical supply for three Phase 1 arms of the

RTX-240 clinical trial; and

? introduced frozen drug substance for RTX-224, potentially enabling inventory

storage for more than two years.

Additional accomplishments include:

? greater than 90% lot success rate for RTX-240 and RTX-321 clinical supply in

the 50L bioreactor;

? hundreds of doses administered across all three arms of our RTX-240 Phase 1/2

trial, RTX-321 Phase 1 trial and RTX-224 Phase 1/2 trial;

? high transduction efficiency, with greater than 90% of cells transduced with

therapeutic proteins; and

? highly consistent protein expression (dual or triple).




We have the potential to significantly expand our manufacturing capabilities and
plan to stage additional investments based on future needs and in preparation
for potential pivotal trial and eventual commercialization.

Funding Overview


Since our inception, we have focused substantially all of our resources on
building our proprietary RED PLATFORM, establishing and protecting our
intellectual property portfolio, conducting research and development activities,
developing our manufacturing process and manufacturing product candidate
material, organizing and staffing our company, business planning, raising
capital and providing general and administrative support for these operations.
We do not have any products approved for sale and have not generated any revenue
from product sales. To date, we have funded our operations with proceeds from
the sale of preferred stock and issuance of debt and with proceeds from our
public offerings.

On July 20, 2018, we completed our IPO pursuant to which we issued and sold
12,055,450 shares of common stock, inclusive of 1,572,450 shares pursuant to the
full exercise of the underwriters' option to purchase additional shares. We
received proceeds of $254.3 million after deducting underwriting discounts and
commissions and other offering costs. In August 2019, we entered into a
Distribution Agreement with J.P. Morgan Securities LLC, Jefferies LLC and SVB
Leerink LLC with respect to an at-the-market, or ATM, offering program under
which we may offer and sell, from time to time at our sole discretion, shares of
our common stock, having aggregate gross proceeds of up to $100.0 million. We
have not yet sold any shares of our common stock under the ATM offering program.
In March 2021, we completed an underwritten public offering, or the March 2021
Offering, pursuant to which we issued and sold 6,896,552 shares of common stock.
We received proceeds of $187.2 million, after deducting underwriting discounts
and commissions and other offering costs.

Since our inception, we have incurred significant operating losses. Our ability
to generate any product revenue or product revenue sufficient to achieve
profitability will depend on the successful development and eventual
commercialization of one or more of our product candidates. We reported net
losses of $52.4 million for the three months ended March 31, 2022 and
$196.5 million for the year ended December 31, 2021. As of March 31, 2022, we
had an accumulated deficit of $729.4 million. We expect to continue to incur
significant expenses and operating losses for at

                                       18

Table of Contents

least the next several years. We expect that our expenses and capital requirements will increase in connection with our ongoing activities, particularly if, and as, we:

conduct clinical trials for our product candidates and to the extent we ? continue to experience delays, setbacks or disruptions to our preclinical

studies, clinical trials or clinical supply chain due to the ongoing COVID-19

pandemic;

? further develop our RED PLATFORM;

? continue to discover and develop additional product candidates;

? maintain, expand and protect our intellectual property portfolio;

? hire additional clinical, scientific, manufacturing and commercial personnel;

? expand in-house manufacturing capabilities, including through the operation and

any future renovation or expansion of our manufacturing facility;

establish a commercial manufacturing source and secure supply chain capacity ? sufficient to provide commercial quantities of any product candidates for which

we may obtain regulatory approval;

? acquire or in-license other product candidates and technologies;

? seek regulatory approvals for any product candidates that successfully complete

clinical trials;

? establish a sales, marketing and distribution infrastructure to commercialize

any products for which we may obtain regulatory approval; and

add operational, financial and management information systems and personnel, ? including personnel to support our product development and planned future

commercialization efforts, as well as to continue to support the requirements

of a public company.




We will not generate revenue from product sales unless and until we successfully
complete clinical development and obtain regulatory approval for our product
candidates. If we obtain regulatory approval for any of our product candidates,
we expect to incur significant expenses related to developing our
commercialization capability to support product sales, marketing and
distribution. Further, we expect to continue to incur costs associated with
operating as a public company.

