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 the related notes to those statements included elsewhere in this Quarterly Report on Form 10-Q and in our Annual Report on Form 10-K filed with the Securities and Exchange Commission, or SEC, on March 14, 2022. In addition to historical financial information, the following discussion and analysis contains forward-looking statements that involve risks, uncertainties and assumptions. Our actual results may differ materially from those anticipated in these forward-looking statements as a result of many factors, including those discussed in "Risk Factors" in Part II, Item 1A. You should carefully read the section entitled "Risk Factors" to gain an understanding of the important factors that could cause actual results to differ materially from our forward- looking statements.

Overview

We are a clinical-stage genetic medicines company pioneering a new approach to the care of cardiovascular disease, or CVD, transforming treatment from chronic management to single-course gene editing medicines. Despite advances in treatment over the last 50 years, CVD remains the leading cause of death worldwide. The current paradigm of chronic care is fragile-requiring rigorous patient adherence, extensive healthcare infrastructure and regular healthcare access-and leaves many patients without adequate care. Our goal is to disrupt the chronic care model for CVD by providing a new therapeutic approach with single-course in vivo gene editing treatments focused on addressing the root causes of this highly prevalent and life-threatening disease. Our initial two programs target PCSK9 and ANGPTL3, respectively, genes that have been extensively validated as targets for lowering blood lipids, such as low-density lipoprotein cholesterol, or LDL-C. We believe that editing these genes could potently and durably lower LDL-C throughout the lifetime of patients with or at risk for atherosclerotic cardiovascular disease, or ASCVD, the most common form of CVD.

Our approach leverages multiple breakthroughs in 21st century biomedicine-human genetic analysis, gene editing, messenger RNA, or mRNA, -based therapies and lipid nanoparticle, or LNP, delivery-to target genes that are predominantly expressed in the liver and disrupt the production of proteins that cause CVD. We are advancing a pipeline of single-course in vivo gene editing programs, each designed to mimic natural disease resistance mutations and turn off specific genes in order to lower blood lipids, thereby reducing the risk of ASCVD. We intend to initially develop these programs for the treatment of patients with familial hypercholesterolemia, or FH, a genetic disease that causes life-long severely elevated blood LDL-C, leading to increased risk of early-onset ASCVD. If our programs are successful in FH, we believe they could also provide a potential treatment for the broader population of patients with established ASCVD. Ultimately, we believe that these treatments could potentially be developed for administration to people at risk for ASCVD as a preventative measure similar to the way that certain vaccines offer long-term protection against infectious diseases.

We were incorporated in March 2018 and commenced operations shortly thereafter. Since our inception, we have devoted substantially all of our resources to building our gene editing and LNP technology and advancing development of our portfolio of programs, establishing and protecting our intellectual property, conducting research and development activities, organizing and staffing our company, business planning, raising capital and providing general and administrative support for these operations. To date, we have financed our operations primarily through the sales of our preferred stock and through the sale of our common stock in our initial public offering, or IPO, and our follow-on public offering.

On June 21, 2021, we completed our IPO in which we issued and sold an aggregate of 16,141,157 shares of our common stock, including 2,105,368 shares of common stock sold pursuant to the underwriters' full exercise of their option to purchase additional shares, at a public offering price of $19.00 per share, for aggregate net proceeds of $281.6 million, after deducting underwriting discounts and offering expenses payable by us. Upon completion of the IPO, all 256,682,054 shares of outstanding convertible preferred stock automatically converted into 27,720,923 shares of common stock. Through June 30, 2022, we had raised an aggregate of $523.2 million in gross proceeds from the sale of our preferred stock in private placements and common stock in our IPO.

On July 18, 2022, we entered into a Strategic Collaboration and License Agreement, or the Vertex Agreement, with Vertex Pharmaceuticals Incorporated, or Vertex, for an exclusive, four-year worldwide research collaboration focused on developing in vivo gene editing candidates toward an undisclosed target for the treatment of a single liver disease. Pursuant to the Vertex Agreement, Vertex paid us $25.0 million in an upfront payment on July 20, 2022. We are eligible to receive (i) success payments of up to $22 million for each product candidate (up to a maximum of $66 million) that achieves the applicable development criteria and (ii) up to an aggregate of $340 million in development and commercial milestone payments. We are also eligible to receive tiered single-digit royalties on net sales, subject to specified reductions.



                                       19

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

On July 18, 2022, in connection with the execution of the Vertex Agreement, we also entered into a stock purchase agreement with Vertex, or the Stock Purchase Agreement, for the sale and issuance of 1,519,756 shares of our common stock to Vertex at a price of $23.03 per share, which is equal to the five-day volume-weighted average share price as of July 15, 2022, for an aggregate purchase price of $35.0 million, or the Private Placement. The Private Placement closed on July 20, 2022.

On July 25, 2022, we issued and sold 9,583,334 shares of our common stock, including an additional 1,250,000 shares of common stock sold pursuant to the underwriters' full exercise of their option to purchase additional shares of common stock, at a public offering price of $27.00 per share, for aggregate net proceeds of approximately $242.9 million after deducting underwriting discounts and commissions of approximately $15.5 million and offering costs of approximately $0.3 million.

We are a clinical-stage company. To date, we have not generated any revenue from product sales and do not expect to generate revenue from the sale of products for the foreseeable future. Prior to the execution of the Vertex Agreement, on July 18, 2022, we had not recorded any revenue. Since our inception, we have incurred significant operating losses. Our net losses for the three and six months ended June 30, 2022 were $40.9 million and $71.1 million, respectively. Our net losses for the three and six months ended June 30, 2021 were $53.0 million and $66.2 million, respectively. As of June 30, 2022, we had an accumulated deficit of $258.0 million.

