The following discussion contains forward-looking statements that involve risks
and uncertainties. Our actual results could differ materially from those
discussed here. Factors that could cause or contribute to such differences
include, but are not limited to, those discussed in this section as well as
factors described in Part II, Item 1A - "Risk Factors" and "Special Note
Regarding Forward-Looking Statements" included elsewhere in this Quarterly
Report on Form 10-Q.
Overview
Dicerna Pharmaceuticals, Inc. ("we", "us," "our," "the Company," or "Dicerna")
is a biopharmaceutical company focused on discovering, developing, and
commercializing medicines that are designed to leverage ribonucleic acid
interference ("RNAi") to silence genes selectively that cause or contribute to
disease. Using our proprietary GalXC™ RNAi technology platform, Dicerna is
committed to developing RNAi-based therapies with the potential to treat both
rare and more prevalent diseases. By silencing disease-causing genes, Dicerna's
GalXC platform has the potential to address conditions that are difficult to
treat with other modalities. Initially focused on hepatocytes, Dicerna has
continued to innovate and is exploring new applications of its RNAi technology
beyond the liver, targeting additional tissues and enabling new therapeutic
applications. In addition to our own pipeline of core discovery and clinical
candidates, Dicerna has established collaborative relationships with some of the
world's leading pharmaceutical companies, including Novo Nordisk A/S ("Novo"),
Roche, Eli Lilly and Company ("Lilly"), Alexion Pharmaceuticals, Inc. (together
with its affiliates, "Alexion"), Boehringer Ingelheim International GmbH ("BI"),
and Alnylam Pharmaceuticals, Inc. ("Alnylam"). Between Dicerna and our
collaborative partners, we currently have more than 20 active discovery,
preclinical, or clinical programs focused on rare, cardiometabolic, viral,
chronic liver, and complement-mediated diseases, as well as neurodegeneration
and pain.
All of our drug discovery and development efforts are based on the therapeutic
modality of RNAi, a highly potent and specific mechanism for silencing the
activity of a targeted gene. In this naturally occurring biological process,
double-stranded RNA molecules induce the enzymatic destruction of the messenger
ribonucleic acid ("mRNA") of a target gene that contains sequences that are
complementary to one strand of the therapeutic double-stranded RNA molecule. Our
approach is to design proprietary double-stranded RNA molecules that have the
potential to engage the enzyme Dicer and initiate an RNAi process to silence a
specific target gene. Our GalXC RNAi platform utilizes a proprietary
GalNAc-mediated structure of double-stranded RNA molecules. For our current
clinical programs, our GalXC RNAi platform has been configured for subcutaneous
delivery to the liver. Due to the enzymatic nature of RNAi, a single GalXC
molecule incorporated into the RNAi machinery can destroy hundreds or thousands
of mRNAs from the targeted gene.
The GalXC RNAi platform and other proprietary RNAi delivery technologies support
Dicerna's long-term strategy to retain a full or substantial ownership stake in
our programs, subject to the evaluation of potential licensing opportunities as
they may arise, and to invest internally in programs for diseases with focused
patient populations, such as certain rare diseases or diseases with
well-characterized genetic targets. These certain rare disease programs, which
include our nedosiran and A1AT product(s) programs, represent opportunities that
we believe carry a relatively higher probability of success, with genetically
and molecularly defined disease markers, high unmet medical need, a limited
number of centers of excellence to facilitate reaching these patients, and the
potential for more rapid clinical development paths to regulatory approval. For
more complex diseases with multiple gene dysfunctions and/or larger patient
populations, we continue to pursue collaborations that can provide the enhanced
scale, resources, and commercial infrastructure required to maximize these
prospects.
We currently view our operations and manage our business as one segment which
encompasses the discovery, research, and development of treatments based on our
RNAi technology platform.
Executive Summary
Our results of operations for and liquidity and capital resources as of the nine
months ended September 30, 2020 include the following:
•  In January 2020, we entered into a non-cancelable real property lease
agreement for 61,282 square feet of office space at 75 Hayden Avenue in
Lexington, Massachusetts. The original term is estimated to commence during the
fourth quarter of 2020 and is for 125 months with options to extend for two
additional successive periods of five years thereafter. The aggregate total
fixed rent is approximately $41.8 million. In addition, in July 2020, we entered
into an amendment to the 75 Hayden Avenue lease. The 75 Hayden Avenue lease
amendment expands the square footage leased under the 75 Hayden Avenue lease to
contain a total of 91,728 rentable square feet. The 75 Hayden Avenue lease
amendment increases monthly base rent by an average of $0.2 million per month.
                                       28
--------------------------------------------------------------------------------
  Table of Contents
•  In January 2020, we received a $200.0 million upfront payment from Roche
associated with a collaboration agreement executed in October 2019 to which we
were a party. The agreement with Roche is to progress RG6346, the
investigational therapy in Phase 1 clinical development, toward worldwide
development and commercialization, and includes an option for the companies to
collaborate in the discovery, development, and commercialization of
oligonucleotide therapeutics intended for the treatment of chronic hepatitis B
virus ("HBV") infection. In April 2020, Roche selected its first target under
the research and development portion of the agreement.
•  In January 2020, we received a $175.0 million upfront payment from Novo
associated with a collaboration agreement executed in November 2019 to which we
were a party. The agreement with Novo is for the use of the Company's
proprietary GalXC platform to progress novel therapies for the treatment of
liver-related cardiometabolic diseases towards clinical development and
commercialization.
•In February 2020, we issued and sold approximately $40.0 million of shares of
our common stock to a single institutional investor through our "at-the-market"
sales facility with Cowen and Company, LLC. In this transaction, we sold an
aggregate of 2,077,500 shares of common stock at a price of $19.25 per share,
resulting in approximately $39.2 million after a deduction of approximately $0.8
million in sales commissions. The shares in the offering were sold pursuant to a
shelf registration statement declared effective by the Securities and Exchange
Commission ("SEC") on May 31, 2018 and a prospectus supplement filed with the
SEC on June 1, 2018.
•In April 2020, we and Alnylam entered into a collaboration and license
agreement (the "A1AT Agreement") and a patent cross-license agreement (the "PH
Agreement"). Under the A1AT Agreement, we and Alnylam will work to develop and
commercialize investigational RNAi therapeutics for the treatment of alpha-1
antitrypsin ("A1AT") deficiency-associated liver disease ("alpha-1 liver
disease"). Under the PH Agreement, we and Alnylam cross-license our respective
intellectual property related to Alnylam's lumasiran and our nedosiran
investigational programs for the treatment of primary hyperoxaluria ("PH"). The
non-exclusive license agreement provides for Alnylam to pay mid- to
high-single-digit royalties to Dicerna based on global net sales of lumasiran
and for Dicerna to pay low-single-digit royalties to Alnylam on global net sales
of nedosiran. The non-exclusive cross-license agreement ensures that each party
has the freedom to develop and commercialize its respective product candidate.
•In June 2020, the Food and Drug Administration ("FDA") granted rare pediatric
disease designation for nedosiran for the treatment of PH. Under the FDA's rare
pediatric disease designation program, the FDA may grant a priority review
voucher to a Sponsor who receives a product approval for a "rare pediatric
disease." Subject to FDA approval of nedosiran for the treatment of PH, we would
be eligible to receive a voucher that may be redeemed to receive priority review
for a subsequent marketing application for a different product candidate or
which could be sold or transferred.
•In July 2020, we entered into a second amendment to the lease agreement for our
facility in Colorado. The Colorado facility lease amendment approves Dicerna to
operate a biotechnology laboratory on the premises.
•In August 2020, we provided updated guidance related to the enrollment
completion timeline for our PHYOX2 pivotal trial of nedosiran in patients with
PH after previously removing formal guidance due to COVID-19 impacts. Revised
guidance anticipates enrollment completion in PHYOX2 in the fourth quarter of
2020.
•We believe we have sufficient capital, along with anticipated milestone and
other payments from existing collaborations, to fund the execution of our
current clinical and operating plans into 2023.
•The following table provides a summary of revenue recognized for the three and
nine months ended September 30, 2020 and 2019:
                 THREE MONTHS ENDED            NINE MONTHS ENDED
                   SEPTEMBER 30,                 SEPTEMBER 30,
                 2020           2019          2020           2019
Novo         $     3,552      $     -      $   7,604      $      -
Roche             18,686            -         58,670             -
Lilly             12,073        5,274         31,142         8,346
Alexion           13,639        1,451         23,464         2,954
BI                   925        1,310          2,471         5,524