As a result, we will need substantial additional funding to support our
continuing operations and pursue our growth strategy. Until such time as we can
generate significant revenue from product sales, if ever, we expect to finance
our operations through a combination of public and private equity financings,
debt financings, borrowings under our credit facility, collaborations, strategic
alliances and marketing, distribution and licensing arrangements. We may be
unable to raise additional funds or enter into such other agreements or
arrangements when needed on favorable terms, or at all. We implemented certain
cost reduction actions in April 2022, which are intended to focus our capital on
advancing our cancer and autoimmune programs and technology platform. If we are
unable to obtain additional funding, we will implement further cost reduction
actions that delay, scale back or discontinue some or all of our research and
development programs and technology platform activities in order to further
extend our forecasted cash runway. These actions could adversely affect our
business prospects. As of May 10, 2022, the issuance date of the interim
condensed consolidated financial statements, we expect that our cash, cash
equivalents and investments will be sufficient to fund our operating expenses,
capital expenditure requirements and debt service payments into the second half
of 2023.

Because of the numerous risks and uncertainties associated with pharmaceutical
product development, we are unable to accurately predict the timing or amount of
increased expenses or when, or if, we will be able to achieve or maintain

                                       19

Table of Contents

profitability. Even if we are able to generate product sales, we may not become profitable. If we fail to become profitable or are unable to sustain profitability on a continuing basis, then we may be unable to continue our operations at planned levels and be forced to reduce or terminate our operations.

See "-Liquidity and Capital Resources."

Impact of the Ongoing COVID-19 Pandemic



Since March of 2020 and throughout the ongoing COVID-19 pandemic, we have
implemented various precautionary measures to protect the health and safety of
our employees, partners and prospective clinical trial participants, to comply
with applicable national, state and local governmental orders, proclamations
and/or directives in effect at any time aimed at minimizing the spread of
COVID-19 and to minimize disruption to our operations. Such measures have
included, at certain times, the elimination of business travel, shifting to
remote work wherever possible and implementing rotating laboratory work
schedules to reduce the number of people onsite at our facilities, advance
ordering of certain raw materials impacted by delays in the global supply chain,
as well as working with our external partners and clinical sites to utilize
virtual clinical trial site training and monitoring, minimizing patient visits
and instituting telemedicine to minimize patient exposure. We will continue to
use these, and other precautionary measures, as required until such time as the
ongoing COVID-19 pandemic, including any subsequent outbreak whether or not due
to emerging variants thereof, is contained.

While the ongoing COVID-19 pandemic has impacted manufacturing, supply chain and
clinical trial activities worldwide, including those of our suppliers, vendors
and clinical trial sites, these disruptions have not significantly impacted our
results of operations to date. The ultimate impact on our operations, however,
is unknown and will depend on future developments, such as the duration, spread
and intensity of the pandemic, among others, which are highly uncertain and
cannot be predicted with confidence. In particular, global developments
concerning COVID-19, including the identification of new strains of coronavirus,
and the magnitude of interventions to contain the spread of viruses, such as
government-mandated quarantines, shelter-in-place mandates, restrictions on
travel, shutdowns for non-essential businesses, requirements regarding social
distancing, impact of government-imposed restrictions on the global supply
chain, including through use of the Defense Production Act, distribution of
vaccines and other public health safety measures, will determine the impact of
the pandemic on our business. We are continuing to monitor the latest
developments regarding the ongoing COVID-19 pandemic and its impact on our
business, financial condition, results of operations and prospects. However, any
resulting financial impact cannot be reasonably estimated at this time and may
have a material adverse impact on our business, financial condition and results
of operations.

Components of Our Results of Operations

Revenue



To date, we have not generated any revenue from product sales and do not expect
to generate any revenue from the sale of products in the near future. If our
development efforts for our product candidates are successful and result in
regulatory approval or license or collaboration agreements with third parties,
we may generate revenue in the future from product sales, payments from
collaboration or license agreements that we may enter into with third parties,
or any combination thereof.

Operating Expenses

Research and Development Expenses

Research and development expenses consist of costs incurred for our research activities, including our drug discovery efforts, and the development and manufacturing of our product candidates, which include:

employee-related expenses, including salaries, related benefits and stock-based ? compensation expense for employees engaged in research and development


  functions;


                                       20

  Table of Contents

expenses incurred in connection with the preclinical and clinical development ? of our product candidates and research programs, including under agreements

with third parties, such as consultants, contractors and contract research

organizations, or CROs;

the cost of developing and scaling our manufacturing process and manufacturing

product candidates for use in our preclinical studies and clinical trials, ? including those produced in our manufacturing facility as well as components

that are produced under agreements with third parties, such as consultants,

contractors and any contract manufacturing organizations, or CMOs, that we may

engage;

? laboratory supplies and research materials;

? facilities, depreciation and other expenses, which include direct and allocated

expenses for rent and maintenance of facilities and insurance; and

? payments made under third-party licensing agreements.