We expect to continue to incur significant expenses and increasing operating losses in connection with ongoing development activities related to our portfolio of programs as we advance VERVE-101 in our heart-1 clinical trial; continue our preclinical development of other product candidates; advance these product candidates toward clinical development; further develop base editing and novel gene editing technology, delivery technology and manufacturing capabilities; seek to discover and develop additional product candidates including VERVE-201, our development candidate targeting ANGPTL3; maintain, expand enforcement, defend, and protect our intellectual property portfolio; hire research and development and clinical personnel; ultimately establish a sales, marketing and distribution infrastructure to commercialize any products for which we may obtain marketing approval; 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; and add operational, legal, compliance, financial and management information systems and personnel to support our research, product development, future commercialization efforts and operations as a public company.

As a result, we will need substantial additional funding to support our continuing operations and pursue our 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 equity offerings, debt financings and other sources of capital, which may include collaborations or licensing arrangements with other companies or other strategic transactions. If we are unable to raise capital or obtain adequate funds when needed or on acceptable terms, we may be required to delay, limit, reduce or terminate our research and development programs or any future commercialization efforts or grant rights to develop and market product candidates that we would otherwise prefer to develop and market ourselves.

Because of the numerous risks and uncertainties associated with product development, we are unable to predict the timing or amount of increased expenses or when or if we will be able to achieve profitability. Even if we are able to generate revenue from 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.

As of June 30, 2022, we had cash, cash equivalents and marketable securities of $293.6 million. We believe that our existing cash, cash equivalents and marketable securities, including the net proceeds of approximately $242.9 million from the July 2022 follow-on public offering, $35.0 million from the Private Placement with Vertex and $25.0 million upfront payment pursuant to the Vertex Agreement, will enable us to fund our operating expenses and capital expenditure requirements into the second half of 2025. We have based this estimate on assumptions that may prove to be wrong, and we could exhaust our available capital resources sooner than we expect. To finance our operations beyond that point we will need to raise additional capital, which cannot be assured. See "Liquidity and capital resources."

Recent Developments

VERVE-101

Our lead product candidate, VERVE-101, is designed to permanently turn off the PCSK9 gene in the liver. PCSK9 is a highly validated target that plays a critical role in controlling blood LDL-C through its regulation of the LDL receptor, or LDLR. Reduction of PCSK9 protein in the blood improves the ability of the liver to clear LDL-C from the blood. VERVE-101 utilizes LNP-mediated delivery to target the liver and base editing technology to make a single base change at a specific site in the PCSK9 gene in order to disrupt PCSK9 protein production.



                                       20

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

VERVE-101 is being developed for the treatment of patients with heterozygous familial hypercholesterolemia, or HeFH, which affects approximately 1.3 million people in the United States and approximately 31 million worldwide. We are strategically developing VERVE-101 initially in patients with HeFH, recognizing that the unmet need is highest in those patients and the benefit-risk profile may be more favorable. We intend to use a stepwise clinical development plan for VERVE-101, evaluating efficacy and safety in higher-risk populations first, and then if successful, expanding into broader population of patients with established ASCVD, and ultimately to those at risk for ASCVD in the general population.

In an ongoing in vivo proof-of-concept study of a precursor formulation of VERVE-101 in non-human primates, or NHPs, we observed substantial lowering of LDL-C levels that was sustained over an extended period of time following treatment. In this study, following a single intravenous infusion of a base editor targeting PCSK9, we observed an average reduction of blood PCSK9 protein of 89% accompanied by an average reduction of blood LDL-C levels of 59% at two weeks after treatment. This LDL-C reduction was maintained at an average of greater than 60% for 20 months following treatment. If we are able to achieve similar reductions in PCSK9 protein levels in humans, we believe this could result in marked and sustained LDL-C reductions of approximately 60%, which would potentially offer superior cumulative LDL-C lowering to what has been clinically demonstrated with other PCSK9-targeting treatment modalities.

In an ongoing preclinical study with VERVE-101 in NHPs, we observed 70% mean editing following a single administration of 1.5 mg/kg dose at the PCSK9 target gene site in liver biopsies taken at day 15. In this study, we also observed an average reduction in blood PCSK9 protein of 86% accompanied by an average reduction of blood LDL-C levels of 62% at two weeks after treatment. These reductions were durable when assessed 12 months after treatment, with mean reduction in blood PCSK9 protein of 89% and blood LDL-C levels of 68%.

In our preclinical studies in NHPs to date, VERVE-101 has been well tolerated following a single administration with only mild elevations in liver function tests that resolved within two weeks.

In GLP toxicology studies, we observed that the highest dose that did not produce a significant increase in adverse effects relative to control was 2.0 mg/kg and 5.0 mg/kg in NHPs and a mouse model, respectively. In the toxicology studies in NHPs, we observed transient mild elevations in liver function tests and evidence of minimal to mild single cell hepatocyte necrosis two days after dosing and all findings were reversible and observed to be resolved at a 90-day measurement. In the mouse model, we did not observe any differences between wild-type mice or the HeFH mouse disease model.

In April 2022, we presented data from a comprehensive preclinical assessment of the potential for VERVE-101 to cause unintended, or off-target, DNA edits in primary human liver cells from multiple donors. We used multiple methods consistent with recent guidance from the U.S. Food and Drug Administration to identify more than 3,000 sites with the greatest experimental or bioinformatic similarity to the on-target site. We then used a sequencing assay to determine whether administration of VERVE-101 resulted in off-target editing at those sites. We did not observe any statistically significant off-target editing after treatment with VERVE-101 at the identified sites. We also evaluated the potential for off-target editing in non-target cells (spleen cells, adrenal cells, and hematopoietic stem cells) and other cellular contexts (pediatric human liver cells and human liver cell lines) and identified only two sites with statistically significant editing above untreated controls. The two instances of off-target editing occurred at doses greater than the dose we expect to achieve saturation for on-target editing. Based on these assessments, we believe that VERVE-101 has a low risk of off-target genomic modifications that would be expected to have an associated clinical adverse effect.