Total        $    48,875      $ 8,035      $ 123,351      $ 16,824


COVID-19 Update
On March 11, 2020, the World Health Organization declared COVID-19 a pandemic.
The global spread of COVID-19 has created significant volatility, uncertainty,
and economic disruption worldwide. Governments in affected regions have
implemented,
                                       29
--------------------------------------------------------------------------------
  Table of Contents
and may continue to implement, safety precautions which include quarantines,
travel restrictions, business closures, and other public health safety measures.
We have been impacted by mandatory work from home edicts directed by local
governments in the jurisdictions in which we operate. However, essential work
exemptions continue to permit critical research and development and laboratory
activities for limited personnel. Those exemptions enable some continued
discovery research and activities supporting our collaborative agreements and
our own programs. Externally, the COVID-19 pandemic has resulted in slower
enrollment in our clinical trials, and we have undertaken efforts to mitigate
potential impacts to our business including those related to conducting clinical
trials and managing our supply chain. Our operating results could be affected by
delays or suspensions of clinical development associated with COVID-19, which
have impacted and may continue to impact global healthcare systems and our trial
sites' enrollment in our clinical trials, such as we have seen in the nedosiran
pivotal PHYOX2 and DCR-A1AT Phase 1/2 studies, and delays in the supply chain
related to COVID-19. We continue to be alert to the potential for disruptions
that could arise from COVID-19 and monitor the Food and Drug Administration's
and other health authorities' guidance for the conduct of clinical trials during
this time.
The current supply of our investigational medicines continues to be sufficient
to support ongoing and planned clinical trials. Based on current evaluations,
our supply chain continues to appear intact at this time and able to meet the
next 12 months of clinical, nonclinical, and chemistry, manufacturing, and
control ("CMC") supply demands across all programs. We have undertaken efforts
to mitigate potential future impacts to the supply chain by increasing our stock
of critical starting materials required to meet our needs and our collaborative
partners' needs through 2021 and by identifying and engaging alternative
suppliers. We continue to be alert to the potential for disruptions that could
arise from COVID-19 and remain in close contact with suppliers.
It is difficult to predict what the lasting impact of the pandemic will be, and
if we or any of the third parties with whom we engage were to experience
additional shutdowns or other prolonged business disruptions. Our ability to
conduct our business in the manner and on the timelines presently planned could
have a material adverse impact on our business, results of operations, and
financial condition. In addition, a recurrence or "second wave" of COVID-19
cases could cause other widespread or more severe impacts depending on where
infection rates are highest. We will continue to monitor developments as we deal
with the disruptions and uncertainties relating to the COVID-19 pandemic. Please
refer to the "Financial Operations Overview" section below for specific
anticipated effects on our financial statement line items.
Development Programs
In choosing which development programs to internally advance, we apply the
scientific, clinical, and commercial criteria that we believe allow us to best
leverage our GalXC RNAi platform and maximize value. Using our GalXC RNAi
technology, and applying the criteria of our development focus, we have created
a pipeline of core liver-focused therapeutic programs for development by
Dicerna. For opportunities that were not selected as a core program opportunity,
we have sought partners to fund the discovery, and subsequently drive the
development of, these non-core opportunities in exchange for upfront payments,
milestone payments, royalties on product sales, and potentially other economic
and operational arrangements. Our current collaborations with Novo, Lilly,
Alexion, and BI resulted from this effort. For core programs targeting rare
diseases, we intend to develop these programs internally through approval,
subject to partnership opportunities that arise, such as our collaboration with
Alnylam. For core programs targeting larger populations, we may seek development
partners, such as our collaboration with Roche on RG6346, under various economic
and operational arrangements. Together, our core program pipeline and our
pipeline of non-core collaborative programs constitute a broad and growing
therapeutic pipeline that we believe may result in multiple valuable approved
products based on our GalXC technology.
In addition to the programs listed in our pipeline, we are exploring a variety
of potential programs involving gene targets in the liver, central nervous
system ("CNS"), and other tissues, which we may elevate in the future to be
either a core program or a non-core collaborative program. Under our
collaborations with Novo, Roche, and Lilly, our collaborators have rights to
nominate additional programs for discovery by Dicerna and subsequent development
by the nominating collaborator, which will become part of our non-core pipeline.
Our four core programs are: nedosiran for the treatment of PH, RG6346 for the
treatment of HBV infection, DCR-A1AT and ALN-AAT02 for the treatment of A1AT
deficiency-associated liver disease, and a program for the treatment of a more
prevalent condition involving the liver.
We conduct clinical trials in various countries around the world, including the
U.S. and other areas heavily impacted by the COVID-19 pandemic. The current
supply of our investigational medicines is sufficient to support ongoing and
planned clinical trials. Based on current evaluations, our supply chains
continue to appear intact to meet the next 12 months of clinical, nonclinical,
and CMC supply demands across all programs. We have undertaken efforts to
mitigate potential future impacts to the supply chain by increasing our stock of
critical starting materials required to meet our needs and our collaborative
partners' needs through 2021 and by identifying and engaging alternative
suppliers. Additionally, there have been delays in the nedosiran pivotal PHYOX2
and DCR?-?A1AT Phase 1/2 studies. As a result, and based on the most recent
updates from clinical sites impacted by COVID-19 and
                                       30
--------------------------------------------------------------------------------
  Table of Contents
precautionary measures related to the pandemic, the Company regularly evaluates
its expectations related to clinical development milestones.
The tables below set forth the stage of development of our various GalXC RNAi
platform product candidates as of November 5, 2020:

