UNITED STATES
SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549
_____________________
 
FORM 8-K
_____________________
 
CURRENT REPORT
 
Pursuant to Section 13 or 15(d) of the Securities Exchange Act of 1934
 
Date of Report (Date of earliest event Reported): June 30, 2023
 
Scholar Rock Holding Corporation
(Exact Name of Registrant as Specified in Charter)
 
Delaware
001-38501
82-3750435
(State or Other Jurisdiction of Incorporation)
(Commission File Number)
(I.R.S. Employer Identification Number)


301 Binney Street, 3rd Floor, Cambridge, MA 02142
(Address of Principal Executive Offices) (Zip Code)
 
(857) 259-3860
(Registrant’s telephone number, including area code)
 
(Former name or former address, if changed since last report)
 
Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions:
 
   Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425)
   Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12)
   Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b))
   Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c))
 
Securities registered pursuant to Section 12(b) of the Act:
 
Title of each class
Trading Symbol(s)
Name of each exchange on which registered
Common Stock, par value $0.001 per share
SRRK
The Nasdaq Global Select Market

 
Indicate by check mark whether the registrant is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (17 CFR §230.405) or Rule 12b-2 of the Securities Exchange Act of 1934 (17 CFR §240.12b-2). Emerging growth company
 
If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act. 


Item 7.01. Regulation FD Disclosure.

On June 30, 2023, Scholar Rock Holding Corporation (the “Company”) issued a press release announcing 36-month topline results from its TOPAZ Phase 2 clinical trial for apitegromab for patients with nonambulatory spinal muscular atrophy (“SMA”). A copy of the press release is attached hereto as Exhibit 99.1.

On June 30, 2023, Thomas O. Crawford, M.D., Professor of Neurology and Pediatrics at Johns Hopkins University and the lead principal investigator of the Company’s TOPAZ trial, presented at two podium presentations at the Cure SMA Research & Clinical Care Meeting (“Cure SMA”) and presented new data from the Company’s Phase 2 TOPAZ trial extension period evaluating patient outcomes after 36 months of treatment with apitegromab. A copy of the podium presentations at Cure SMA may be accessed by visiting the Publications & Posters section of the Company’s website at https://scholarrock.com/our-science/publications-posters/.

On July 12, 2023, the Company will host a conference call and webcast at 9:00 am ET to discuss the 36-month data from the TOPAZ Phase 2 clinical trial and apitegromab’s potential to advance the standard of care in SMA. A copy of the presentation slides to be used by the Company during the conference call and webcast is attached hereto as Exhibit 99.2. A live webcast of the conference call may be accessed by visiting the Investors & Media section of the Company’s website at http://investors.scholarrock.com.

The information in this report furnished pursuant to Item 7.01 and Exhibit 99.1 shall not be deemed “filed” for the purposes of Section 18 of the Securities Exchange Act of 1934, as amended (the “Exchange Act”), or otherwise subject to the liabilities of that section nor shall such information be deemed incorporated by reference in any filing under the Securities Act of 1933, as amended, or the Exchange Act, regardless of the general incorporation language of such filing, except as shall be expressly set forth by specific reference in such filing. It may only be incorporated by reference in another filing under the Exchange Act or the Securities Act of 1933, as amended, if such subsequent filing specifically references the information furnished pursuant to Item 7.01 and Exhibit 99.1 of this report.

Item 8.01. Other Events.

TOPAZ Phase 2 Clinical Trial Update

On June 30, 2023, the Company announced new data from the Phase 2 TOPAZ trial extension period evaluating patient outcomes at 36-months of treatment with apitegromab. These data showed that continued treatment with apitegromab over the extended treatment period was associated with substantial and sustained improvement in motor function, as well as improvements in patient-reported outcome measures in patients with nonambulatory Types 2 and 3 SMA receiving survival motor neuron (SMN)-targeted therapy.

Nonambulatory patients (ages 2-21) experienced substantial and sustained gains in Hammersmith Functional Motor Scale-Expanded (HFMSE) and Revised Upper Limb Module (RULM) scores over the 36-month extended treatment period from baseline:

 
12-Month Data
24-Month Data
36-Month Data
Mean Change from Baseline in HFMSE (95% Confidence Interval)
3.6 points
(1.2, 6.0)
N=32
4.2 points
(1.9, 6.6)
N=29
4.0 points
(1.0, 6.9)
N=28
Mean Change from Baseline in
RULM (95% Confidence Interval)
1.3 points
(0.2, 2.3)
N=31
2.3 points
(1.2, 3.3)
N=31
2.4 points
(1.1, 3.7)
N=27

For the 36-month data, an observed case analysis was conducted, which pooled data for all nonambulatory patients (including those patients on 20 mg/kg of apitegromab for the full duration of the trial, and those who switched from 2 mg/kg to 20 mg/kg at various time intervals in year 2) and was based upon the available data. These analyses exclude data for patients post scoliosis surgery.

Nonambulatory patients (ages 2-21) had improvements in PEDI-CAT (measure of activities of daily living) and PROMIS-Fatigue (a patient-reported outcome tool measuring fatigue) that were consistent and sustained at 36 months. The mean change in PEDI-CAT daily activity domain from baseline at 36 months was 2.2 (95% CI: –0.1, 4.5; N=17), indicating an improvement in the ability to perform daily activities. The mean change in PROMIS-Fatigue from baseline at 36 months was –4.6 (95% CI: –8.7, –0.5; N=14), indicating a decline in fatigue. These improvements in PEDI-CAT and PROMIS-Fatigue were generally consistent with improvements in motor function across the 36 months of the study period.

Treatment-emergent adverse events (TEAEs) at 36 months were consistent with previous reports at 12 and 24 months, with no new findings after an aggregate of 198 patient-years of exposure. TEAEs were mostly mild to moderate in severity, and generally consistent with the underlying patient population and background therapy. The five most common TEAEs were headache, pyrexia, COVID-19, nasopharyngitis, and upper respiratory tract infection. No deaths or suspected unexpected serious adverse reactions or hypersensitivity reactions were observed with apitegromab at 36 months. A total of 21 serious TEAEs were reported over the 36-month treatment period. No patients displayed positive titers for apitegromab antibodies (ADA).

More than 90 percent of nonambulatory patients remained on treatment in the extension study.