We have received clearance of our first clinical trial application, or CTA, for VERVE-101 in New Zealand and in July 2022, we announced that the first patient has been dosed with VERVE-101 in our heart-1 clinical trial. The trial is underway and is enrolling patients with HeFH in New Zealand. We anticipate obtaining regulatory clearances for a CTA in the United Kingdom and an investigational new drug application, or IND, in the United States in the second half of 2022. We are on track to complete the remaining IND-enabling studies in the third quarter of 2022.

The heart-1 clinical trial is designed to enroll approximately 40 adult HeFH patients with established ASCVD and evaluate the safety and tolerability of VERVE-101 administration, with additional analyses for pharmacokinetics and reductions in blood PCSK9 protein and LDL-C. The trial includes three parts - (A) a single ascending dose portion, followed by (B) an expansion single-dose cohort, in which additional participants will receive the selected potentially therapeutic dose and (C) an optional second-dose cohort, in which eligible participants in lower dose cohorts in Part A have the option to receive a second treatment at the selected potentially therapeutic dose. We expect to report interim clinical data from the heart-1 clinical trial including safety parameters, blood PCSK9 level, and blood LDL-C level in 2023.

ANGPTL3 Program

VERVE-201, our development candidate targeting ANGPTL3, is designed to permanently turn off the ANGPTL3 gene in the liver. We plan to develop this program initially for the treatment of homozygous familial hypercholesterolemia, or HoFH, which affects approximately 1,300 patients in the United States. Similar to our approach with VERVE-101, we plan to expand the clinical development of VERVE-201 in a stepwise fashion beyond HoFH to patients with established



                                       21

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

ASCVD who may need additional LDL-C and/or triglycerides reduction. Ultimately, we believe that VERVE-201 may also be useful to people at risk for ASCVD as a preventative measure in the general population. We plan to present updated preclinical data supporting VERVE-201's advancement at the ESC 2022 Congress. We expect to begin IND-enabling studies in the second half of 2022.

We plan to utilize internally developed GalNAc-LNP technology in VERVE-201 to deliver a base editor targeting the ANGPTL3 gene to the liver. In patients with HoFH, delivery of base editors with standard LNPs to the liver is challenging due to the deficiency of LDLR, which is known to mediate LNP uptake. We have developed proprietary LNPs with a GalNAc ligand designed to bind to asialoglycoprotein receptors, in the liver, which bypass LDLR, thereby enabling uptake into the liver in HoFH patients.

In an ongoing proof of concept study of an ANGPTL3 base editor in NHPs (n=4), we observed a 96% reduction in blood ANGPTL3 protein from baseline, with follow-up out to approximately 20 months. In addition, no long-term impacts were observed on markers of liver toxicity, as measured by alanine aminotransferase (ALT) and bilirubin levels following treatment administration.

In addition, in our preclinical studies of our ANGPTL3 program using a single treatment of two different formulations of our proprietary GalNAc-LNPs to deliver an ANGPTL3-targeted base editor we observed approximately 94% (n=3) and 97% (n=3) reduction in blood ANGPTL3 protein, and reductions in LDL-C of nearly 100 mg/dL, which was an approximately 35% reduction from baseline. We conducted these studies in an internally developed NHP model of HoFH, which we created by editing the LDLR gene in wild-type NHPs and eliminating LDLR expression in the livers of NHPs using a Cas9 and dual guide RNA strategy encapsulated in standard LNPs, which led to nearly 70% whole liver DNA editing at the LDLR gene and resulted in an approximately 94% reduction in LDLR protein in the liver and a six-fold increase in blood LDL-C.

We have also assessed the potential broad utility of our proprietary GalNAc-LNP approach for delivery of our ANGPTL3-targeted base editor, in a preclinical study evaluating delivery efficiency of an ANGPTL3 base editor using either a GalNAc-LNP or a standard LNP without GalNAc in wild-type NHPs with normal livers. In these studies, we observed that wild-type NHPs treated with our ANGPTL3-targeted base editor delivered via our GalNAc-LNP had an approximately 89% reduction in ANGPTL3 protein compared to an approximately 74% reduction in wild-type NHPs treated with a standard LNP. A follow up study in which we performed dose-response assessments at three dose levels (0.75 mg/kg, 1.5 mg/kg and 3.0 mg/kg) in wild-type NHPs confirmed the observation that the addition of GalNAc-lipid to a standard LNP enhanced potency of liver base editing. In this study, we observed greater editing potency of a ANGPTL3 precursor formulation with our GalNAc-LNP than with standard LNPs at day 14. We also observed that the use of a GalNAc-LNP led to reductions in blood ANGPTL3 protein at each dose level at day 14, including a 51% reduction at 0.75mg/kg, an 83% reduction at 1.5 mg/kg and a 98% reduction at 3.0 mg/kg. In this study, the alanine aminotransferase profiles over 14 days following treatment did not differ between NHPs treated with GalNAc-LNP versus a standard LNP. We also evaluated the biodistribution of editing following administration of an ANGPTL3 precursor at a dose of 1.5 mg/kg using our GalNAc-LNP and observed editing to be localized to the liver with minimal biodistribution to other organs.

We believe this data suggests that GalNAc-LNP delivery may have broad utility for liver editing in other indications.

We are continuing to invest and build out capabilities in the development of novel and optimized GalNAc-targeting ligands, optimal lipid anchors, optimal compositions and ratios of LNP components, and optimal processes of addition and LNP formation with targeting ligands. We believe GalNAc provides a delivery platform for patients with both forms of FH and potentially may be applicable in other applications where liver-directed delivery is advantageous.

Impact of COVID-19 on our business

In March 2020, COVID-19 was declared a global pandemic by the World Health Organization and to date, the COVID-19 pandemic continues to present a substantial public health and economic challenge around the world. The length of time and full extent to which the COVID-19 pandemic may directly or indirectly impact our business, results of operations and financial condition will depend on future developments that are highly uncertain, subject to change and difficult to predict. We, our contract manufacturing organizations, or CMOs, and our contract research organizations, or CROs, experienced temporary reductions in the capacity to undertake research-scale production and to execute some preclinical studies. While these operations have since normalized, we, together with our CMOs and CROs, continue to closely monitor the impact of the COVID-19 pandemic on these operations.