[[Image Removed: drna-20200930_g1.jpg]]
Status of Dicerna Programs
Research
We continue to advance the GalXC RNAi platform as it is applied to therapeutic
targets expressed in hepatocytes using GalNAc conjugates for both our partnered
research and development programs and our internal liver-targeted programs. All
Dicerna-partnered programs include one or more liver-targeted applications of
the GalXC RNAi platform.
In addition, we continue to explore applications of our RNAi technology against
therapeutic gene targets expressed in tissues other than the liver, including
targets expressed in the CNS and other extrahepatic tissues and organs of high
therapeutic interest, and have made significant progress. Significant gene
target knockdown has been achieved in multiple cell types and regions of the CNS
and other extrahepatic tissues, in both rodents and nonhuman primates. These
extrahepatic applications are based on structurally and chemically modified
and/or alternatively conjugated forms of our RNAi platform as initially applied
to the liver in our ongoing clinical programs.
On August 6, 2020, we presented our first preclinical data demonstrating
expansion of our technology and discovery efforts beyond our hepatocyte-focused
GalXC RNAi technology to CNS, skeletal muscle, and adipose tissues. Results from
preclinical assays demonstrated consistent and durable CNS-wide target mRNA
knockdown using novel constructs regardless of route of administration
(intrathecal [IT] or intracisterna magna [ICM]), and reduction in target mRNA in
skeletal muscle and adipose tissue using optimized chemistries, resulting in
equivalent and potentially highly durable target knockdown regardless of dosing
regimens.
Our current core GalXC RNAi platform development programs are as follows:
Nedosiran for Primary Hyperoxaluria
We are developing our lead GalXC product candidate, nedosiran, which is in
pivotal clinical development, for the treatment of PH type 1 ("PH1"), PH type 2
("PH2"), and PH type 3 ("PH3"). PH is a family of severe, ultra-rare, genetic
liver disorders characterized by the overproduction of oxalate, a highly
insoluble metabolic end-product that is eliminated from the body mainly by the
kidneys. In patients with PH, the kidneys are unable to eliminate fully the
large amount of oxalate that is produced. The accumulation of oxalate
compromises the renal system, which may result in severe damage to the kidneys
and other organs.
                                       31
--------------------------------------------------------------------------------
  Table of Contents
PH encompasses three genetically distinct, autosomal-recessive, inborn errors of
glyoxalate metabolism characterized by the overproduction of oxalate. PH1, PH2,
and PH3 are each characterized by a specific enzyme deficiency. PH1 is caused by
a deficiency of glyoxalate aminotransferase, PH2 is caused by a deficiency of
glyoxalate reductase/hydroxypyruvate reductase, and PH3 is caused by a
deficiency of 4-hydroxy-2-oxoglutarate aldolase. Patients with PH are
predisposed to the development of recurrent urinary tract (urolithiasis) and
kidney (nephrolithiasis) stones, composed of calcium oxalate crystals. Stone
formation is accompanied by nephrocalcinosis in some patients with PH1 or PH2.
This deposition of calcium crystals in the renal parenchyma produces tubular
toxicity and renal damage that is compounded by the effects of renal
calculi-related obstruction and frequent superimposed infections. Based on the
evaluation of genome sequence databases, there may be as many as 16,000 people
with PH in the U.S. and major European countries.
The ultimate goal of any PH therapy is for patients to live normal lives without
the need to comply with hyperhydration and urine alkalization supporting
therapies in order to prevent the formation of new calcium oxalate crystals. To
best achieve this ultimate goal, Dicerna has developed a once-monthly fixed-dose
injection regimen of nedosiran. This once-monthly formulation of nedosiran is
designed to avoid the sudden oxalate spikes that could occur with less frequent
administration or missed dosages, which could result in the formation of kidney
stones and renal failure. To maximize patient convenience, we are developing
pre-filled syringes to enable self-administration by most PH patients without
the need for involvement of a healthcare provider for dosing.
In 2018, nedosiran received Orphan Drug Designation from the FDA, and the
European Medicines Agency's ("EMA") Committee for Orphan Medicinal Products
("COMP") designated nedosiran as an orphan medicinal product for the treatment
of PH. In June 2020, the FDA granted rare pediatric disease designation for
nedosiran. Under the FDA's rare pediatric disease designation program, the FDA
may grant a priority review voucher to a Sponsor who receives a product approval
for a rare pediatric disease. Subject to FDA approval of nedosiran for the
treatment of PH, we would be eligible to receive a voucher that may be redeemed
to receive priority review for a subsequent marketing application for a
different product candidate or which could be sold or transferred.
As of November 2019, we had completed all study participant dosing and follow-up
in PHYOX™1, a Phase 1 single-ascending-dose study of nedosiran in healthy
volunteers and study participants with PH1 or PH2. The primary objective of the
study was to evaluate the safety, tolerability, pharmacokinetics, and
pharmacodynamics of single-ascending doses of nedosiran. Secondary endpoints
included the change in 24-hour urinary oxalate excretion from baseline, defined
as the mean of two 24-hour collections during screening. The trial was divided
into two groups:
•Group A was a placebo-controlled, single-blind study and included 25 healthy
volunteers at a single site in the United Kingdom with five cohorts dosed at
0.3, 1.5, 3.0, 6.0, or 12.0 mg/kg of nedosiran or placebo (3:2 randomization).
•Group B was an open-label study and included 18 participants, 15 with PH1 and
three with PH2, and included three cohorts of participants dosed at 1.5, 3.0,
and 6.0 mg/kg of nedosiran and a fourth cohort with mixed dosing. Group B
participants were enrolled among three sites in the European Union, one in the
United Kingdom, and one in the United States.
Group A dosing was completed in March 2018, and Group B dosing was completed in
January 2019. We first reported interim results from the PHYOX1 trial on
September 5, 2018 and subsequently presented updated results at the American
Society of Nephrology's ("ASN") Kidney Week in San Diego on October 25, 2018, at
the German Society of Pediatric Nephrology 50th Annual Meeting in Cologne,
Germany on March 28, 2019, at the Oxalosis and Hyperoxaluria Foundation
International Hyperoxaluria Workshop in Boston, Massachusetts on June 20, 2019,
and at the ASN's Kidney Week Annual Meeting in Washington, D.C. on November 7,
2019.
With respect to efficacy data in PHYOX1, as of September 2019, nedosiran was
associated with normalization or near-normalization of urinary oxalate levels in
14 of 18 adult patients with PH1 or PH2 following single-dose administration.
The PHYOX1 investigators also reported that the three participants with PH2 (one
of whom received a single 1.5 mg/kg dose of nedosiran; the other two received a
3.0 mg/kg dose) achieved a mean maximal reduction of 24-hour urinary oxalate of
49% (range: 39% to 66%) with one participant reaching normalization and another
participant reaching near-normalization at one or more post-dose time points.
The PH2 patient who did not achieve normalization or near-normalization was
dosed at the 1.5 mg/kg level.
Data from the PHYOX1 trial showed that nedosiran was associated with
normalization or near-normalization of urinary oxalate levels in 14 of 18 adult
participants with PH1 or PH2 following single-dose administration. Nedosiran was
generally well tolerated based on data from 18 participants (15 adults and three
adolescents [participants 13-16 years old]) with PH1 (n=15) or PH2 (n=3) and 25
adult healthy volunteers. As of the last data cut in March 2020, seven serious
adverse events ("SAEs") had occurred in six participants in Group B; none was
deemed related to the study drug, and all seven SAEs had resolved. A total of
seven participants dosed with nedosiran experienced mild or moderate
injection-site reactions (defined as occurring four hours or more after
injection), all of which resolved without intervention in a mean of 25 hours. No
clinically meaningful safety signals were observed, including no clinically
significant liver function test abnormalities.
                                       32
--------------------------------------------------------------------------------
  Table of Contents
PHYOX3 Long-Term, Multidose, Open-Label Extension Study
Following positive Phase 1 data from PHYOX1 in 2019, a single-ascending-dose
study of nedosiran in healthy volunteers and study participants with PH1 or PH2,
we received approval to proceed with the Phase 2 ("PHYOX2," our multidose,
double-blind, randomized, placebo-controlled pivotal trial of nedosiran) and
Phase 3 ("PHYOX3") studies of nedosiran in PH. We initiated dosing of
participants transitioning from the PHYOX1 trial into the PHYOX3 study, a
long-term, multi-dose, open-label extension study. Unlike the PHYOX2 trial,
which requires screening and enrollment of new participants, patients are
permitted to transition into the PHYOX3 trial from any previous nedosiran trial
in which they have participated. As of April 2020, we had implemented a protocol