Item 9.01. Financial Statements and Exhibits.
 
(d) Exhibits
 
Exhibit
No.
 
Description
 
 
 
 
 
104
 
Cover Page Interactive Data File (embedded within the Inline XBRL document)


SIGNATURE
 
Pursuant to the requirements of the Securities Exchange Act of 1934, the registrant has duly caused this report to be signed on its behalf by the undersigned hereunto duly authorized.

 
Scholar Rock Holding Corporation
 
 
 
 
 
 
Date: June 30, 2023 By:
/s/ Junlin Ho
 
 
Junlin Ho
 
 
General Counsel and Corporate Secretary
 
Exhibit 99.1

New 36-Month Apitegromab Extension Data Reinforce Long-Term Substantial and Sustained Improvement of Motor Function in Phase 2 TOPAZ Trial Patients with Nonambulatory Spinal Muscular Atrophy

- Improvements in patient-reported outcomes consistent with gains in motor function scores

- Safety profile at 36 months consistent with previous reports with no new safety findings; more than 90 percent of nonambulatory patients remained on study

- Enrollment progressing in pivotal Phase 3 SAPPHIRE registrational trial, anticipated completion in Q3 2023

- Scholar Rock to host virtual investor event on July 12 at 9:00 AM EST to discuss the current SMA treatment landscape and apitegromab’s potential to advance the standard of care

CAMBRIDGE, Mass.--(BUSINESS WIRE)--June 29, 2023--Scholar Rock (NASDAQ: SRRK), a Phase 3, clinical-stage biopharmaceutical company focused on the treatment of serious diseases in which protein growth factors play a fundamental role, today announced new data from the Phase 2 TOPAZ trial extension period evaluating patient outcomes at 36 months of treatment with apitegromab. These data showed that continued treatment with apitegromab over the extended treatment period was associated with substantial and sustained improvement in motor function, as well as improvements in patient-reported outcome measures in patients with nonambulatory Types 2 and 3 spinal muscular atrophy (SMA) receiving survival motor neuron (SMN)-targeted therapy. Detailed results were presented today by Thomas Crawford, M.D., of Johns Hopkins Medicine, and the lead principal investigator of the TOPAZ trial, during two podium presentations at the Cure SMA Research & Clinical Care Meeting in Orlando, Florida.

“These promising long-term data highlight the therapeutic potential of muscle-targeted therapies, such as apitegromab, to help those with SMA address persistent weakness,” said Dr. Crawford. “While SMN-targeted therapies play an important role in preventing further loss of motor neurons, many people still experience persistent or progressive symptoms due to preexisting motor neuron degeneration. Incorporating a muscle-targeted therapy with apitegromab’s clinical profile into the treatment paradigm could allow patients to sustain or potentially achieve new gains in motor functioning.”

“We are excited to share these new Phase 2 data that support apitegromab’s long-term durability of effect and consistent tolerability and safety profile. The results further strengthen our confidence in apitegromab’s therapeutic potential for patients with SMA, as well as validate Scholar Rock’s unique approach to selectively inhibiting the pro and latent forms of myostatin,” said Jay Backstrom, M.D., MPH, President and Chief Executive Officer of Scholar Rock. “In addition to the sustained benefit observed with consistent HFMSE scores, we saw continued improvement in RULM scores, and reductions in fatigue as reported by patients—all of which can be important factors in performing activities of daily living. We remain committed to advancing the standard of care for people with SMA, and we look forward to sharing updates on our pivotal Phase 3 SAPPHIRE trial, which we anticipate will complete enrollment in the third quarter of 2023.”


Substantial and Sustained Gains in Motor Function Observed Over the Extended Treatment Period: Nonambulatory patients (ages 2-21) experienced substantial and sustained gains in Hammersmith Functional Motor Scale-Expanded (HFMSE) and Revised Upper Limb Module (RULM) scores over the 36-month extended treatment period from baseline:

 

12-Month Data

24-Month Data

36-Month Data

Mean Change from Baseline in HFMSE (95% Confidence Interval)

3.6 points

(1.2, 6.0)

N=32

4.2 points

(1.9, 6.6)

N=29

4.0 points

(1.0, 6.9)

N=28

Mean Change from Baseline in RULM (95% Confidence Interval)

1.3 points

(0.2, 2.3)

N=31

2.3 points

(1.2, 3.3)

N=31

2.4 points

(1.1, 3.7)

N=27

For the 36-month data, an observed case analysis was conducted, which pooled data for all nonambulatory patients (including those patients on 20 mg/kg of apitegromab for the full duration of the trial, and those who switched from 2 mg/kg to 20 mg/kg at various time intervals in year 2) and was based upon the available data. These analyses exclude data for patients post scoliosis surgery.

Improvement in Patient-Reported Outcomes Consistent with Improvements in Motor Function: Nonambulatory patients (ages 2-21) had improvements in PEDI-CAT (measure of activities of daily living) and PROMIS-Fatigue (a patient-reported outcome tool measuring fatigue) that were consistent and sustained at 36 months. The mean change in PEDI-CAT daily activity domain from baseline at 36 months was 2.2 (95% CI: –0.1, 4.5; N=17), indicating an improvement in the ability to perform daily activities. The mean change in PROMIS-Fatigue from baseline at 36 months was –4.6 (95% CI: –8.7, –0.5; N=14), indicating a decline in fatigue. These improvements in PEDI-CAT and PROMIS-Fatigue were generally consistent with improvements in motor function across the 36 months of the study period.

Consistent Safety Data: Treatment-emergent adverse events (TEAEs) at 36 months were consistent with previous reports at 12 and 24 months, with no new findings after an aggregate of 198 patient-years of exposure. TEAEs were mostly mild-to-moderate in severity, and generally consistent with the underlying patient population and background therapy. The five most common TEAEs were headache, pyrexia, COVID-19, nasopharyngitis, and upper respiratory tract infection. No deaths or suspected unexpected serious adverse reactions or hypersensitivity reactions were observed with apitegromab at 36 months. A total of 21 serious TEAEs were reported over the 36-month treatment period. No patients displayed positive titers for apitegromab antibodies (ADA).

More than 90 percent of nonambulatory patients remained on treatment in the extension study.