We also plan to continue to closely monitor the ongoing impact of the COVID-19 pandemic on our employees and our other business operations. In an effort to provide a safe work environment for our employees, we have, among other things, limited employees in our office and lab facilities to those where on-site presence is needed for their job activities, increased the cadence of sanitization of our office and lab facilities, implemented various social distancing measures in our offices and labs including replacing all in-person meetings with virtual interactions. Recently, additional employees



                                       22

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

have returned to our office and lab facilities in limited capacities. We continue to provide personal protective equipment and recommend regular COVID-19 testing for employees and visitors present in our office and lab facilities. We continue to monitor the impact and effects of the COVID-19 pandemic and our response to it, and we expect to continue to take actions as may be required or recommended by government authorities or as we determine are in the best interests of our employees and other business partners in light of the pandemic.

While the COVID-19 pandemic did not significantly impact our business or results for the three and six months ended June 30, 2022 and 2021, the length and extent of the pandemic, its consequences, and containment efforts will determine the future impact on our operations and financial condition.

License and collaboration agreements

We have obligations under various license and collaboration agreements to make potentially significant milestone and success payments in the future and to pay royalties on sales of any product candidates covered by those agreements that eventually achieve regulatory approval and commercialization. For information regarding these agreements, see Note 8, "License agreements" and Note 14, "Subsequent events" to our condensed consolidated financial statements included in this Quarterly Report on Form 10-Q.

Components of our results of operations

Revenue

Through June 30, 2022 and prior to the execution of the Vertex Agreement, we had not recorded any revenue. We do not expect to generate any revenue from the sale of products in the near future and unless and until we successfully complete development and obtain regulatory approval for one or more of our product candidates. If our development efforts for our product candidates are successful and result in regulatory approval or we successfully enter into license or collaboration agreements with third parties, in addition to the Vertex Agreement, we may generate revenue in the future from product sales, payments from such additional third-party collaboration or license agreements, or any combination thereof.

Operating expenses

Research and development expenses

Research and development expenses consist of costs incurred in performing research and development activities, which include:

the cost to obtain and maintain licenses to intellectual property, such as those with the President and Fellows of Harvard College, or Harvard, The Broad Institute, Inc., or Broad, Beam Therapeutics Inc., or Beam, Acuitas Therapeutics, Inc., or Acuitas, and others, and related future payments should certain development and regulatory milestones be achieved;

personnel-related expenses, including salaries, bonuses, benefits and stock-based compensation for employees engaged in research and development functions;

expenses incurred in connection with the discovery and preclinical development of our research programs, including under agreements with third parties, such as consultants, contractors and CROs;

the cost of developing and validating our manufacturing process for use in our preclinical studies and ongoing, planned and future clinical trials, including the cost of raw materials used in our research and development activities;

the cost of laboratory supplies and research materials; and

facilities, depreciation and other expenses, which include direct and allocated expenses for rent and maintenance of facilities and insurance.

We expense research and development costs as incurred. Nonrefundable 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 benefits are consumed.

In the early phases of development, our research and development costs are often devoted to proof-of-concept studies that are not necessarily allocable to a specific target; therefore, we have not yet begun tracking our expenses on a program-by-program basis.

Research and development activities are central to our business model. We expect that our research and development expenses will continue to increase for the foreseeable future as we advance our programs and product candidates into and through clinical development, as we continue to develop additional product candidates, and as we continue to



                                       23

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

develop our gene editing and LNP technology. We also expect our discovery research efforts and our related personnel costs will increase and, as a result, we expect our research and development expenses, including costs associated with stock-based compensation, will increase above historical levels. In addition, we may incur additional expenses related to milestone and royalty payments payable to third parties with whom we may enter into license, acquisition and option agreements to acquire the rights to future product candidates.

At this time, we cannot reasonably estimate or know the nature, timing and costs of the efforts that will be necessary to complete the preclinical and clinical development of, and obtain regulatory approval for, any of our product candidates or programs. The successful development of our product candidates is highly uncertain. This is due to the numerous risks and uncertainties associated with product development, 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;

raising additional funds necessary to complete preclinical and clinical development of our product candidates;

the timing of filing and acceptance of INDs or comparable foreign applications that allow commencement of planned and future clinical trials for our product candidates;

the successful initiation, enrollment and completion of clinical trials;

our ability to achieve positive results from our ongoing and future clinical programs that support a finding of safety and effectiveness and an acceptable risk-benefit profile in the intended patient populations of any product candidates we may develop;

our ability to successfully develop, obtain regulatory approval for, and then successfully commercialize, our product candidates for the expected indications and patient populations;

our ability to hire and retain key research and development personnel;

the costs associated with the development of any additional product candidates we develop or acquire through collaborations;

our ability to establish and maintain agreements with third-party manufacturers for clinical supply for our clinical trials and commercial manufacturing, if our product candidates are approved;

the terms and timing of any existing or future collaboration, license or other arrangement, including the terms and timing of any milestone payments thereunder;

our ability to establish and obtain intellectual property protection and regulatory exclusivity for our product candidates and enforce and defend our intellectual property rights and claims;

our ability to commercialize products, if and when approved, whether alone or in collaboration with others;

our ability to maintain a continued acceptable safety, tolerability and efficacy profile of our product candidates following approval; and

the effects of the COVID-19 pandemic.

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

General and administrative expenses

General and administrative expenses consist primarily of personnel-related costs, including salaries, benefits and stock-based compensation, for personnel in our executive, intellectual property, business development, and administrative functions. General and administrative expenses also include legal fees relating to intellectual property and corporate matters, professional fees for accounting, auditing, tax and consulting services, insurance costs, travel, and direct and allocated facility-related expenses and other operating costs.

We anticipate that our general and administrative expenses will increase in the future to support increased research and development activities. We also expect to continue to incur increased costs associated with being a public company, including costs of accounting, audit, legal, regulatory and tax-related services associated with maintaining compliance with Nasdaq and SEC requirements, director and officer insurance costs, and investor and public relations costs.