amendment with local institutional review boards ("IRBs") and had transitioned
certain site visits to a combination of at-home nurse visits with investigator
telehealth assessments for dose administration and safety follow-up, as a result
of impacts from COVID-19.
The primary endpoint of PHYOX3 is to evaluate the impact of monthly nedosiran
administration on the annual rate of decline in estimated glomerular filtration
rate ("eGFR"), a measure of kidney function. The PHYOX3 trial will also evaluate
the long-term effect of nedosiran on urinary oxalate ("Uox") excretion, new
stone formation, progression of nephrocalcinosis, and the potential to enable
the gradual decrease or elimination of patients' supportive hyperhydration
therapies.
In August 2020, we presented interim data from the PHYOX3 trial up to July 10,
2020. Included in the analysis were 11 patients who had received at least five
monthly doses of nedosiran delivered subcutaneously. There was an extended
washout period between participants' completion of PHYOX1 during which
participants were anticipating a return to 80% of Uox PHYOX1 baseline prior to
starting in PHYOX3. Some patients did not return to this range and were
permitted to enroll in PHYOX3 with lower baseline levels. The mean body surface
area ("BSA") adjusted baseline Uox of these 11 participants in PHYOX1 was 1.323
mmol/1.73m2 BSA/24 hr, and the mean BSA-adjusted observable baseline Uox level
for these same participants initiating PHYOX3 and included in this analysis was
0.926 mmol/1.73m2 BSA/24 hr. One hundred percent of PH1 participants (eight of
eight) that had rolled over from PHYOX1 and received five doses of nedosiran in
the PHYOX3 study had normalization or near normalization at Day 120: of these,
near-normalization was reached in 63% of the PH1 participants (five of eight).
For PH1 participants, the mean Uox level achieved was within the normal range
(mean Uox - 0.404 mmol/1.73m2 BSA/24 hr). Thirty-three percent of PH2
participants (one of three) achieved normalization at Day 120. We define normal
and near-normal Uox as below 0.46 mmol/1.73m2 BSA/24 hr and from 0.46 to 0.6
mmol/1.73m2 BSA/24 hr, respectively. As of July 10, 2020, five of the 17
participants enrolled in PHYOX3 had achieved and maintained normal Uox
concentrations on at least three consecutive visits, making them eligible for
gradual reduction in fluid intake.
In October 2020, we presented new interim data from the PHYOX3 trial. A total of
16 trial participants from the completed PHYOX1 trial had entered the PHYOX3
trial. Of these, 13 who had reached 180 Days and had received six monthly doses
in the PHYOX3 trial were included in this analysis. Three participants were not
included in the efficacy analysis, as they had not yet reached Day 180 at the
time of the analysis. All 13 participants (10 with PH1 and three with PH2)
receiving nedosiran achieved normal (12 of 13) or near-normal (one of 13) Uox
excretions at one or more timepoints. Of these, all 10 (100%) of the
participants with PH1, and two of the three (67%) participants with PH2,
achieved normal Uox excretions at one or more visits.
In this interim analysis, nedosiran was generally well tolerated, and no serious
safety concerns were identified in this study. There were no treatment
discontinuations or study withdrawals during the observation period. Two
participants had serious treatment-emergent adverse events ("TEAEs")
(pyelonephritis and nephrolithiasis) that were determined by the investigator to
be unrelated to nedosiran treatment. The most common TEAEs were mild to moderate
administration-site reactions.
PHYOX2 Multidose, Double-Blind, Randomized, Placebo-Controlled Pivotal Trial
In March 2020, we provided an overview of our response to the COVID-19 pandemic
that included an update on business continuity and clinical development
milestones, including milestones related to the PHYOX clinical development
program for nedosiran. The update outlined Dicerna's intention to work closely
with local IRBs to implement a protocol amendment in accordance with recently
adopted regulatory guidance that would allow for the transition of remaining
site visits to at-home nurse visits for drug administration and safety
follow-up. As a result of enrollment progress and temporary suspension of
further clinical trial activities at multiple sites as of the March update, the
Company withdrew its previous guidance for completion of enrollment in PHYOX2 in
the second quarter of 2020. During April and May 2020, Dicerna transitioned
certain site visits in the PHYOX2 trial to a combination of at-home nurse visits
and investigator telehealth assessments for drug administration and safety
follow-up following trial sites' operational changes resulting from the COVID-19
pandemic. Enrollment in the PHYOX2 trial continues at a number of sites
globally. Given the fluid nature of the COVID-19 pandemic and the evolving and
extraordinary actions undertaken by clinical trial sites globally, we continue
to evaluate our clinical plan related to nedosiran, and, at this time, expect to
complete PHYOX2 enrollment in the fourth quarter of 2020. We also anticipate the
last patient to complete this study in the first half of 2021 and to submit a
New Drug Application ("NDA") in the third quarter of 2021.
In addition to the PHYOX2 clinical trial, we have multiple additional trials
planned. Patient enrollment in a study of patients with PH3 (PHYOX4) is expected
in the fourth quarter of 2020. Enrollment in a study of PH1 and PH2 patients
with severe renal impairment, including those on dialysis (PHYOX7), and in a
study of PH1 and PH2 patients aged 0-5 years with relatively intact renal
                                       33
--------------------------------------------------------------------------------
  Table of Contents
function (PHYOX8) is expected to begin in the first quarter of 2021. In
addition, we initiated an observational study in the third quarter of 2020 in
patients with PH3 (PHYOX-OBX) to evaluate the association between urinary
oxalate and the rate of kidney stone formation.
In discussions with the FDA, we received feedback indicating alignment on a path
to the full approval of nedosiran for the treatment of PH1 and PH2 based on
achievement of substantial reduction of urinary oxalate excretion in patients
with PH1 and PH2 after nedosiran administration versus those to whom the placebo
was administered in the PHYOX2 pivotal trial. With this feedback, we believe we
have a path to seek full approval in both PH1 and PH2 based on PHYOX2 results.
In July 2019, we received Breakthrough Therapy Designation from the FDA for the
development of nedosiran for the treatment of PH1. In June 2020, we announced
that the FDA granted rare pediatric disease designation for nedosiran for the
treatment of PH. We plan to continue our dialogue with the FDA regarding
endpoints for studies involving patients with PH3 as part of the PHYOX clinical
development program for nedosiran, and potentially an expansion of the
Breakthrough Therapy Designation to include PH2.
In order to enhance our ability to bring nedosiran to market, in April 2020, we
entered into the Alnylam Cross-License Agreement for the intellectual property
for ours and Alnylam's respective investigational programs for the treatment of
PH. The non-exclusive cross-license agreement ensures that each party has the
freedom to develop and commercialize its respective product candidates. The
cross-license agreement provides for Alnylam to pay mid- to high-single-digit
royalties to Dicerna based on global net sales of lumasiran and for Dicerna to
pay low-single-digit royalties to Alnylam on global net sales of nedosiran.
Commercial readiness activities continue across the organization to ensure the
timing of appropriate infrastructure, processes, and capabilities to support
Dicerna's evolution to a fully integrated biopharmaceutical company. The primary
focus is on U.S. commercialization infrastructure for nedosiran and the Medical
Affairs and Commercial teams that have been established. Additional
infrastructure and commercialization activities are paced to the PHYOX programs
and NDA preparations. Outside the U.S., active discussions for regional and/or
multinational commercial collaboration partners are underway.
RG6346 for Chronic Hepatitis B Virus Infection
Our GalXC RNAi platform-based product candidate for the treatment of chronic HBV
infection, RG6346, is currently being tested in a Phase 1 clinical trial. Roche
will be responsible for initiating Phase 2 development of RG6346. HBV is the
world's most common serious liver infection and affects an estimated 292 million
people worldwide. Chronic HBV infection is characterized by the presence of the
HBV surface antigen ("HBsAg") for six months or more. In order to be optimally
positioned to develop and commercialize our product candidate, RG6346, in
combination with other novel drugs, we entered into a research collaboration and
licensing agreement with Roche in October 2019. Under the terms of the
agreement, we are leading the development of RG6346 through the current Phase 1
trial, and pending favorable results, Roche intends to further develop RG6346
with the overall goal of developing a combination regimen to achieve a long-term
functional cure of chronic HBV in combination with additional Roche product
candidates. The agreement also provides an option for the companies to
collaborate in the discovery, development, and commercialization of
oligonucleotide therapeutics intended for the treatment of chronic HBV.
The DCR-HBVS-101 clinical trial is a Phase 1, randomized, placebo-controlled,