We expense research and development costs as incurred. Advance payments that we
make for goods or services to be received in the future for use in research and
development activities are recorded as prepaid expenses. The prepaid amounts are
expensed as the related goods are delivered or the services are performed.

Our direct research, manufacturing and development expenses are tracked on a
program-by-program basis for clinical candidates. These consist mostly of fees,
reimbursed materials, testing and other costs paid to consultants, contractors,
CMOs and CROs, as well as the cost of materials incurred for internal
manufacturing. In addition, we allocate the cost of operating our manufacturing
facility to research and development program costs, consisting of associated
personnel costs, other than stock-based compensation expense, and manufacturing
facility costs, including depreciation. We do not allocate costs associated with
our platform development, early-stage research and shared research and
development, including associated personnel costs, laboratory supplies,
non-manufacturing facilities expenses and other indirect costs, to research and
development programs, because these costs are deployed across multiple programs
and our technology platform and, as such, are not separately classified.

Product candidates in later stages of clinical development generally have higher
development costs than those in earlier stages of clinical development, due to
the increased size and duration of later stage clinical trials. Therefore, we
have reduced preclinical and other development activities to advance our current
clinical programs. As a result, we expect research and development expenses
related to those preclinical and other development activities to decrease while
we focus on achieving clinical endpoints. The successful development and
commercialization of our product candidates is highly uncertain. This is due to
the numerous risks and uncertainties associated with product development and
commercialization, including the following:

? the timing and progress of preclinical and clinical development activities;

? the number and scope of preclinical and clinical programs we decide to pursue;

? our ability to raise the additional funds necessary to complete preclinical and

clinical development of and commercialize our drug candidates;

? the progress of the development efforts of parties with whom we may enter into

collaboration arrangements;

? our ability to maintain our current research and development programs and to

establish new ones;

? our ability to establish new licensing or collaboration arrangements;

the successful initiation and completion of clinical trials with safety, ? tolerability and efficacy profiles that are satisfactory to the U.S. Food and

Drug Administration, or FDA, or any comparable foreign regulatory authority;




                                       21

  Table of Contents

? the continued impact of the ongoing COVID-19 pandemic on our operations;

? the receipt and related terms of regulatory approvals from applicable

regulatory authorities;

? the availability of specialty raw materials for use in production of our

product candidates;

? our ability to consistently manufacture our product candidates for use in

clinical trials;

? our ability to operate a manufacturing facility, or secure manufacturing supply

through relationships with third parties;

? our ability to obtain and maintain patents, trade secret protection and

regulatory exclusivity, both in the United States and internationally;

? our ability to protect our rights in our intellectual property portfolio;

? our ability to successfully commercialize our product candidates, if and when

approved;

? our ability to obtain and maintain third-party insurance coverage and adequate

reimbursement;

? the acceptance of our product candidates, if approved, by patients, the medical

community and third-party payors;

? competition with other products; and

? a continued acceptable safety profile of our therapies following approval.

A change in the outcome of any of these variables with respect to the development of any of our product candidates could significantly change the costs and timing associated with the development of that product candidate. We may never succeed in obtaining regulatory approval for any of our product candidates.

General and Administrative Expenses



General and administrative expenses include salaries and related costs,
including stock-based compensation, for personnel in executive, finance and
administrative functions. General and administrative expenses also include
direct and allocated facility-related costs, as well as professional fees for
legal, patent, consulting, investor and public relations, accounting and audit
services.

Other Income (Expense)

Interest Income

Interest income consists of interest earned on our invested cash balances.

Interest Expense

Interest expense consists of interest owed on outstanding borrowings under our Loan Agreement (as defined below), as well as amortization of debt discount.

Other Income, Net



Other income, net consists of miscellaneous income and expense unrelated to our
core operations.