Details of the podium presentations at SMA Research & Clinical Care Meeting are as follows:

Title: Effect of apitegromab on PEDI-CAT and PROMIS-Fatigue questionnaire at 36 months in patients with Type 2 and nonambulatory Type 3 spinal muscular atrophy
Presentation type: Oral presentation
Presenter: Thomas O. Crawford, M.D., Professor of Neurology and Pediatrics, Johns Hopkins University
Date and time: Friday, June 30, 2023, 10:40 AM EST
Location: Disney Swan and Dolphin Hotels, Orlando, FL


Title: Effect of apitegromab on motor function at 36 months in patients with Type 2 and nonambulatory Type 3 spinal muscular atrophy
Presentation type: Oral presentation
Presenter: Thomas O. Crawford, M.D., Professor of Neurology and Pediatrics, Johns Hopkins University
Date and time: Friday, June 30, 2023, 11:00 AM EST
Location: Disney Swan and Dolphin Hotels, Orlando, Florida
For conference information, visit https://www.researchandclinicalcaremeeting.com/

The presentations will be made available in the Publications & Posters section of Scholar Rock’s website following the presentation.

Conference Call/Webcast:

Scholar Rock will host a virtual investor event on July 12 at 9:00 AM EST to discuss the current SMA treatment landscape and apitegromab’s potential to advance the standard of care for patients with nonambulatory Types 2 and 3 SMA. Click here to register and listen to the webcast. A link to the webcast of this event is also available on the Investors & Media section of the Scholar Rock website at http://investors.scholarrock.com.

An archived replay of the webcast will be available on Scholar Rock’s website at: https://scholarrock.com/ for approximately 90 days following the presentation.

About the Phase 2 TOPAZ Trial

The TOPAZ trial is an ongoing proof-of-concept, open-label Phase 2 trial evaluating the safety and efficacy of apitegromab in patients with Types 2 and 3 SMA. In the main treatment period, patients were dosed intravenously every four weeks as monotherapy or with nusinersen, an approved SMN-targeted therapy. The trial enrolled 58 patients in the U.S. and Europe. The primary efficacy endpoints were mean change from baseline in Revised Hammersmith Scale (RHS) score at 12 months for the ambulatory population (Cohort 1), and mean change from baseline in HFMSE score at 12 months for the nonambulatory population (Cohorts 2 and 3). The trial also includes multiple 12-month extension periods designed to evaluate longer-term patient outcomes.

About the Phase 3 SAPPHIRE Trial

SAPPHIRE is an ongoing randomized, double-blind, placebo-controlled, phase 3 clinical trial evaluating the safety and efficacy of apitegromab in nonambulatory patients with Types 2 and 3 SMA who are receiving SMN therapy (either nusinersen or risdiplam). Approximately 156 patients aged 2-12 years old are anticipated to be enrolled in the main efficacy population. These patients will be randomized 1:1:1 to receive for 12 months either apitegromab 10 mg/kg, apitegromab 20 mg/kg, or placebo by intravenous (IV) infusion every 4 weeks. An exploratory population of approximately 48 patients aged 13-21 years old will also separately be evaluated. These patients will be randomized 2:1 to receive either apitegromab 20 mg/kg or placebo. For more information about SAPPHIRE, visit www.clinicaltrials.gov.


About Apitegromab

Apitegromab is an investigational fully human monoclonal antibody inhibiting myostatin activation by selectively binding the pro- and latent forms of myostatin in the skeletal muscle. It is the first muscle-targeted treatment candidate to demonstrate clinical proof of concept in spinal muscular atrophy (SMA). Myostatin, a member of the TGFβ superfamily of growth factors, is expressed primarily by skeletal muscle cells, and the absence of its gene is associated with an increase in muscle mass and strength in multiple animal species, including humans. Scholar Rock believes that our highly selective targeting of pro- and latent forms of myostatin with apitegromab may lead to a clinically meaningful improvement in motor function in patients with SMA. The U.S. Food and Drug Administration (FDA) has granted Fast Track, Orphan Drug and Rare Pediatric Disease designations, and the European Medicines Agency (EMA) has granted Priority Medicines (PRIME) and Orphan Medicinal Product designations, to apitegromab for the treatment of SMA. The efficacy and safety of apitegromab have not been established and apitegromab has not been approved for any use by the FDA or any other regulatory agency.

About SMA

Spinal muscular atrophy (SMA) is a rare, genetic neuromuscular disease that afflicts an estimated 30,000 to 35,000 people in the United States and Europe. The disease is characterized by the loss of motor neurons, atrophy of the voluntary muscles of the limbs and trunk, and progressive muscle weakness. While there has been progress in the development of therapeutics that address the loss of motor neurons, there continues to be a high unmet need for therapies that directly address the progressive muscle weakness that leads to loss of motor function in SMA.

About Scholar Rock

Scholar Rock is a clinical-stage biopharmaceutical company focused on the discovery and development of innovative medicines for the treatment of serious diseases in which signaling by protein growth factors plays a fundamental role. Scholar Rock is creating a pipeline of novel product candidates with the potential to transform the lives of patients suffering from a wide range of serious diseases, including neuromuscular disorders, cancer, and fibrosis. Scholar Rock’s approach to targeting the molecular mechanisms of growth factor activation enabled it to develop a proprietary platform for the discovery and development of monoclonal antibodies that locally and selectively target these signaling proteins at the cellular level. By developing product candidates that act in the disease microenvironment, the Company intends to avoid the historical challenges associated with inhibiting growth factors for therapeutic effect. Scholar Rock believes its focus on biologically validated growth factors may facilitate a more efficient development path. For more information, please visit www.ScholarRock.com or follow Scholar Rock on Twitter (@ScholarRock) and LinkedIn (https://www.linkedin.com/company/scholar-rock/).


Availability of Other Information About Scholar Rock

Investors and others should note that we communicate with our investors and the public using our company website www.scholarrock.com, including, but not limited to, company disclosures, investor presentations and FAQs, Securities and Exchange Commission filings, press releases, public conference call transcripts and webcast transcripts, as well as on Twitter and LinkedIn. The information that we post on our website or on Twitter or LinkedIn could be deemed to be material information. As a result, we encourage investors, the media and others interested to review the information that we post there on a regular basis. The contents of our website or social media shall not be deemed incorporated by reference in any filing under the Securities Act of 1933, as amended.