                                       24

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

Other income

Change in fair value of antidilution rights liability

Change in fair value of antidilution rights liability consists of remeasurement gains or losses associated with changes in the antidilution rights liability associated with our license agreements with Harvard and Broad, or the Harvard/Broad License Agreement, and Broad, or the Broad License Agreement.

The antidilution rights represented the obligation to issue additional shares of common stock to Harvard and Broad following the completion of preferred stock financings and other equity financings, which was fully satisfied upon the closing of our IPO. At the inception of the agreements, the liability for the antidilution rights was recorded at fair value with the cost recorded as research and development expense and were remeasured at each reporting period with changes recorded in other income (expense) while the instruments are outstanding.

The antidilution rights liability was partially satisfied in 2019 and 2020 and was satisfied in full in June 2021 upon the closing of our IPO with the issuance of an additional 878,098 shares of our common stock.

Change in fair value of success payment liability

We are also obligated to pay to Harvard and Broad tiered success payments in the event our average market capitalization exceeds specified thresholds ascending from a high nine-digit dollar amount to $10.0 billion, or sale of our company for consideration in excess of those thresholds. In the event of a change of control of our company or a sale of our company, we are required to pay any related success payment in cash within a specified period following such event. Otherwise, the success payments may be settled at our option in either cash or shares of our common stock, or a combination of cash and shares of our common stock. The maximum aggregate success payments that could be payable by us are $31.3 million. At inception of the agreements, the success payment liabilities were recorded at fair value with the cost recorded as research and development expense and are being remeasured at each reporting period with charges recorded in other income (expense) while the instrument is outstanding.

Depending on our valuation, the fair value of the success payment liability, and the corresponding changes in fair value that we record in our statements of operations, could fluctuate significantly from period to period.

During the year ended December 31, 2021, certain success payment obligations were triggered, and amounts due to Harvard and Broad totaled $6.3 million. These amounts were settled in cash in November 2021. The remaining success payment obligations will continue to be revalued at the end of each reporting period.

Interest and other income, net

Interest and other income primarily consisted of interest earned on our marketable securities and other miscellaneous income and expenses unrelated to our core operations.

Income tax

During the three and six months ended June 30, 2022 and 2021, we recorded a full valuation allowance on federal and state deferred tax assets since management does not forecast that we will be in a taxable position in the near future.



                                       25

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

Results of operations

Comparison of three months ended June 30, 2022 and 2021

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



                                                        Three months ended
                                                                  June 30,
(in thousands)                                         2022           2021         Change
Operating expenses:
Research and development                         $   33,125     $   13,423     $   19,702
General and administrative                            9,067          3,541          5,526
Total operating expenses                         $   42,192     $   16,964     $   25,228
Other income (expense):
Change in fair value of antidilution rights
liability                                                 -        (25,970 )       25,970
Change in fair value of success payment
liability                                               938        (10,036 )       10,974
Interest and other income, net                          308              5            303
Total other income (expense)                     $    1,246     $  (36,001 )   $   37,247
Net loss                                         $  (40,946 )   $  (52,965 )   $   12,019

Research and development expenses

The following table summarizes our research and development expenses for the three months ended June 30, 2022 and 2021:



                                                        Three months ended
                                                                  June 30,
(in thousands)                                         2022           2021         Change
Employee-related expenses                       $    10,917     $    4,113     $    6,804
External expenses associated with preclinical
studies performed by outside consultants,
including third-party CROs                            7,462          3,681          3,781
Raw material costs and external expenses
associated with manufacturing activities,
including third-party CMOs                            5,552          2,990          2,562
License and milestone payments                        3,047             25          3,022
Lab supplies                                          2,369          1,138          1,231
Facility-related costs (including
depreciation)                                         1,602            821            781
Clinical trial costs                                  1,305              -          1,305
Other research and development costs                    871            655            216

Total research and development expenses $ 33,125 $ 13,423 $ 19,702

Research and development expenses were $33.1 million for the three months ended June 30, 2022, compared to $13.4 million for the three months ended June 30, 2021. The increase of $19.7 million was primarily due to the following:

an increase in personnel-related costs of $6.8 million, including stock-based compensation driven by an increase in headcount of employees involved in research and development activities;

an increase in external expenses associated with preclinical studies (primarily animal-study costs) performed by outside consultants, including third-party CROs, of $3.8 million;

an increase in raw material costs and external expenses associated with developing and validating our manufacturing activities, including third-party CMOs, for use in our preclinical studies and clinical trial of $2.6 million;

an increase in research and development expense attributed to license and milestone payments of $3.0 million in 2022;

an increase in lab supplies of $1.2 million due to increased headcount and company growth;

an increase in facility-related costs (including depreciation) and other allocated miscellaneous expenses of $0.8 million due to increased investment in research and development;

an increase in clinical trial costs of $1.3 million associated with our heart-1 clinical trial, a Phase 1b clinical trial of VERVE-101; and

an increase in other research and development costs of $0.2 million, primarily due to an increase in professional fees and consulting fees in support of increased investment in research and development activities.



                                       26

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

We expect that our research and development expenses will continue to increase for the foreseeable future as we advance our programs and product candidates into and through clinical development, and as we continue to develop additional product candidates and invest in our technology and manufacturing capabilities.

General and administrative expenses

General and administrative expenses were $9.1 million for the three months ended June 30, 2022, compared to $3.5 million for the three months ended June 30, 2021. The increase of approximately $5.5 million was primarily attributable to the following:

an increase of $3.5 million in personnel, facility and other expenses stemming from an increase in headcount to support our growth;

an increase of $1.4 million in legal and professional service fees, primarily due to increased professional fees for audit, tax and consulting services; and

an increase in other miscellaneous expenses of $0.6 million, primarily due to increased insurance expense for our directors and officers insurance policy and other increases in software, IT, and other miscellaneous charges.

We anticipate that our general and administrative expenses will increase in the future to support increased research and development activities.