double-blind study designed to evaluate the safety and tolerability of RG6346 in
healthy volunteers and in patients with chronic HBV. Secondary objectives are to
characterize the pharmacokinetic profile of RG6346 and to evaluate preliminary
pharmacodynamic effects on markers of HBV antiviral efficacy, including
reductions of HBsAg and HBV deoxyribonucleic acid ("DNA") levels in blood. The
Phase 1 clinical trial is divided into three groups:
•Group A is a single-ascending-dose arm in which 30 healthy volunteers received
a dose of RG6346 (0.1, 1.5, 3.0, 6.0, or 12.0 mg/kg) or placebo, with a
four-week follow-up period. Group A dosing was completed in August 2019.
•Group B is a single-dose arm in which eight participants with chronic HBV who
are naïve to nucleoside analog therapy received a 3.0 mg/kg dose of RG6346 or
placebo, with a follow-up period of at least 12 weeks. We initiated Group B
dosing in the third quarter of 2019, in parallel with Group C at the 3.0 mg/kg
dose level. Group B dosing was completed in May 2020.
•Group C is a multiple-ascending-dose arm in which RG6346 (1.5, 3.0, or 6.0
mg/kg) or placebo was administered to 18 participants with chronic HBV who are
already being treated with nucleoside analogs, with a treatment and follow-up
period of 16 weeks or more.
In August 2020, we announced interim top-line data from all originally planned
cohorts from the ongoing DCR-HBVS-101 study. Patients within Groups B and C were
randomized 5:3 and 2:1, to RG6346 treatment versus placebo, respectively.
Participants in Groups B and C were eligible to enter into an extended follow-up
observation period if reductions of ?1.0 log10 IU/mL of HBsAg from baseline were
maintained at the end of the double-blind treatment period (12 weeks/85 days for
Group B and 16 weeks/112 days for Group C). Enrollment for the originally
planned cohorts was completed in June 2020, and results from an interim analysis
of data
                                       34
--------------------------------------------------------------------------------
  Table of Contents
available as of June 25, 2020 were presented publicly during our R&D Day event
in August 2020 and are summarized below. As of the interim analysis, two
participants in the Group C 6.0 mg/kg group had not yet reached Day 112. The
last participant dose in the 6.0 mg/kg dose group was administered in September
2020, and the follow-up phase of the study is ongoing as of November 5, 2020.
•Data from the June 25, 2020 interim analysis showed that nine of 10 patients
who received RG6346 and completed the treatment period in Group C achieved ?1.0
Log10 IU/mL reduction in HBsAg at Day 112 and continued in the extended
follow-up period.
•At Day 112, the mean reduction in HBsAg from baseline for each Group C cohort
was:
•1.39 log10 IU/mL (n=4) for the 1.5 mg/kg cohort
•1.80 log10 IU/mL (n=4) for the 3.0 mg/kg cohort
•1.84 log10 IU/mL (n=2) for the 6.0 mg/kg cohort; two participants had not yet
reached Day 112.
•Six of the 10 participants who completed Day 112 had HBsAg <100 IU/mL.
•Of the data presented, durability of response was demonstrated up to Day 392,
and the first patient dosed in the study had reached Day 392 during the
conditional follow-up period with a 2.21 log10 IU/mL reduction in HBsAg (<100
IU/mL).
In addition, after a single 3.0 mg/kg dose of RG6346, two participants in Group
B (naïve to nucleoside analog therapy) experienced protocol-defined transient
alanine aminotransferase ("ALT") flares, and one patient experienced a
near-flare. In each case, synthetic and excretory liver function was preserved
and there was a coinciding drop in blood HBsAg, suggesting a potential
beneficial flare and immune response in relation to the observed ALT elevations.
A similar self-resolving flare and response was demonstrated in one Group C
participant receiving 6.0 mg/kg of RG6346.
No SAEs were observed with RG6346 treatment in any group. The most commonly
reported adverse events were mild or moderate injection-site events. No
dose-limiting toxicities were observed, and there were no safety-related
discontinuations. No dose-/exposure-dependent increases in frequency or severity
of safety parameters were noted, and participants with ALT/aspartate
aminotransferase ("AST") or gamma-glutamyl transferase ("GGT") elevations were
all self-resolving and associated with preserved liver synthetic and excretory
function. All observed ALT elevations above the upper limit of normal were
self-resolving and associated with preserved liver synthetic and excretory
function. Dose-/exposure-dependent increases in frequency or severity of any
other safety parameters were not noted.
In agreement with Roche, we are enrolling two additional optional open-label,
fixed dose Group C cohorts, Cohort 4C and Cohort 5C. Cohorts 4C and 5C will
evaluate fixed dosing regimens over an extended conditional follow-up period to
capture the rebound kinetics of HBsAG from baseline levels. Cohort 4C is a
single-ascending-dose arm with a total follow-up duration of up to 48 weeks.
Cohort 5C is a multiple dose cohort with a total follow-up duration of up to 72
weeks.
In April 2020, Roche nominated the first of up to five targets under the
research and development portion of our collaboration agreement.
DCR-A1AT and ALN-AAT02 for Alpha-1 Antitrypsin Deficiency-Associated Liver
Disease
Our GalXC RNAi platform-based product candidate for the treatment of A1AT
deficiency-associated liver disease, DCR-A1AT, is currently being tested in a
Phase 1/2 clinical study, which we believe could enable a pivotal trial without
additional studies. A1AT deficiency is an inherited disorder that can lead to
liver disease in children and adults and lung disease in adults. The disorder is
caused by mutations in a gene called SERPINA1. This gene, when functioning
normally, provides instructions for making the A1AT protein, which protects the
body from an enzyme called neutrophil elastase. This enzyme is released from
white blood cells to fight infection, but it can attack normal tissues if not
tightly controlled by A1AT. Mutations in the SERPINA1 gene can result in a
deficiency of A1AT or, most commonly, an abnormal form of the protein that
cannot control neutrophil elastase. Accumulation of abnormal A1AT protein in the
liver can lead to liver disease. Uncontrolled neutrophil elastase can also
destroy alveoli (small air sacs in the lungs) and cause lung disease.
A1AT deficiency occurs all over the world, though its prevalence varies by
population. The disorder affects roughly one in 1,500 to 3,500 individuals with
European ancestry but is uncommon in people of Asian descent. Congenital A1AT
deficiency is estimated to affect 2.4 people out of every 10,000 in the EU.
In March 2020, the FDA granted orphan drug designation to DCR-A1AT for the
treatment of A1AT deficiency. In December 2019, the European Commission granted
orphan drug designation to DCR-A1AT for the treatment of congenital A1AT
deficiency based on a positive opinion from the COMP of the EMA. We submitted a
CTA to the Swedish Medical Products Agency in June 2019 to conduct a
first-in-human Phase 1/2 study of DCR-A1AT.
                                       35
--------------------------------------------------------------------------------
  Table of Contents
In April 2020, we entered into the Alnylam Collaboration Agreement. Under the
Alnylam Collaboration Agreement, Alnylam's ALN-AAT02 and Dicerna's DCR-A1AT,
each in Phase 1/2 development, will be explored for the treatment of alpha-1
liver disease. Under the agreement, we assume responsibility for both ALN-AAT02
and DCR-A1AT at our cost and may progress one or both of these investigational
medicines through clinical development. We will select which product candidate
to advance in development for the treatment of patients with alpha-1 liver
disease. At the completion of Phase 3, Alnylam has the no-cost opportunity to
opt in to commercialize the selected candidate in countries outside the U.S.
where it already has a commercialization infrastructure in place. If Alnylam
exercises its opt-in right, each party will pay tiered royalties to the other
party based on net product sales generated in its territory at rates dependent
on which candidate is commercialized. In the event Alnylam waives its
commercialization option, we will retain worldwide rights to commercialize the
selected candidate in exchange for milestones and royalties payable to Alnylam,
also at a rate dependent on which candidate is ultimately commercialized.
The initial DCR-A1AT-101 clinical trial is a Phase 1/2 randomized,
placebo-controlled study designed to evaluate the safety and tolerability of
DCR-A1AT in healthy volunteers and in patients with A1AT deficiency-associated
liver disease. Secondary objectives are to characterize the pharmacokinetic
profile of DCR-A1AT, to evaluate preliminary pharmacodynamics on serum A1AT
protein concentrations, and to characterize the effect of DCR-A1AT on A1AT
deficiency-associated liver disease evaluated by liver biopsy. Exploratory
objectives include characterization of the effect of DCR-A1AT on A1AT
deficiency-associated liver disease evaluated by biochemical markers as well as
the effect on liver stiffness.
Following our business update in March 2020, further enrollment of healthy
volunteers in the Phase 1/2 trial of DCR-A1AT was effectively paused due to site
restrictions related to the COVID-19 pandemic; however, all subjects in the
current dosing cohort have since completed their remaining visits. Additional