                                       22

  Table of Contents

Income Taxes

Since our inception, we have not recorded any income tax benefits for the net
losses we have incurred in each year or for our research and development tax
credits generated, as we believe, based upon the weight of available evidence,
that it is more likely than not that all of our net operating loss, or NOL,
carryforwards and tax credits will not be realized. As of December 31, 2021, we
had U.S. federal and state net operating loss carryforwards of $534.2 million
and $534.8 million, respectively, which may be available to offset future
taxable income. The federal NOLs include $37.2 million, which expire at various
dates through 2037, and $497.0 million, which carryforward indefinitely. The
state NOLs expire at various dates through 2041. As of December 31, 2021, we
also had U.S. federal and state research and development tax credit
carryforwards of $22.7 million and $15.6 million, respectively, which may be
available to offset future tax liabilities and begin to expire in 2034 and 2026,
respectively. We have recorded a full valuation allowance against our net
deferred tax assets at each balance sheet date.

Results of Operations

Comparison of the Three Months Ended March 31, 2022 and 2021

The following table summarizes our results of operations for the three months ended March 31, 2022 and 2021:



                                         Three Months Ended March 31,
                                          2022                      2021       Change

                                                      (in thousands)
Revenue                             $              -             $        -  $        -
Operating expenses:
Research and development                      38,299                 27,677      10,622
General and administrative                    12,563                 13,240       (677)
Total operating expenses                      50,862                 40,917       9,945
Loss from operations                        (50,862)               (40,917)     (9,945)
Other income (expense):
Interest income                                   48                     26          22
Interest expense                             (1,629)                (1,748)         119
Other income, net                                 31                    309       (278)

Total other income (expense), net            (1,550)                (1,413)

      (137)
Net loss                            $       (52,412)             $ (42,330)  $ (10,082)

Research and Development Expenses



                                                               Three Months Ended March 31,
                                                              2022                          2021       Change

                                                                            (in thousands)
Research and development program expenses:
Rare disease                                            $               -                 $     197   $  (197)
Cancer                                                             17,011                    11,742      5,269
Platform development, early-stage research and
unallocated expenses:
Personnel-related                                                   8,832                     6,388      2,444
Stock-based compensation expense                                    3,477                     2,531        946
Contract research and development                                   2,605                     1,328      1,277
Laboratory supplies and research materials                          2,725                     1,759        966
Facility related and other                                          3,649                     3,732       (83)
Total research and development expenses                 $          38,299                 $  27,677   $ 10,622


Research and development expenses were $38.3 million for the three months ended
March 31, 2022, compared to $27.7 million for the three months ended March 31,
2021. The increase in direct costs of $5.3 million related to RTX-240, RTX-321
and RTX-224, was principally related to clinical research organization, or

CRO,
costs and internal

                                       23

  Table of Contents

manufacturing costs incurred in connection with all three programs. We have
prioritized the advancement of clinical endpoints and, therefore, expect these
costs to decrease in future periods. Platform development, early-stage research
and unallocated expenses increased by $5.6 million principally due to increases
of $2.4 million in personnel-related costs and $0.9 million in stock-based
compensation related to the increase in headcount to support our expanded
operations. Additionally, increases of $1.3 million in contract research and
development and $1.0 million in laboratory supplies and research materials are
related to drug discovery activities and platform development.

General and Administrative Expenses



                                                               Three Months Ended March 31,
                                                              2022                          2021       Change

                                                                            (in thousands)
Personnel-related                                       $           3,486                 $   3,125   $     361
Stock-based compensation expense                                    4,783                     6,111     (1,328)
Professional and consultant fees                                    2,632                     2,275         357
Facility related and other                                          1,662                     1,729        (67)
Total general and administrative expenses               $          12,563                 $  13,240   $   (677)


General and administrative expenses were $12.6 million for the three months
ended March 31, 2022, compared to $13.2 million for the three months ended March
31, 2021. The decrease in general and administrative expenses of $0.7 million
was primarily due to a decrease in stock-based compensation expense of $1.3
million, which was driven by stock option awards that fully vested during the
third quarter of 2021. The reduction in stock-based compensation was offset by
an increase in personnel-related expenses of $0.4 million driven by additions to
headcount in our general and administrative function and an increase in
professional and consultant fees of $0.4 million resulting primarily from
additional costs to support operations as a public company.

Interest Income


Interest income was less than $0.1 million for the three months ended March 31,
2022, compared to less than $0.1 million for the three months ended March 31,
2021. The change in interest income was not significant during the period.