Other income (expense)

Change in fair value of antidilution rights liability

The decrease in the change in the fair value of the antidilution rights liability was as a result of the liability being fully settled during the three months ended June 30, 2021 with the issuance of 878,098 shares of our common stock.

Change in fair value of success payments liability

During the three months ended June 30, 2022, the change in fair value of the success payments liability was primarily due to the decrease in the fair value of our common stock, which resulted in a fair value adjustment of $0.9 million to other income. During the three months ended June 30, 2021, the change in fair value for the success payments liability of $10.0 million was primarily due to the increase in the fair value of our common stock.

Interest and other income, net

The increase of $0.3 million in interest and other income, net for the three months ended June 30, 2022 compared to the three months ended June 30, 2021 was primarily attributable to increasing interest rates on marketable security balances.

Comparison of six months ended June 30, 2022 and 2021

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



                                                          Six months ended
                                                                  June 30,
(in thousands)                                         2022           2021         Change
Operating expenses:
Research and development                             57,614     $   24,768     $   32,846
General and administrative                           16,503          6,257         10,246
Total operating expenses                         $   74,117     $   31,025     $   43,092
Other income (expense):
Change in fair value of antidilution rights
liability                                                 -        (25,574 )       25,574
Change in fair value of success payment
liability                                             2,615         (9,654 )       12,269
Interest and other income, net                          390             25            365
Total other income (expense)                     $    3,005     $  (35,203 )   $   38,208
Net loss                                         $  (71,112 )   $  (66,228 )   $   (4,884 )




                                       27

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

Research and development expenses

The following table summarizes our research and development expenses for the six months ended June 30, 2022 and 2021:



                                                         Six months ended
                                                                 June 30,
(in thousands)                                        2022           2021         Change
Employee-related expenses                       $   19,849     $    6,806     $   13,043
External expenses associated with preclinical
studies performed by outside consulting
services, including third-party CROs                13,702          7,691          6,011
Raw material costs and external expenses
associated with manufacturing activities,
including third-party CMOs                           9,657          5,385          4,272
Lab supplies                                         3,775          1,869          1,906
License and milestone payments                       3,400             80          3,320
Facility-related costs (including
depreciation)                                        3,031          1,600          1,431
Clinical trial costs                                 1,317              -          1,317
Other research and development costs                 2,883          1,337          1,546

Total research and development expenses $ 57,614 $ 24,768 $ 32,846

Research and development expenses were $57.6 million for the six months ended June 30, 2022, compared to $24.8 million for the six months ended June 30, 2021. The increase of $32.8 million was primarily due to the following:

an increase in personnel-related costs of $13.0 million, including stock-based compensation, driven by an increase in headcount of employees involved in research and development activities;

an increase in external expenses associated with preclinical studies (primarily animal-study costs) performed by outside consultants, including third-party CROs, of $6.0 million;

an increase in raw material costs and external expenses associated with developing and validating our manufacturing activities, including third-party CMOs, for use in our preclinical studies and clinical trial of $4.3 million;

an increase in lab supplies of $1.9 million due to increased headcount and company growth;

an increase in research and development expense attributed to license and milestone payments of $3.3 million in 2022;

an increase in facility-related costs (including depreciation) and other allocated miscellaneous expenses of $1.4 million due to increased investment in research and development;

an increase in clinical trial costs of $1.3 million associated with our heart-1 clinical trial, a Phase 1b clinical trial of VERVE-101; and

an increase in other research and development costs of approximately $1.6 million, primarily due to an increase in professional fees and consulting fees in support of increased investment in research and development activities.

We expect that our research and development expenses will continue to increase for the foreseeable future as we advance our programs and product candidates into and through clinical development, and as we continue to develop additional product candidates and invest in our technology and manufacturing capabilities.

General and administrative expenses

General and administrative expenses were $16.5 million for the six months ended June 30, 2022,compared to $6.3 million for the six months ended June 30, 2021. The increase of $10.2 million was primarily attributable to the following:

an increase of $6.5 million in personnel, facility and other expenses stemming from an increase in headcount to support our growth;

an increase of $2.1 million in legal and professional service fees, primarily due to increased professional fees for audit, tax and consulting services; and

an increase in other miscellaneous expenses of $1.6 million, primarily due to increased insurance expense of $1.3 million for our directors and officers insurance policy and other increases in software, IT, and other miscellaneous charges.

We anticipate that our general and administrative expenses will increase in the future to support increased research and development activities.



                                       28

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

Other income (expense)

Change in fair value of antidilution rights liability

The decrease in the change in the fair value of the antidilution rights liability was as a result of the liability being fully settled during the six months ended June 30, 2021 with the issuance of 878,098 shares of our common stock.

Change in fair value of success payments liability

During the six months ended June 30, 2022, the change in fair value of the success payments liability was primarily due to the decrease in the fair value of our common stock, which resulted in a fair value adjustment of $2.6 million to other income. During the six months ended June 30, 2021, the change in fair value for the success payments liability of $9.7 million was primarily due to the increase in the fair value of our common stock.

Interest and other income, net

The increase of $0.4 million in interest and other income, net for the six months ended June 30, 2022 compared to the six months ended June 30, 2021 was primarily attributable to higher balances and increasing interest rates on marketable security balances.

Liquidity and capital resources

Sources of liquidity and capital

Since our inception in 2018, we have incurred significant operating losses. We expect to incur significant expenses and operating losses for the foreseeable future as we advance the preclinical and, if successful, clinical development of our programs. To date, we have funded our operations primarily through equity offerings. Through June 30, 2022, we had raised an aggregate of $523.2 million in gross proceeds from sales of our preferred stock in private placements and common stock in our IPO.

In June 2021, we completed our IPO in which we issued 16,141,157 shares of our common stock, including 2,105,368 shares of common stock sold pursuant to the underwriters' full exercise of their option to purchase additional shares, at a public offering price of $19.00 per share. We received net proceeds from our IPO of $281.6 million, after deducting underwriting discounts and offering expenses payable by us. In June 2021, we issued 878,098 shares of our common stock to Harvard and Broad as final settlement of the antidilution rights liability.