Scholar Rock® is a registered trademark of Scholar Rock, Inc.

Forward-Looking Statements

This press release contains "forward-looking statements" within the meaning of the Private Securities Litigation Reform Act of 1995, including, but not limited to, statements regarding Scholar Rock’s future expectations, plans and prospects, including without limitation, Scholar Rock’s expectations regarding its growth, strategy, progress and timing of its clinical trials for apitegromab, and other product candidates and indication selection and development timing, the ability of any product candidate to perform in humans in a manner consistent with earlier nonclinical, preclinical or clinical trial data, and the potential of its product candidates and proprietary platform. The use of words such as “may,” “might,” “could,” “will,” “should,” “expect,” “plan,” “anticipate,” “believe,” “estimate,” “project,” “intend,” “future,” “potential,” or “continue,” and other similar expressions are intended to identify such forward-looking statements. All such forward-looking statements are based on management's current expectations of future events and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in or implied by such forward-looking statements. These risks and uncertainties include, without limitation, that preclinical and clinical data, including the results from the Phase 2 clinical trial of apitegromab, and are not predictive of, may be inconsistent with, or more favorable than, data generated from future clinical trials of the same product candidates, including, without limitation, the Phase 3 clinical trial of apitegromab in SMA , Scholar Rock’s ability to provide the financial support, resources and expertise necessary to identify and develop product candidates on the expected timeline, the data generated from Scholar Rock’s nonclinical and preclinical studies and clinical trials, information provided or decisions made by regulatory authorities, competition from third parties that are developing products for similar uses, Scholar Rock’s ability to obtain, maintain and protect its intellectual property, Scholar Rock’s dependence on third parties for development and manufacture of product candidates including, without limitation, to supply any clinical trials, Scholar Rock’s ability to manage expenses and to obtain additional funding when needed to support its business activities and establish and maintain strategic business alliances and new business initiatives, and the impacts of public health pandemics on business operations and expectations, as well as those risks more fully discussed in the section entitled "Risk Factors" in Scholar Rock’s Quarterly Report on Form 10-Q for the quarter ended March 31, 2023, as well as discussions of potential risks, uncertainties, and other important factors in Scholar Rock’s subsequent filings with the Securities and Exchange Commission. Any forward-looking statements represent Scholar Rock’s views only as of today and should not be relied upon as representing its views as of any subsequent date. All information in this press release is as of the date of the release, and Scholar Rock undertakes no duty to update this information unless required by law.

Contacts

Scholar Rock:

Investors
Rushmie Nofsinger
Scholar Rock
rnofsinger@scholarrock.com
ir@scholarrock.com
857-259-5573

Media
Teagan White
Finn Partners
teagan.white@finnpartners.com
media@scholarrock.com
650-766-3955

Exhibit 99.2

 Deep InsightsAdvancingImpactful Medicines  Company Overview | June 2023  
 

 © Scholar Rock, Inc. All rights reserved. 2023.   Forward-Looking Statements  Various statements in this presentation concerning the future expectations, plans and prospects of Scholar Rock, Inc. (“Scholar Rock”), including without limitation, Scholar Rock’s expectations regarding its strategy, its product candidate selection and development timing, including timing for the initiation of and reporting results from its clinical trials for apitegromab, SRK-181, and other product candidates and indication selection and development timing, its cash runway, the ability of any product candidate to perform in humans in a manner consistent with earlier nonclinical, preclinical or clinical trial data, and the potential of its product candidates and proprietary platform. The use of words such as “may,” “could,” “might,” “will,” “should,” “expect,” “plan,” “anticipate,” “believe,” “estimate,” “project,” “intend,” “future,” “potential,” or “continue,” and other similar expressions are intended to identify such forward-looking statements for the purposes of the safe harbor provisions under The Private Securities Litigation Reform Act of 1995. All such forward-looking statements are based on management's current expectations of future events and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in or implied by such forward-looking statements. These risks and uncertainties include, without limitation, that preclinical and clinical data, including the results from the Phase 2 trial of apitegromab or Part A of the Phase 1 trial of SRK-181, are not predictive of, may be inconsistent with, or more favorable than, data generated from future clinical trials of the same product candidate, including the Phase 3 clinical trial of apitegromab in SMA and Part B of the Phase 1 clinical trial of SRK-181, respectively, Scholar Rock’s ability to provide the financial support, resources and expertise necessary to identify and develop product candidates on the expected timeline, the data generated from Scholar Rock’s nonclinical and preclinical studies and clinical trials, information provided or decisions made by regulatory authorities, competition from third parties that are developing products for similar uses, Scholar Rock’s ability to obtain, maintain and protect its intellectual property, the success of Scholar Rock’s current and potential future collaborations, Scholar Rock’s dependence on third parties for development and manufacture of product candidates including, without limitation, to supply any clinical trials, Scholar Rock’s ability to manage expenses and to obtain additional funding when needed to support its business activities and establish and maintain strategic business alliances and new business initiatives, and the impacts of current macroeconomic and geopolitical events, hostilities in Ukraine, increasing rates of inflation and rising interest rates, on business operations and expectations, as well as those risks more fully discussed in the section entitled "Risk Factors" in Scholar Rock’s Quarterly Report on Form 10-Q for the quarter ended March 31, 2023, as well as discussions of potential risks, uncertainties, and other important factors in Scholar Rock’s subsequent filings with the Securities and Exchange Commission.  Any forward-looking statements represent Scholar Rock’s views only as of today and should not be relied upon as representing its views as of any subsequent date. All information in this press release is as of the date of the release, and Scholar Rock undertakes no duty to update this information unless required by law. Apitegromab and SRK-181 are investigational drug candidates under evaluation.  Apitegromab and SRK-181 have not been approved for any use by the FDA or any other regulatory agency and the safety and efficacy of apitegromab and SRK-181 have not been established. 
 