safety precautions have been implemented, including testing of any participants
who present with symptoms consistent with COVID-19. As of late April 2020, the
Scientific Review Committee for the DCR-A1AT Phase 1/2 trial confirmed that the
study could continue, and we began enrolling the next dosing cohort in May 2020,
which has since been completed.
We are currently targeting program selection for advancement into Phase 2 in the
first quarter of 2021.
DCR-Proprietary
We are currently pursuing development of a proprietary candidate for the
treatment of a more prevalent condition involving the liver. Our goal is to
submit an Investigational New Drug ("IND") or Clinical Trial Application ("CTA")
filing in mid-2021.
Collaborative Program Update
Eli Lilly and Company
During the first quarter of 2020, Lilly selected LY3819469, a GalXC molecule for
the second collaboration target in cardiometabolic disease, for advancement into
preclinical development.
Lilly currently has a goal of filing an IND or CTA for LY3561774 in late 2020.
Critical Accounting Policies and Significant Judgments and Estimates
Our discussion and analysis of our financial condition and results of operations
is based on our condensed consolidated financial statements, which have been
prepared in accordance with accounting principles generally accepted in the U.S.
The preparation of our condensed consolidated financial statements requires us
to make estimates and apply judgments that affect the reported amounts of assets
and liabilities and the disclosure of contingent assets and liabilities at the
date of the condensed consolidated financial statements, as well as the revenue
and expenses incurred during the reported periods. We base our estimates on
historical experience and on various other factors that we believe are
reasonable under the circumstances. Actual results may differ from these
estimates and could have a material impact on our condensed consolidated
financial statements.
The critical accounting policies that we believe impact significant judgments
and estimates used in the preparation of our financial statements presented in
this report are described in "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 filed
with the SEC on February 28, 2020. There have been no significant changes to our
critical accounting policies as disclosed in our most recently filed Annual
Report on Form 10-K during the nine months ended September 30, 2020.
                                       36
--------------------------------------------------------------------------------
  Table of Contents
Recent Accounting Pronouncements
A summary of significant recent accounting pronouncements that we have adopted
or expect to adopt is included in Note 1 - Description of Business and Basis of
Presentation to our condensed consolidated financial statements (see Part I,
Item 1 - "Financial Statements" of this Quarterly Report on Form 10-Q).
Financial Operations Overview
Revenue
Our revenue to date has been generated primarily through research funding,
license fees and other upfront payments, option exercise fees, milestone
payments, and preclinical development payments under our research collaboration
arrangements with Novo, Roche, Lilly, Alexion, and BI. We have not generated any
commercial product revenue, nor do we expect to generate any product revenue in
the near-term.
In the future, we may generate revenue from a combination of research and
development payments, license fees and other upfront payments, milestone
payments, product sales, and royalties in connection with our current or future
collaborations with partners, and product sales from our internally developed
products. We expect that any revenue we generate will fluctuate in future
periods as a result of the timing of our or our collaborators' achievement of
preclinical, clinical, regulatory, and commercialization milestones, to the
extent achieved, the timing and amount of any payments to us related to such
milestones, and the extent to which any of our product candidates are approved
and successfully commercialized by us or a collaborator. Delays in or changes to
the research and development plans and timelines related to our collaboration
agreements are likely due to the COVID-19 pandemic. Because we recognize the
majority of our collaboration revenue on a cost-to-cost measure of progress,
revenues recognized in the near-term future may be lower than originally
anticipated and could be recognized over an extended period of time as a result.
Research and development expenses
Research and development expenses consist of costs associated with our research
activities, including discovery and development of our molecules and drug
delivery technologies, clinical and preclinical development activities, and
research activities under our research collaboration and license agreements. Our
research and development expenses include:
•direct research and development expenses incurred under arrangements with third
parties, such as contract research organizations, contract manufacturing
organizations, and consultants;
•platform-related lab expenses, including discovery research, lab supplies,
license fees, and consultants;
•employee-related expenses, including salaries, benefits, and stock-based
compensation expense; and
•facilities, depreciation, and other allocated expenses, which include direct
and allocated expenses for rent and maintenance of facilities, depreciation of
leasehold improvements and equipment, and laboratory and other supplies.
We expense research and development costs as they are incurred. We account for
non-refundable advance payments for goods and services that will be used in
future research and development activities as expenses when the service has been
performed or when the goods have been received. A significant portion of our
research and development costs are not tracked by project, as they benefit
multiple projects or our technology platform.
Delays in or changes to our research and development plans and timelines, which
impact both our internal and external resources, have occurred and may continue
to occur due to the COVID-19 pandemic. Internally, we have been impacted by
mandatory work from home edicts directed by the local governments in the
jurisdictions in which we operate. However, essential work exemptions continue
to permit critical research and development and laboratory activities for
limited personnel. Those exemptions enable some continued discovery research and
activities supporting our collaborative agreements and our own programs. We also
anticipate that the timing of hiring additional personnel may shift into later
periods than initially anticipated. Externally, a number of our clinical trial
sites have delayed and may continue to delay trial-related activities as a
result of COVID-19. Any of these factors could cause the timing of the research
and development expenses we expect to incur to shift into later periods and have
the potential to cause us to expend more funds than originally contemplated as a
result of needing to extend clinical development activities.
General and administrative expenses
General and administrative expenses consist primarily of salaries and related
benefits, including stock-based compensation, related to our executive, finance,
legal, business development, commercial, and support functions. Other general
and administrative expenses include travel expenses, professional legal fees,
audit, tax, and other professional services, and allocated facility-related
costs not otherwise included in research and development expenses.
                                       37
--------------------------------------------------------------------------------
  Table of Contents
Delays in or changes to our research and development plans and timelines due to
the COVID-19 pandemic may also impact our support functions and the timing of
hiring additional personnel, which could cause the timing of certain general and
administrative expenses we expect to incur to shift into later periods.
Other income (expense)
Other income (expense) consists primarily of interest income. Interest income
consists of income earned on our cash and cash equivalents, held-to-maturity
investments, and restricted cash equivalents. We expect that interest income
will continue to decrease due to recent decreases in interest rates.
Results of Operations
Comparison of the Three and Nine Months Ended September 30, 2020 and 2019
The following table summarizes the results of our operations for the periods
indicated (amounts in thousands, except percentages):
                                     THREE MONTHS ENDED
                                       SEPTEMBER 30,
                                    2020           2019         $ CHANGE      % CHANGE
Revenue                          $  48,875      $   8,035      $ 40,840               *
Operating expenses:
Research and development            54,814         30,086        24,728         82.2  %
General and administrative          16,961         10,619         6,342         59.7  %
Total operating expenses            71,775         40,705        31,070         76.3  %
Loss from operations               (22,900)       (32,670)        9,770        (29.9) %
Other income (expense):
Interest income (expense), net       1,050          1,880          (830)       (44.1) %
Other income                             1              -             1        100.0  %
Total other income, net              1,051          1,880          (829)       (44.1) %
Net loss                         $ (21,849)     $ (30,790)     $  8,941        (29.0) %