As of June 30, 2022, we had $293.6 million in cash, cash equivalents and marketable securities. On July 20, 2022, we received $25.0 million from Vertex pursuant to the Vertex Agreement. Additionally, on July 20, 2022, we sold and issued 1,519,756 shares of our common stock to Vertex in connection with the Private Placement at a price of $23.03 per share for an aggregate purchase price of $35.0 million. On July 25, 2022, we issued and sold 9,583,334 shares of our common stock, including an additional 1,250,000 shares of common stock sold pursuant to the underwriters' full exercise of their option to purchase additional shares of common stock, at a public offering price of $27.00 per share, for aggregate net proceeds of approximately $242.9 million after deducting underwriting discounts and commissions of approximately $15.5 million and offering costs of approximately $0.3 million.

On July 1, 2022, we entered into an Open Market Sale AgreementSM, or the Sales Agreement, with Jefferies LLC, or Jefferies, as agent, pursuant to which we may offer and sell shares of our common stock having an aggregate offering price of up to $150.0 million from time to time through Jefferies under an "at-the-market offering" program. Upon delivery of a placement notice and subject to the terms and conditions of the Sales Agreement, Jefferies may sell the shares of our common stock by any method permitted by law deemed to be an "at-the-market offering" as defined in Rule 415(a)(4) promulgated under the Securities Act of 1933, as amended. We may sell the shares in amounts and at times to be determined by us from time to time subject to the terms and conditions of the Sales Agreement, but we have no obligation to sell any shares under the Sales Agreement. We agreed to pay Jefferies a commission of up to 3% of the gross proceeds of any sales of common stock pursuant to the Sales Agreement. We or Jefferies may suspend or terminate the offering of shares upon notice to the other party and subject to other conditions. On July 20, 2022, we delivered written notice to Jefferies that we were suspending and terminating the prospectus, or the ATM Prospectus, related to the sale of our common stock pursuant to the Sales Agreement. As a result, we will not make any sales of our common stock pursuant to the Sales Agreement unless and until a new prospectus, prospectus supplement or registration statement is filed. The Sales Agreement remains in full force and effect. As of the termination of the at-the-market offering, we had not issued and sold any shares of our common stock under the ATM Prospectus.



                                       29

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

Cash flows



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


                                                                    Six months ended
                                                                            June 30,
(in thousands)                                                    2022          2021
Cash used in operating activities                            $ (60,265 )   $ (28,482 )
Cash provided by investing activities                           67,874        30,203
Cash provided by financing activities                              950       376,732

Net increase in cash, cash equivalents and restricted cash $ 8,559 $ 378,453






Operating activities

For the six months ended June 30, 2022, net cash used in operating activities was $60.3 million, consisting primarily of our net loss of $71.1 million, partially offset by changes in our operating assets and liabilities of approximately $0.1 million and by the following non-cash changes: stock-based compensation of $9.9 million, depreciation expense of $1.2 million, non-cash lease expense of $1.0 million and amortization of investment premiums of $1.2 million offset by the non-cash change in the fair value of the success payment liability of $2.6 million.

For the six months ended June 30, 2021, net cash used in operating activities was $28.5 million, consisting primarily of our net loss of $66.2 million and a decrease in our operating assets and liabilities of approximately $1.5 million. These amounts were partially offset by the following non-cash changes: $25.6 million associated with the fair value change in antidilution rights liability, $9.7 million associated with the fair value change in success payments liability, stock-based compensation of $2.0 million, non-cash lease expense of approximately $0.8 million, depreciation expense of $0.7 million and amortization of investment premiums of $0.4 million.

Investing activities

For the six months ended June 30, 2022, net cash provided by investing activities was $67.9 million and consisted of maturities of marketable securities of approximately $147.7 million, partially offset by purchases of marketable securities of $74.2 million and purchases of property and equipment of $5.6 million, primarily related to lab equipment.

For the six months ended June 30, 2021, net cash provided by investing activities was $30.2 million and consisted of maturities of marketable securities of $43.8 million, offset partially by purchases of marketable securities of $11.2 million and purchases of property and equipment of $2.4 million, primarily related to lab equipment.

Financing activities

For the six months ended June 30, 2022, net cash provided by financing activities was $1.0 million, consisting of proceeds from exercises of stock options of approximately $0.7 million and issuance of shares through our employee stock purchase plan of $0.3 million.

For the six months ended June 30, 2021, net cash provided by financing activities was $376.7 million, consisting primarily of the net proceeds from the issuance of Series B Preferred Stock of $93.8 million, net proceeds from our IPO of $285.2 million and proceeds from exercises of stock options of $0.5 million, offset partially by payment of IPO expenses of $2.8 million.

Funding requirements

Our operating expenses and future funding requirements are expected to increase substantially as we continue to advance our portfolio of programs.

Specifically, our expenses will increase if and as we:

conduct our ongoing heart-1 clinical trial for VERVE-101 in New Zealand and, if the IND and CTA are cleared, as applicable, in the United States and United Kingdom;

continue our current research programs and our preclinical development of product candidates from our current research programs;

seek to identify additional research programs and additional product candidates;

advance our existing and future product candidates into clinical development;



                                       30

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

initiate preclinical studies and clinical trials for any additional product candidates we identify and develop or expand development of existing programs into additional patient populations;

maintain, expand, enforce, defend and protect our intellectual property portfolio and provide reimbursement of third-party expenses related to our patent portfolio;

seek regulatory and marketing approvals for any of our product candidates that we develop;

seek to identify, establish and maintain additional collaborations and license agreements, and the success of those collaborations and license agreements;

make milestone payments to Beam under our amended and restated collaboration and license agreement with Beam, milestone payments to Acuitas under our non-exclusive license agreement with Acuitas, and milestone payments or success payments to Harvard and Broad under the Harvard/ Broad License Agreements, and potential payments to other third parties under our other collaboration agreements or under any additional future collaboration or license agreements that we obtain;

ultimately establish a sales, marketing, and distribution infrastructure to commercialize any drug products for which we may obtain marketing approval, either by ourselves or in collaboration with others;

generate revenue from commercial sales of product candidates we may develop for which we receive marketing approval;

further develop base editing and novel gene editing technology;

hire additional personnel including research and development, clinical and commercial personnel;

add operational, financial and management information systems and personnel, including personnel to support our product development;

acquire or in-license products, intellectual property, medicines and technologies;

satisfy any post-marketing requirements, such as a cardiovascular outcomes trial;

establish commercial-scale current good manufacturing practices capabilities through a third-party or our own manufacturing facility; and

continue to operate as a public company.