 Scholar Rock: Transforming Patient Lives, Addressing High Unmet Medical Need  3  Global leader in TGFβ superfamily biology   Targeting the latent forms of growth factors   Exquisite selectivity to deliver differentiated therapies   Revolutionary Platform  Rich preclinical pipeline focused on high unmet patient needs   Phase 3 SAPPHIRE study underway, enrollment completion expected in Q3 2023; data readoutexpected in 2024   Phase 1 proof-of-concept DRAGON study underway in immuno-oncology   Neuromuscular and Beyond  Compelling proof-of-concept TOPAZ data informed Phase 3 SAPPHIRE study design  Seasoned leadership team with track record of clinical and commercial success   Anticipated cash runway into 2025  Positioned  for Success  Commercial planning underway for apitegromab (SMA) in US and Europe   Broad platform, including promising early-stage assets, provides opportunities to advance alone or in partnership  Strategic Optionality  
 

 Revolutionary Approach to Regulating TGFβ Superfamily Implicated in Devastating Diseases  4  Scholar Rock’s R&D Platform  Transforming Medical Practice  Selectively target the latent form of growth factors in the microenvironment of cells and tissues with uniquely designed antibodies  Overcome the challenges that plague traditional approaches that target the “mature” growth factor or its receptors, which are difficult to differentiate and lead to unintended negative effects  Scholar Rock’s TargetLatent Growth Factor  Traditional Target“mature” growth factor  TGFβ Superfamily: Highly Sought-After Targets   Dysregulation plays a role in devastating diseases that have a high unmet need, including:  Neuromuscular disorders    Fibrosis  Oncology  Recognized by the industry as important targets given their fundamental roles in regulating a variety of biological processes 
 

 DISCOVERY/PRECLINICAL  PHASE 1  PHASE 2  PHASE 3  ANTICIPATED 2023 MILESTONES  SPINAL MUSCULAR ATROPHYApitegromab (selective anti- pro and latent myostatin)  36-month TOPAZ data  SAPPHIRE: last patient enrolled expected in Q3  IMMUNO-ONCOLOGYSRK-181 (selective context-independent, anti-latent TGFβ-1)  Rolling clinical data updates  ANEMIASelective anti-RGMc  IND-enabling studies  FIBROSIS  Selective context-dependent (LTBP1 & LTBP3) anti-latent TGFβ-1   IND-enabling studies  Robust Pipeline of Novel Product Candidates   5  Potential to transform the lives of people living with awide range of serious diseases, including neuromuscular disorders, oncology, and fibrosis  LTBP3=latent transforming growth factor beta binding protein 3; LTBP1=Latent Transforming Growth Factor Beta Binding Protein 1; RGM=Repulsive guidance molecule; TGFβ-1 =Transforming Growth Factor Beta-1 
 

 Leadership Team: Experienced in Drug Development and Commercialization  6  Jay Backstrom, MD, MPH  President & CEO  30 years of clinical R&D experience, leading multiple successful regulatory approvals  Ted Myles, MBA  Chief Operating Officer & CFO  25 years of progressive experience in clinical and commercial-stage companies   25 years of experience leading HR, culture transformation, leadership development, DEI, and talent management  Caryn Parlavecchio  Chief Human Resources Officer  15 years of experience leading and advising life sciences companies in areas of legal and compliance   Junlin Ho, JD  General Counsel &Corporate Secretary  Mo Qatanani, PhD  SVP, Research  15 years of industry experience on the strategic and operational sides of research & development  Tracey Sacco  Chief Commercial Officer  20 years of commercial leadership experience, including product launch and global commercial strategy  Jing Marantz, MD, PhD  Chief Medical Officer   20 years of development and medical leadership experience across neurology, hematology/oncology, and rare diseases 
 

 Apitegromab: The Next Potential Transformative Therapy for Patients with Spinal Muscular Atrophy (SMA) 
 

 Apitegromab: Transformative Potential to Change the Standard of Care  8  Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy (SMA). Apitegromab has not been approved for any use by the FDA or any other regulatory agency, and its safety and efficacy have not been established.  Phase 2 TOPAZ TrialDemonstrated substantial and sustained functional improvements in Type 2 and nonambulatory Type 3 SMA patients  Apitegromab at a Glance   First and only muscle-targeted investigational treatment to demonstrate clinical proof-of-concept in SMA  Phase 3 SAPPHIRE TrialRegistrational trial with topline 12-month data readout expected in 2024  ONYX Open-Label Extension Study Evaluating the long-term safety and efficacy of apitegromab in patients who have completed TOPAZ or SAPPHIRE  
 

 There is further potential to regain vital muscle function by also addressing the progressive muscle atrophy and associated weakness of SMA   SMN therapies slow further degeneration of motor neurons1  Hallmarks of SMAProgressive Motor Neuron Loss and Muscle Atrophy Leading to Progressive Muscle Weakness   9  SMA, spinal muscular atrophy; SMN, survival motor neuron.1. Hua Y, et al. Nature. 2011;478(7367):123-6. 2. Figure adapted from: SMA Foundation Overview. http://www.smafoundation.org/wp-content/uploads/2012/03/SMA-Overview.pdf.; Accessed April 18, 2021.   ...but do not directly address muscle atrophy  Muscle fiber atrophy  Motor neuron  degeneration2 
 

 10  *TOPAZ Phase 2 trial evaluated patients with Type 2 and 3 SMA (did not include Type 1)  ** 2/3 of overall patient population has type 2 or 3 SMA, including ambulatory and nonambulatory patients. Percentages reflected here do not add up to 100% because the prevalences of Types 0, 4, and unknown are excluded.  Percentages represent percent of prevalent patients.  1. Lally et al, Orphanet Journal of Rare Diseases, 2017; 2. SMA Europe. SMATracker. About SMA. Accessed January 24, 2022. https://smatracker.eu/what-is-spinal-muscular-atrophy; 3. National Organization for Rare Disorders. Spinal muscular atrophy. Accessed January 24, 2022. https://rarediseases.org/rare-diseases/spinal-muscular-atrophy/. 4. Cure SMA. Care Series Booklet. Accessed September 19, 2021. 2020. https://www.curesma.org/wpcontent/uploads/2020/08/08262020_Understanding_SMA_vWeb.pdf. 5. Cure SMA. State of SMA 2022. May 31, 2022. https://www.curesma.org/wp-content/uploads/2022/06/9042022_State-of-SMA_vweb.pdf Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Severe, progressive disabilities; unable to walk independently  Significant, progressive motor function impairment; many lose ambulation  Infant onset; unable to sit up independently  Type 3  25%  Type 2  39%  Type 1  25%  TOPAZ* 12-month results showed transformative potential in nonambulatory Types 2 and 3 patients  ~2/3 of overall   patient population5**  GLOBAL DISEASE:  30,000-35,000 affected in US and Europe1, 2, 3,4  Cure SMA. State of SMA 2022. May 31, 2022  ALMOST 70% OF INDIVIDUALS WITH SMA IN THE U.S. HAVE RECEIVED AN FDA-APPROVED TREATMENT  
 