Interest Expense



Interest expense was $1.6 million for the three months ended March 31, 2022,
compared to $1.7 million for the three months ended March 31, 2021. The change
in interest expense was not significant during the period.

Other Income, Net



Other income, net was less than $0.1 million for the three months ended March
31, 2022, compared to $0.3 million for the three months ended March 31, 2021.
The decrease in other income, net was due to the expiration of our sublease
agreement in the third quarter of 2021, concurrent with the expiration of the
associated lease.

Liquidity and Capital Resources


Since our inception, we have incurred significant operating losses. We have not
yet commercialized any of our product candidates and we do not expect to
generate revenue from sales of any product candidates for several years, if at
all. To date, we have funded our operations with proceeds from the sale of
preferred stock, with the issuance of debt, with proceeds from our IPO and, most
recently, with proceeds from our March 2021 Offering, described and defined
further below. As of March 31, 2022, we had cash, cash equivalents and
investments of $176.5 million. In July 2018, we completed our IPO, pursuant to
which we issued and sold 12,055,450 shares of common stock, inclusive of
1,572,450 shares pursuant to the full exercise of the underwriters' option to
purchase additional shares. We received proceeds of $254.3 million, after
deducting underwriting discounts and commissions and other offering costs. In
December 2018, we entered into a loan and security agreement, which was amended
in June 2021, which provides for aggregate borrowings of up to $75.0 million. As
of March 31, 2022, $75.0 million is outstanding under the agreement and
principal payments

                                       24

  Table of Contents

commence in July 2024. In March 2021, we completed the March 2021 Offering, pursuant to which we issued and sold 6,896,552 shares of common stock. We received proceeds of $187.2 million, after deducting underwriting discounts and commissions and other offering costs.

Cash Flows



The following table summarizes our sources and uses of cash for each of the
periods presented:

                                                                  Three Months Ended March 31,
                                                                   2022                       2021
                                                                         (in thousands)
Cash used in operating activities                            $        (47,135)            $ (38,739)
Cash provided by (used in) investing activities                       (80,847)                41,752
Cash provided by financing activities                                       76               193,908

Net increase in cash, cash equivalents and restricted cash $ (127,906)

$  196,921

Operating Activities



During the three months ended March 31, 2022, operating activities used
$47.1 million of cash, primarily resulting from our net loss of $52.4 million,
offset by net non-cash charges of $10.0 million, predominantly consisting of
stock-based compensation expense. Net cash used in our operating assets and
liabilities for the three months ended March 31, 2022 consisted of a $6.3
million decrease in accounts payable, accrued expenses and other current
liabilities, other long-term liabilities and operating lease liabilities, offset
by a decrease in prepaid expenses and other current assets and operating lease,
right-of-use asset of $1.5 million.

During the three months ended March 31, 2021, operating activities used
$38.7 million of cash, primarily resulting from our net loss of $42.3 million,
offset by net non-cash charges of $10.9 million, predominantly consisting of
stock-based compensation expense. Net cash used in our operating assets and
liabilities for the three months ended March 31, 2021 consisted of a $8.3
million decrease in accounts payable, accrued expenses and other current
liabilities, other long-term liabilities and operating lease liabilities, offset
by a decrease in prepaid expenses and other current assets and operating lease,
right-of-use asset of $1.0 million.

Investing Activities



During the three months ended March 31, 2022, net cash used in our investing
activities was $80.8 million, consisting of purchases of investments of $78.4
million and purchases of property, plant and equipment of $2.4 million. Our cash
purchases of property, plant and equipment primarily relate to renovation
expenditures incurred at our manufacturing facility in Smithfield, Rhode Island.

During the three months ended March 31, 2021, net cash provided by investing
activities was $41.8 million, consisting of sales and maturities of investments
of $42.5 million, offset by purchases of property, plant and equipment of
$0.7 million. Our cash purchases of property, plant and equipment relate to the
purchase of computer and laboratory equipment installed in our manufacturing
facility in Smithfield, Rhode Island and our laboratory space in Cambridge,
Massachusetts.

Financing Activities

During the three months ended March 31, 2022, net cash provided by financing activities of $0.1 million consisted of proceeds received from issuance of common stock upon exercise of stock options of $0.1 million.



During the three months ended March 31, 2021, net cash provided by financing
activities of $193.9 million consisted primarily of proceeds of $188.0 million,
net of commissions and underwriting discounts, and proceeds received from
issuance of common stock upon exercise of stock options of $5.9 million from the
Offering completed in March 2021.