* Percentage change not meaningful


                                     NINE MONTHS ENDED
                                       SEPTEMBER 30,
                                    2020           2019         $ CHANGE       % CHANGE
Revenue                          $ 123,351      $  16,824      $ 106,527               *
Operating expenses:
Research and development           151,361         74,521         76,840        103.1  %
General and administrative          53,549         29,126         24,423         83.9  %
Total operating expenses           204,910        103,647        101,263         97.7  %
Loss from operations               (81,559)       (86,823)         5,264         (6.1) %
Other income (expense):
Interest income (expense), net       5,382          6,034           (652)       (10.8) %
Other income                            16              -             16        100.0  %
Total other income, net              5,398          6,034           (636)       (10.5) %
Net loss                         $ (76,161)     $ (80,789)     $   4,628         (5.7) %

* Percentage change not meaningful


                                       38
--------------------------------------------------------------------------------
  Table of Contents
Revenue
The following tables provide a summary of revenue recognized (amounts in
thousands):
              THREE MONTHS ENDED
                SEPTEMBER 30,
              2020           2019        $ CHANGE      % CHANGE
Novo      $     3,552      $     -      $  3,552        100.0  %
Roche          18,686            -        18,686        100.0  %
Lilly          12,073        5,274         6,799        128.9  %
Alexion        13,639        1,451        12,188               *
BI                925        1,310          (385)       (29.4) %

Total     $    48,875      $ 8,035      $ 40,840               *

* Percentage change not meaningful


              NINE MONTHS ENDED
                SEPTEMBER 30,
             2020           2019        $ CHANGE       % CHANGE
Novo      $   7,604      $      -      $   7,604        100.0  %
Roche        58,670             -         58,670        100.0  %
Lilly        31,142         8,346         22,796        273.1  %
Alexion      23,464         2,954         20,510               *
BI            2,471         5,524         (3,053)       (55.3) %

Total     $ 123,351      $ 16,824      $ 106,527               *


* Percentage change not meaningful
The increase in revenue for the three and nine months ended September 30, 2020
is primarily attributable to increased activities and associated costs under the
recent collaboration agreement with Roche, as well as under the Alexion and
Lilly collaboration agreements, as all three agreements are recognized as
revenue on a cost-to-cost measure of progress method.
Research and development expenses
The following table summarizes our research and development expenses incurred
during the periods indicated (amounts in thousands):
                                                       THREE MONTHS ENDED
                                                          SEPTEMBER 30,
                                                     2020               2019            $ CHANGE             % CHANGE

Direct research and development expenses $ 29,567 $ 17,116 $ 12,451

                    72.7  %
Platform-related and discovery research expenses      3,843             4,319              (476)                  (11.0) %
Employee-related expenses                            18,483             7,797            10,686                   137.1  %
Facilities, depreciation, and other expenses          2,921               854             2,067                   242.0  %
Total                                            $   54,814          $ 30,086          $ 24,728                    82.2  %



                                                       NINE MONTHS ENDED
                                                         SEPTEMBER 30,
                                                     2020              2019            $ CHANGE             % CHANGE

Direct research and development expenses $ 84,757 $ 40,628 $ 44,129

                   108.6  %
Platform-related and discovery research expenses    10,488             9,782               706                     7.2  %
Employee-related expenses                           47,359            21,399            25,960                   121.3  %
Facilities, depreciation, and other expenses         8,757             2,712             6,045                   222.9  %
Total                                            $ 151,361          $ 74,521          $ 76,840                   103.1  %