 Motor Function With SMN Therapies as Assessed by HFMSE HFMSE appears to Plateau After Initial Gains  1. Mercuri E, et al. Presented at: World Muscle Society Congress 2020, P. 257  2. Oskoui M, et al. Presented at: 2021 Muscular Dystrophy Association Clinical & Scientific Conference; March 15-18, 2021. Poster 80.   HFMSE, Hammersmith Functional Motor Scale–Expanded; SE, standard error.  *MFM was primary efficacy endpoint of SUNFISH. HFMSE was a secondary endpoint. This third-party information is provided for background only and is not intended to convey or imply a comparison to the TOPAZ clinical trial results.   Overall population age 2-25   Change in HFMSE* Over Four Years with Risdiplam2   Risdiplam  n=  120  120  119  117  109  106  89  99  101  97  Placebo  n=  60  60  58  58  Risdiplam  Placebo (months 0-12)  Mean (+/-SE) Change in HFMSE  Total Score From Baseline*  0  30  48  6  12  18  24  36  42  Visit, months  11  Change in HFMSE Over Four Years with Nusinersen1   Overall population age 2-12   Mean (+SE) Change in HFMSE  Total Score From Baseline  Analysis Visit, days  Initial Treatment  CHERISH  Chronic Maintenance Phase  SHINE  1  92  169  253  350  450  690  930  1170  1410  1650  Nusinersen  n=  84  82  84  84  83  76   83   83   79   61   20  Placebo  n=  42  41  41  42  42  39  Nusinersen in CHERISH and SHINE  Sham control in CHERISH 
 

 Patients and caregivers want new therapies to address the following unmet needs2* :   Improving and Sustaining Muscle Function Remains an Unmet Need  12  HFMSE=Hammersmith Functional Motor Scale-Expanded   *Percentages represent percent of patients who named these unmet needs when asked “What are your most significant current unmet needs that you hope new therapies would address”?  1. Mercuri E et al.; N Engl J Med 2018; 378:625-635; DOI: 10.1056/NEJMoa1710504; cherish trial results. 2. 2022 Community Update Survey, Cure SMA.This third-party information is provided for background only and is not intended to convey or imply a comparison to the TOPAZ clinical trial results.  Mean improvement in HFMSE experienced by patients with  nonambulatory Types 2/3 SMA in nusinersen Phase 3 CHERISH trial1  3.9-point increase in HFMSE from nusinersen(4.9 point increase relative to sham control)  HFMSE Score at Month 15  Total Possible HFMSE Score of 66  Unmet need remains substantial   Increase muscle strength   Improve daily activities   Stabilize or gain new motor function   Reduce fatigue   97%  92%  89%  83% 
 

 Apitegromab Offers Significant Potential to Address Unmet Needs   13  * Based on Animal Model Data; 1. Long KK, et al. Hum Mol Genet. 2019;28(7):1077-1088; 2. Pirruccello-Straub M, et al. Sci Reports. 2018;8(1):2292. doi:10.1038/s41598-018-20524-9 3. Figure adapted from: SMA Foundation Overview. http://www.smafoundation.org/wp-content/uploads/2012/03/SMA-Overview.pdf.; Accessed April 18, 2021. For illustrative purposes only  Apitegromab is a  MUSCLE-TARGETED APPROACH  designed to improve motor function*1,2  Myostatin is a negative modulator of muscle growth   Strong preclinical evidence indicates upstream targeting of structurally differentiated pro- and latent myostatin avoids undesirable off-target effects  Apitegromab  Muscle fiber atrophy  Apitegromab selectively inhibits myostatin and has the potential to build muscle and strength to improve certain patient outcomes  Motor neuron  degeneration3 
 

 Phase 2 TOPAZ Trial: Safety and Efficacy Data from First Muscle-Targeted Treatment Candidate in SMA 
 

 Phase 2 Trial Design1,2  15  Ambulatory Patients (Revised Hammersmith Scale)  Safety  RHS mean change from baseline at 12 months  Ages 5-21  Type 3 SMA  Open-label, single-arm  Apitegromab 20 mg/kg IV q4w  Receiving nusinersen  Apitegromab monotherapy  Nonambulatory Patients (Hammersmith Functional Motor Scale Expanded)  Safety  HFMSE mean change from baseline at 12 months  COHORT 2 N=15  Ages 5-21  Type 2 or Type 3 SMA  Apitegromab 20 mg/kg IV q4w  Open-label, single-arm  Receiving nusinersen  (initiated ≥ 5 years of age)  Safety  HFMSE mean change from baseline at 12 months  COHORT 3 N=20  Ages ≥ 2  Type 2 SMA   Apitegromab 2 mg/kg IV q4w  Apitegromab 20 mg/kg IV q4w  R1:1  Double-blind, randomized (1:1)  Receiving nusinersen   (initiated< 5 years of age)  TREATMENT (12 months)  PRIMARY ENDPOINTS  All SMA Types 2/3, cohorts defined by age and present ambulatory status at time of enrollment. HFMSE, Hammersmith Functional Motor Scale Expanded; IV, intravenous; q4w, every 4 weeks; SMA, spinal muscular atrophy; SMN, survival motor neuron.   1. Place A, et al. Eu J Neurol. 2021;28(Suppl1):207-334 (EPR-184). 2. Crawford T, et al. TOPAZ Extension: 24-month Efficacy and Safety of Apitegromab in Patients With Later-onset SMA (Type 2 and Type 3 SMA). Presented at CureSMA Annual Conference; June 16-19, 2022.  COHORT 1 N=23 
 