                                       25

Table of Contents

Contractual Obligations and Commitments


The following table summarizes our contractual obligations as of March 31, 2022
and the effects that such obligations are expected to have on our liquidity and
cash flows in future periods:

                                                                     Payments Due by Period
                                                          Less Than                                      More Than
                                                Total       1 Year      1 to 3 Years     3 to 5 Years     5 Years

                                                                         (in thousands)
Operating lease commitments (1)               $  44,298   $    9,082   $       15,097   $        7,873   $   12,246
Debt obligations (2)                             97,739        5,700       

   38,813           53,226            -
Total                                         $ 142,037   $   14,782   $       53,910   $       61,099   $   12,246

(1)Amounts in table reflect payments due for our leases of office and laboratory space in Cambridge, Massachusetts under two operating lease agreements that expire in January 2027 and August 2028, respectively.



(2)Amounts in table reflect the contractually required principal and interest
payments payable under the Loan Agreement. For purposes of this table, the
interest due under the Loan Agreement was calculated using an assumed interest
rate of 8.86% per annum, which was the interest rate in effect as of March

31,
2022.

Loan and Security Agreement

In December 2018, or the Closing Date, we entered into a loan and security
agreement, or, as amended, the Loan Agreement, with SLR Investment Corp.
(formerly Solar Capital Ltd.) as collateral agent for the lenders party thereto
for an aggregate principal amount of $75.0 million. The aggregate principal
amount was funded in three tranches of term loans of $25.0 million each, on the
Closing Date, in June 2019 and in June 2020.

On June 22, 2021, or the Amendment Closing Date, we entered into an amendment,
or the Amendment, to our Loan Agreement. Pursuant to the Amendment, we and our
lenders agreed to extend the interest-only period in respect of our borrowings
under the Loan Agreement from December 21, 2021 until July 1, 2024. The parties
also agreed to extend the final maturity date on which all of our outstanding
obligations under the Loan Agreement become due to June 1, 2026 (from December
21, 2023 originally). An additional tranche in the amount of $35.0 million is
available to us prior to the final maturity date, to be provided at the sole
discretion of the lenders. Interest on the outstanding loan balance will accrue
at a rate of 5.50%, plus the greater of 2.10% or the one-month U.S. LIBOR rate.
Monthly principal payments will commence on July 1, 2024 and will be amortized
over the following 24 months. Certain back-end fees are due to the lender at the
time of final repayment based on the total funded term loans. The term loans are
subject to a prepayment fee of 1.00% if prepayment occurs within the first year
subsequent to the Amendment Closing Date, 0.50% in the second year and 0.25% in
the third year through final maturity date.

The Loan Agreement contains financial covenants that require us to maintain
either a certain minimum cash balance or a minimum market capitalization
threshold. We were in compliance with all such financial covenants as of March
31, 2022. The Loan Agreement contains customary representations, warranties and
covenants and also includes customary events of default, including payment
defaults, breaches of covenants, change of control and a material adverse change
default. Upon the occurrence of an event of default, a default interest rate of
an additional 4.00% per annum may be applied to the outstanding loan balances,
and the lenders may declare all outstanding obligations immediately due and
payable. Borrowings under the Loan Agreement are collateralized by substantially
all of our assets, other than our intellectual property.

Common Stock Sales Agreement



On August 1, 2019, we entered into a Distribution Agreement, or the Distribution
Agreement, with multiple sales agents, pursuant to which the Company may offer
and sell to or through the agents, from time to time, shares of the Company's
common stock, par value $0.001 per share, having an aggregate gross sales price
of up to $100.0 million. Sales, if any, of the Company's shares of common stock
will be made primarily in "at-the-market" offerings, as defined in Rule 415
under the Securities Act. The shares of common stock will be offered and sold
pursuant to our registration statement on

                                       26

Table of Contents



Form S-3 and a related prospectus supplement, both filed with the SEC on August
1, 2019. We intend to use substantially all of the net proceeds from any sale of
shares of the Company's common stock for working capital and other general
corporate purposes. There have been no shares of the Company's common stock sold
under the Distribution Agreement as of March 31, 2022.