Research and development expenses increased for the three months ended
September 30, 2020 compared to the three months ended September 30, 2019
primarily due to direct research and development expenses and employee-related
expenses. The $12.5 million increase in direct research and development expenses
in the three months ended September 30, 2020 included a
                                       39
--------------------------------------------------------------------------------
  Table of Contents
$3.8 million increase in clinical study costs, reflecting increased activities
primarily associated with our nedosiran and DCR-A1AT programs, as well as
increased costs associated with our collaboration with Lilly. In addition,
manufacturing costs increased $3.7 million. Research and development expenses
were also impacted by a $10.7 million increase in employee-related expenses,
which include salaries, benefits, and stock-based compensation. The increase in
employee-related expenses is a result of an increase in research and development
headcount necessary to support our collaboration agreements and expanding
pipeline.
Research and development expenses increased for the nine months ended
September 30, 2020 compared to the nine months ended September 30, 2019
primarily due to direct research and development expenses and employee-related
expenses. The $44.1 million increase in direct research and development expenses
in the nine months ended September 30, 2020 included a $19.3 million increase in
manufacturing costs primarily for drug substance to support our clinical studies
and a $10.4 million increase in clinical study costs, reflecting increased
activities primarily associated with our DCR-A1AT and nedosiran programs, as
well as increased costs associated with our collaboration with Lilly. In
addition, research-related expenses, such as lab supplies and materials,
increased $5.3 million. Research and development expenses were also impacted by
a $26.0 million increase in employee-related expenses, which include salaries,
benefits, and stock-based compensation. The increase in employee-related
expenses is a result of an increase in research and development headcount
necessary to support our collaboration agreements and expanding pipeline.
We expect our overall research and development expenses to continue to increase
for the foreseeable future as we ramp our clinical manufacturing activities,
continue clinical activities associated with our core product candidates, and
continue activities under our existing collaboration agreements.
General and administrative expenses
General and administrative expenses increased for the three months ended
September 30, 2020 compared to the three months ended September 30, 2019. The
$6.3 million increase for the three months ended September 30, 2020 is primarily
due to a $4.6 million increase in employee-related compensation, including
salaries, benefits, and stock-based compensation, due to an increase in
headcount necessary to support our growing operations. In addition, professional
consulting services increased $1.0 million in the three months ended
September 30, 2020.
General and administrative expenses increased for the nine months ended
September 30, 2020 compared to the nine months ended September 30, 2019. The
$24.4 million increase for the nine months ended September 30, 2020 is primarily
due to a $16.3 million increase in employee-related compensation, including
salaries, benefits, and stock-based compensation, due to an increase in
headcount necessary to support our growing operations. In addition, professional
consulting services increased $5.9 million in the nine months ended
September 30, 2020.
We expect general and administrative expenses to continue to increase in the
foreseeable future, largely due to investments in staffing and market readiness
activities.
Liquidity and Capital Resources
We have historically funded our operations primarily through the public offering
and private placement of our securities and consideration received from our
collaborative agreements. As of September 30, 2020, we had cash and cash
equivalents and held-to-maturity investments of $609.9 million compared to
$348.9 million as of December 31, 2019.
During the nine months ended September 30, 2020, we realized $375.0 million in
proceeds from the Novo and Roche collaborations and net proceeds of $39.2
million from the issuance of common stock to a single institutional investor
pursuant to our common stock Sales Agreement with Cowen and Company, LLC as the
sales agent. The shares in the offering were sold pursuant to a
shelf registration statement declared effective by the SEC on May 31, 2018 and
a prospectus supplement filed with the SEC on June 1, 2018.
We believe that our cash, cash equivalents, and held-to-maturity investments,
together with anticipated milestone and other payments from existing
collaborations, provide us with sufficient resources to continue our planned
operations and clinical activities into 2023.
                                       40
--------------------------------------------------------------------------------
  Table of Contents
Cash flows
The following table shows a summary of our condensed consolidated cash flows for
the periods indicated (amounts in thousands):
                                                  NINE MONTHS ENDED
                                                    SEPTEMBER 30,
                                                 2020           2019

Net cash provided by operating activities $ 222,415 $ 14,232 Net cash used in investing activities $ (216,882) $ (19,233) Net cash provided by financing activities $ 48,400 $ 792




Operating activities
Net cash provided by operating activities increased $208.2 million in the nine
months ended September 30, 2020 compared to the nine months ended September 30,
2019, primarily due to a $98.3 million decrease in contract receivables largely
due to the upfront cash payment received from Roche in January 2020 in
connection with our collaboration agreement and a $79.1 million increase in
deferred revenue from the Novo and Roche collaborations.
Investing activities
Net cash used in investing activities increased $197.6 million in the nine
months ended September 30, 2020 compared to the nine months ended September 30,
2019. The increase in net cash used in investing activities primarily relates to
a $309.5 million increase in purchases of held-to-maturity investments for the
nine months ended September 30, 2020 as a result of an increased cash balance
from the Novo and Roche upfront payments, which were partially offset by a
$114.0 million increase in maturities of investments.
Financing activities
Net cash provided by financing activities for the nine months ended
September 30, 2020 increased $47.6 million compared to the nine months ended
September 30, 2019. The increase was primarily due to the receipt of
$39.2 million in net proceeds in February 2020 from the private placement of our
common stock.
Funding requirements
We expect that our primary uses of capital will continue to be commercialization
readiness and launch activities, subject to approval of our development
candidates; third-party clinical research and development services and
manufacturing costs; compensation and related expenses; laboratory and related
supplies; legal and other regulatory expenses; and general overhead costs.
Because of the numerous risks and uncertainties associated with the development
and commercialization of our product candidates and the extent to which we may
enter into additional collaborations with third parties to participate in their
development and commercialization, we are unable to estimate the amounts of
capital outlays and operating expenditures associated with our anticipated
development activities. However, based on our current operating plan, we believe
that our available cash, cash equivalents, held-to-maturity investments, and
anticipated milestone and other payments from existing collaborations will be
sufficient to fund the execution of our current clinical and operating plans
into 2023. Over the balance of 2020 and the year ending December 31, 2021, we
forecast receiving over $100.0 million in cash from our collaborations,
including anticipated milestone achievement. We based this estimate on
assumptions that may prove to be incorrect, and we could utilize our available
capital resources sooner than we currently expect.
Our forecast of the period of time through which our financial resources will be
adequate to support our operations is a forward-looking statement that involves
risks and uncertainties, and actual results could vary materially as a result of
a number of factors. Our future capital requirements are difficult to forecast
and will depend on many factors, including:
•the potential receipt of any milestone payments under the Novo Collaboration
Agreement, Roche Collaboration Agreement, Lilly Collaboration Agreement, Alexion
Collaboration Agreement, BI Agreements, and Alnylam Collaboration Agreement and
the potential payment of any royalties under the Alnylam Collaboration
Agreement;
•the potential payment or receipt of royalty payments under the Alnylam
Cross-License Agreement;
•the terms and timing of any other collaboration, licensing, and other
arrangements that we may establish;
•the initiation, progress, timing, and completion of preclinical studies and
clinical trials for our current and future potential product candidates,
including the impact of COVID-19 on our ongoing and planned research and
development efforts and the timing of a potential commercial launch date for
nedosiran;
                                       41
--------------------------------------------------------------------------------
  Table of Contents
•our alignment with the FDA on regulatory approval requirements;
•the impact of COVID-19 on the operations of key governmental agencies, such as
the FDA, which may delay the development of our current product candidates or
any future product candidates;
•the number and characteristics of product candidates that we pursue;
•the outcome, timing, and cost of regulatory approvals;
•delays that may be caused by changing regulatory requirements;
•the cost and timing of hiring new employees to support our continued growth;
•the costs involved in filing and prosecuting patent applications and enforcing
and defending patent claims;
•the costs of filing and prosecuting intellectual property rights and enforcing
and defending any intellectual property-related claims;
•the costs of responding to and defending ourselves against complaints and
potential litigation;
•the costs and timing of procuring clinical and commercial supplies for our
product candidates;
•the extent to which we acquire or in-license other product candidates and
technologies; and
•the extent to which we acquire or invest in other businesses, product
candidates, or technologies.
Until such time, if ever, that we generate product revenue, we expect to finance
our future cash needs through a combination of public or private equity
offerings, debt financings, and research collaboration and license agreements.
Please see the risk factors set forth in Part II, Item 1A - "Risk Factors" in
this Quarterly Report on Form 10-Q for additional risks associated with our
substantial capital requirements.
Contractual Obligations and Commitments
We have excluded from our condensed consolidated balance sheets $69.2 million of
fixed payments for leases that have not yet commenced for accounting purposes.
We also have obligations to make future payments to licensors that become due
and payable on the achievement of certain development, regulatory, and
commercial milestones. We have not included any such potential obligations on
our condensed consolidated balance sheet since the achievement and timing of
these milestones were not probable or estimable as of September 30, 2020.
Off-Balance Sheet Arrangements
As of September 30, 2020, we did not have any relationships with unconsolidated
entities or financial partnerships, such as entities often referred to as
"special purpose" entities, which would have been established for the purpose of
facilitating off-balance sheet arrangements or other contractually narrow or
limited purposes.
                                       42

--------------------------------------------------------------------------------

Table of Contents

© Edgar Online, source Glimpses