 Motor Function Outcomes by HFMSE Over 36 Months Improvements Were Substantial and Sustained   16  For the 36-month evaluation, an observed case analysis was conducted, which pooled all the nonambulatory patients (Cohorts 2 and 3) and was based upon the available data for given timepoints. This analysis population included patients receiving either low dose (2 mg/kg) or high dose (20 mg/kg) apitegromab (inclusive of patients in Cohort 3 who switched from 2 mg/kg to 20 mg/kg in Year 2). This analysis excludes data post scoliosis surgery from seven patients. One patient did not conduct HFMSE at time of database lock for 24 months, however, this patient had an unscheduled HFMSE score one month prior to their scheduled visit. In the most recent analysis, this result was included in the 24-month analysis. Error bars represent SE. CI represents confidence interval. SMN Rx=SMN therapy. In the age 2-21 group, 18/28 patients achieved ≥ 1-pt gains, and 11/28 patients ≥ 3-pt gains at 36 months. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  n=  35  29  32  29  28  95% CI=  (1.4, 4.1)  (1.2, 6.0)  (1.9, 6.6)  (1.0, 6.9)  n=  29  23  26  23  23  95% CI=  (1.8, 5.0)  (1.8, 7.4)  (2.3, 8.0)  (1.3, 8.3)  Pooled Nonambulatory Patients   Mean Change from Baseline (+SE)  Baseline mean age=7.3 | Time on SMN Rx=24.1m  Age 2-21 YearsAll Doses (N=35)  Baseline mean age=5.5 | Time on SMN Rx=24.6m   Age 2-12 YearsAll Doses (N=29)  Mean Change from Baseline (+SE) 
 

 Motor Function Outcomes by RULM Over 36 Months Improvements Were Substantial and Sustained  17  For the 36-month evaluation, an observed case analysis was conducted, which pooled all the nonambulatory patients (Cohorts 2 and 3) and was based upon the available data for given timepoints. This analysis population included patients receiving either low dose (2 mg/kg) or high dose (20 mg/kg) apitegromab (inclusive of patients in Cohort 3 who switched from 2 mg/kg to 20 mg/kg in Year 2). This analysis excludes data post scoliosis surgery from seven patients. One patient did not conduct RULM at month 24, however, had an unscheduled RULM score one month prior to their scheduled visit. In the most recent analysis, this result was included in the 24-month analysis. Error bars represent standard error (SE). CI represents confidence interval. SMN Rx=SMN therapy. In the age 2-21 group, 15/27 patients achieved ≥ 1-pt gains, and 18/27 patients ≥ 3-pt gains at 36 months. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Baseline mean age=7.3 |Time on SMN Rx=24.1m   Baseline mean age=5.5 |Time on SMN Rx=24.6m   Pooled Nonambulatory Patients   Mean Change from Baseline (+SE)  Age 2-21 YearsAll Doses (N=35)  Age 2-12 YearsAll Doses (N=29)  n=  35  28  31  31  27  95% CI=  (-0.2, 1.4)  (0.2, 2.3)  (1.2, 3.3)  (1.1, 3.7)  n=  29  22  25  25  22  95% CI=  (-0.7, 1.1)  (0.1, 2.4)  (1.0, 3.5)  (1.4, 4.2) 
 

 Time (weeks)  No. of observations  Time (weeks)  No. of observations  Year 3  Year 2  Year 1  Pooled Nonambulatory Patients | Age 2 – 21 | All Doses | Over 36 Months Improvements in PRO Measures Were Consistent with Motor Function  IMPROVEMENT  PEDI-CAT Daily Activities  IMPROVEMENT  IMPROVEMENT  N = 35; Baseline mean age=7.3 |Time on SMN Rx=24.1m   HFMSE  Change from baseline (±SE)  IMPROVEMENT  Year 2  Year 3  Year 1  RULM  Year 3  Year 2  Year 1  PROMIS Fatigue (Proxy)  Change from baseline (±SE)  Year 2  Year 3  Year 1  34  34  34  28  27  29  31  32  32  31  27  27  27  35  35  34  29  28  29  32  32  32  29  27  28  27  31  29  30  21  23  25  25  22  27  23  16  17  15  25  23  22  19  19  22  16  19  18  19  16  14  14  HFMSE=Hammersmith Functional Motor Scale Expanded; OC=observed case; PEDI-CAT=Pediatric Evaluation of Disability Inventory Computer Adaptive Test; PROMIS=Patient Reported Outcome Measurement Information System; RULM=Revised upper limb module; SE=standard error of the mean. SMN Rx=SMN therapy. Data on File. Scholar Rock, Inc. Cambridge, MA. Data cutoff date as of March 13, 2023. The updated PEDI-CAT analysis included additional records (2 at 12 months and 1 at 24 months) that were not available at the time of previous analysis. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established. 
 

 Pooled Nonambulatory Patients New WHO Development Milestones Achieved Over 36 Months   19  Proportion of patients gaining new milestones in TOPAZ  Cohort 2: BL (0%),12m (20%), 24m (7%), 36m (0%)  Cohort 3 (all doses): BL (0%,) 12m (24%), 24m (26%), 36m (30%)  Cohort 3: Randomized to 20mg/kg dose: 12m (25%), 24m (33%), 36m (40%)  *Includes patients who crossed over from 2 mg/kg to 20mg/kg starting week 68 through week 104.   SMN Rx=SMN therapy. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Age (years)  Hands & kneescrawling  Standing with  assistance  Walking with  assistance  Standing alone  Walking  alone  SMN Rx (≥ age 5)  8     9     19  SMN Rx  (< age 5)  2*  4*     5*     2     2     4     5  WHO MILESTONE  X  X  X  X  X  X  X  X  X  X  X  X     X  X  X  X  X  X  X  X  X  X  X  X  X  X  X  Key Takeaways  Patients receiving nusinersen ≥ age 5 mostly maintained WHO milestones  Patients receiving nusinersen < age 5 improved overall: 6 out of 20 gained new milestones over 36 months  BL  12M  24M  36M  Able  X  Unable  No record 
 