Funding Requirements



We expect our expenses to increase substantially in the future as we conduct the
activities necessary to advance our product candidates through development. The
timing and amount of our operating and capital expenditures will depend largely
on:

? the timing and progress of preclinical and clinical development activities;

the commencement, enrollment or results of the planned clinical trials of our ? product candidates or any future clinical trials we may conduct, or changes in

the development status of our product candidates;

? the timing and outcome of regulatory review of our product candidates;

the continued impact of the ongoing COVID-19 pandemic, including from any ? subsequent outbreak whether or not due to emerging variants thereof, on our

operations;

? our decision to initiate a clinical trial, not to initiate a clinical trial or

to terminate an existing clinical trial;

? changes in laws or regulations applicable to our product candidates, including

but not limited to clinical trial requirements for approvals;

? developments concerning our key vendors;

? our ability to obtain materials to produce adequate product supply for any

approved product or inability to do so at acceptable prices;

the costs associated with the operation of our multi-suite manufacturing ? facility and the costs and timing of any future renovation or expansion of the

facility;

? our ability to establish collaborations if needed;

the costs and timing of future commercialization activities, including product ? manufacturing, marketing, sales and distribution, for any of our product

candidates for which we obtain marketing approval;

? the legal patent costs involved in prosecuting patent applications and

enforcing patent claims and other intellectual property claims;

? additions or departures of key scientific or management personnel;

? unanticipated serious safety concerns related to the use of our product

candidates; and

? the terms and timing of any collaboration, license or other arrangement,

including the terms and timing of any milestone payments thereunder.


We believe that our existing cash, cash equivalents and investments will be
sufficient to fund our operating expenses, capital expenditure requirements and
debt service payments into the second half of 2023. We have based this estimate
on assumptions that may prove to be wrong, and we could utilize our available
capital resources sooner than we expect.

                                       27

Table of Contents



Until such time, if ever, as we can generate substantial product revenue, we
expect to finance our operations through a combination of public and private
equity financings, debt financings, collaborations, strategic alliances and
marketing, distribution and licensing arrangements. To the extent that we raise
additional capital through the sale of equity or convertible debt securities,
investors' ownership interest will be diluted, and the terms of these securities
may include liquidation or other preferences that adversely affect investors'
rights as a common stockholder. Debt financing and preferred equity financing,
if available, may involve agreements that include covenants limiting or
restricting our ability to take specific actions, such as incurring additional
debt, making acquisitions or capital expenditures or declaring dividends. If we
raise additional funds through collaborations, strategic alliances or marketing,
distribution or licensing arrangements with third parties, we may have to
relinquish valuable rights to our technologies, future revenue streams, research
programs or drug candidates, or grant licenses on terms that may not be
favorable to us. We implemented certain cost reduction actions in April 2022,
which are intended to focus our capital on advancing our cancer and autoimmune
programs and technology platform. If we are unable to obtain additional funding,
we will implement further cost reduction actions that delay, scale back or
discontinue some or all of our research and development programs and technology
platform activities in order to further extend our forecasted cash runway. These
actions could adversely affect our business prospects.

Off-Balance Sheet Arrangements


We did not have during the periods presented, and we do not currently have, any
off-balance sheet arrangements, as defined in the rules and regulations of the
SEC.

Critical Accounting Policies and Significant Judgments and Estimates


Our condensed consolidated financial statements are prepared in accordance with
generally accepted accounting principles in the United States, or GAAP. The
preparation of our condensed consolidated financial statements and related
disclosures requires us to make estimates, assumptions and judgments that affect
the reported amounts of assets, liabilities, revenue, costs and expenses, and
related disclosures. Our critical accounting policies are described under the
heading "Management's Discussion and Analysis of Financial Condition and Results
of Operations-Critical Accounting Policies and Significant Judgments and
Estimates" in our Annual Report on Form 10-K for the year ended December 31,
2021, filed with the SEC on February 25, 2022. If actual results or events
differ materially from the estimates, judgments and assumptions used by us in
applying these policies, our reported financial condition and results of
operations could be materially affected.

There have been no significant changes to our critical accounting policies from
those described in our Annual Report on Form 10-K for the year ended December
31, 2021 filed with the SEC on February 25, 2022.

Recently Issued Accounting Pronouncements



A description of recently issued accounting pronouncements that may potentially
impact our financial position and results of operations is disclosed in Note 2
to our condensed consolidated financial statements appearing elsewhere in this
Quarterly Report on Form 10-Q.

© Edgar Online, source Glimpses