As of June 30, 2022, we had cash, cash equivalents and marketable securities of $293.6 million. We believe that our existing cash, cash equivalents and marketable securities, including the net proceeds of approximately $242.9 million from the July 2022 follow-on public offering, $35.0 million from the Private Placement with Vertex and the $25.0 million upfront payment from Vertex pursuant to the Vertex Agreement, will enable us to fund our operating expenses and capital expenditure requirements into the second half of 2025. We have based this estimate on assumptions that may prove to be wrong, and we could exhaust our available capital resources sooner than we expect.

Identifying potential product candidates and conducting preclinical testing and clinical trials is a time consuming, expensive and uncertain process that takes years to complete, and we may never generate the necessary data or results required to obtain marketing approval and achieve product sales. In addition, our product candidates, if approved, may not achieve commercial success. Our commercial revenues, if any, will be derived from sales of products that we do not expect to be commercially available for several years, if ever. Accordingly, we will need to obtain substantial additional funds to achieve our business objectives.

Our expectation with respect to our ability to fund current planned operations is based on estimates that are subject to risks and uncertainties. Our operating plan may change as a result of many factors currently unknown to management and there can be no assurance that the current operating plan will be achieved in the time frame anticipated by us, and we may need to seek additional funds sooner than planned.

Adequate additional funds may not be available to us on acceptable terms, or at all. We do not have any source of committed external funds. Market volatility could also adversely impact our ability to access capital as and when needed. Additional capital raised through the sale of equity or convertible debt securities may include liquidation or other preferences. 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, selling or licensing our assets, making capital expenditures or declaring dividends and may require the issuance of warrants.

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



                                       31

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

programs or product candidates or grant licenses on terms that may not be favorable to us. If we are unable to raise additional funds through equity or debt financings or other arrangements when needed or on terms acceptable to us, we may be required to delay, limit, reduce or terminate our product development or future commercialization efforts or grant rights to develop and market product candidates that we would otherwise prefer to develop and market ourselves.

Contractual obligations

During the three months ended June 30, 2022, there were no material changes to our contractual obligations and commitments from those described under the heading "Management's Discussion and Analysis of Financial Condition and Results of Operations-Contractual obligations" in our Annual Report on Form 10-K filed with the SEC on March 14, 2022. Refer to Note 7, "Leases," to the condensed consolidated financial statements appearing in Part I, Item 1 in this Quarterly Report on Form 10-Q for more information on our lease obligations.

Emerging growth company status

As an emerging growth company, or EGC, under the Jumpstart Our Business Startups Act of 2012, or JOBS Act, we may delay the adoption of certain accounting standards until such time as those standards apply to private companies. Other exemptions and reduced reporting requirements under the JOBS Act for EGCs include presentation of only two years of audited financial statements in a registration statement for an IPO, an exemption from the requirement to provide an auditor's report on internal controls over financial reporting pursuant to Section 404(b) of the Sarbanes-Oxley Act of 2002, an exemption from any requirement that may be adopted by the Public Company Accounting Oversight Board regarding mandatory audit firm rotation, and less extensive disclosure about our executive compensation arrangements.

In addition, the JOBS Act provides that an EGC can take advantage of an extended transition period for complying with new or revised accounting standards. This provision allows an EGC to delay the adoption of some accounting standards until those standards would otherwise apply to private companies. We have elected to use this extended transition period for complying with new or revised accounting standards that have different effective dates for public and private companies until the earlier of the date we (i) are no longer an emerging growth company or (ii) affirmatively and irrevocably opt out of the extended transition period provided in the JOBS Act. As a result, our consolidated financial statements may not be comparable to companies that comply with new or revised accounting pronouncements as of public company effective dates.

We may remain classified as an EGC until December 31, 2026, although if the market value of our common stock that is held by non-affiliates exceeds $700 million as of any June 30 before that time or if we have annual gross revenues of $1.07 billion or more in any fiscal year, we would cease to be an emerging growth company as of December 31 of the applicable year. We also would cease to be an EGC if we issue more than $1 billion of non-convertible debt over a three-year period.

Critical accounting policies and significant judgments

This management's discussion and analysis of our financial condition and results of operations is based on our consolidated financial statements, which we have prepared in accordance with U.S. generally accepted accounting principles. The preparation of these financial statements and related disclosures requires us to make estimates, judgments and assumptions that affect the reported amounts of assets, liabilities, and expenses and the disclosure of contingent assets and liabilities in our consolidated financial statements. We base our estimates on historical experience, known trends and events and various other factors that we believe are reasonable under the circumstances, the results of which form the basis for making judgments about the carrying values of assets and liabilities that are not readily apparent from other sources. We evaluate our estimates and assumptions on an ongoing basis. Our actual results may differ from these estimates under different assumptions or conditions.

During the three and six months ended June 30, 2022, there were no material changes to our critical accounting policies from those described in our Annual Report on Form 10-K filed with the SEC on March 14, 2022.

Recently issued accounting pronouncements

See Note 2, "Summary of significant accounting policies - Recently issued accounting pronouncements" to our consolidated financial statements included in our Annual Report on Form 10-K filed with the SEC on March 14, 2022.



                                       32

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

© Edgar Online, source Glimpses