 TOPAZ Safety Summary Over 36 Months  20  Treatment-Emergent Adverse Events (TEAEs)*  2 mg/kg dose   (N=10)  n (%)  20 mg/kg dose   (N=48)  n (%)  Total  (N=58)  n (%)  Any TEAE  10 (100)  46 (95.8)  56 (96.6)  Any serious TEAE  5 (50)  16 (33.3)  21 (36.2)  Any TEAE leading to study drug discontinuation  0  1 (2.1)  1 (1.7)  Any Grade 3 (severe) or higher TEAE  4 (40)  16 (33.3)  20 (34.5)  *Defined as AEs that start after the first dose of study drug or start prior to the administration of study drug and worsen in severity/grade or relationship to investigational medication after the administration of study drug. % = 100 x n/N; % at 12 month. AE, adverse event; TEAE, treatment emergent adverse events. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  TEAEs were consistent with previous reports with no new findings after 198 patient-years of exposure  Most frequently reported TEAEs*: headache (38%), pyrexia (38%), COVID-19 (36%), nasopharyngitis (36%), & upper respiratory tract infection (33%)  TEAEs were mostly mild to moderate in severity and generally consistent with the underlying patient population and nusinersen therapy  No deaths or suspected unexpected serious adverse reactions or hypersensitivity reactions to apitegromab were reported  No patients displayed positive titers for apitegromab antibodies (ADA) 
 

 Summary of TOPAZ Data  Substantial and Sustained Improvement Over 36 MONTHS  21  PRO=Patient Reported Outcome  Data to date has shown substantial clinical benefit that is dose-dependent  TOPAZ data suggest that apitegromab has the potential to transform care in SMA by directly addressing progressive muscle weakness  Benefit continued to improve or was sustained over 36 months  Consistency across functional scales and patient-reported outcomes  Well tolerated profile and low discontinuation rate supports durability of treatment  
 

 Sapphire Phase 3 Pivotal Trial 
 

 SAPPHIRE Phase 3 Design is Optimized by Insights from TOPAZ  23  Substantial HFMSE gains observed in the nonambulatory Type 2/3 SMA cohorts  Exploratory age 2-12 analysis in nonambulatory  Type 2/3 showed transformative potential  HFMSE gains substantial by 12 months of treatment  Dose response seen  (greater effect observed with 20 mg/kg over 2 mg/kg)  Study population: nonambulatory Type 2/3 SMA  Primary efficacy endpoint: HFMSE  Age 2-12 main efficacy population  12-month treatment duration  20 mg/kg apitegromab dose  Also evaluating 10 mg/kg arm (to explore potential that dose between 2 and 20 mg/kg may be comparable to 20 mg/kg)  TOPAZ Learnings  SAPPHIRE Design Elements  HFMSE=Hammersmith Functional Motor Scale Expanded  
 

 Ongoing SAPPHIRE Phase 3 Trial Overview  24  Randomized, double-blind, placebo-controlled, parallel arm design (n=204)  Enrolling patients who are on SMN-directed therapy (nusinersen or risdiplam)  Anticipate completing enrollment in 3Q 2023  TREATMENT (52 weeks)  Apitegromab (20 mg/kg IV q4w) + SMN-directed therapy  Apitegromab (10 mg/kg IV q4w) + SMN-directed therapy  Placebo (IV q4w) + SMN-directed therapy  SCREENING  MAIN POPULATION (n=156)  Ages 2-12  With nonambulatory Types 2 and 3 SMA  N=52  N=52  N=52  R  Stratified to ensure balanced allocation across the three arms:   Age at SMN therapy initiation(age < 5 vs age ≥ 5)   SMN therapy (nusinersen vs. risdiplam)  ENDPOINTS  Primary Efficacy:  Mean HFMSE change from baseline at 12 months   Additional Efficacy Measures:  RULM, WHO, other outcome measures   Safety, PK/PD, ADA  Additional Data Opportunities  Exploratory population (age 13-21), in patients using SMN therapy  Focused upon safety & exploratory efficacy (n=48; 2:1 randomization between apitegromab 20 mg/kg vs placebo)  Separate open-label extension study (after patients complete 12-month treatment period)  Focused upon safety & exploratory long-term efficacy  ClinicalTrials.gov Identifier: NCT05156320  HFMSE=Hammersmith Functional Motor Scale Expanded; RULM=Revised Upper Limb Module; R=randomization; SMA=spinal muscular atrophy; SMN=survival motor neuron. 
 

 Executing on the Promise: Apitegromab SMA Trials  25  SMA=Spinal Muscular Atrophy  *Subject to regulatory approval   WHERE WE AREPhase 3  12-Month  IN PROCESS  Long-term  EXTENSION  3Q 2023 enrollment  2024 data  2025 launch*   ANTICIPATED MILESTONES   WHERE WE WERE Phase 2  12-Month  COMPLETE  24-Month  EXTENSION  COMPLETE  36-Month  EXTENSION  COMPLETE   WHERE WE PLAN TO GO  Ambulatory patients  Under 2 Years of Age (including those treated with gene therapy) 
 

 26  Potential first muscle-targeted therapy in SMA   Robust body of data supports therapeutic potential   Differentiation  Strong pre-clinical evidence indicates upstream targeting of structurally differentiated latent myostatin avoids undesirable off-target effects   Phase 2 TOPAZ trial demonstrated the therapeutic potential of inhibiting the latent forms of growth factors  Strong  Scientific  Rationale  TOPAZ has demonstrated sustained motor function gains to date in patients with nonambulatory Types 2 and 3 SMA   Pivotal Phase 3 SAPPHIRE trial: enrollment completion expected in Q3 2023  FDA has granted Fast Track, Orphan Drug, and Rare Pediatric Disease designations  European Medicines Agency (EMA) has granted Priority Medicines (PRIME) and Orphan designations  Clear  Clinical  Pathway  SMN therapies prevent further degeneration of motor neurons but do not directly address muscle atrophy  Apitegromab is a muscle-targeted approach and has the potential to address this unmet medical need  Global SMA treatment market expected to grow in the next five years   High Unmet  Medical Need  & Significant  Commercial Opportunity  ApitegromabSummary 
 

 SRK-181: Potential Transformative Backbone for a New Era ofCancer Immunotherapy 
 

 Resistance to Checkpoint Inhibitor (CPI) Therapies Remains a Significant Clinical Challenge  28  1. Source: Gores, M. (2022). In the eye of the storm: PD-(L)1 inhibitors weathering turbulence [White paper].  IQVIA. https://www.iqvia.com/library/white-papers/in-the-eye-of-the-storm-pd-l-1-inhibitors-weathering-turbulence   2. Source: Carretero-Gonzalez et al. (2018) Oncotarget 9:8706-8715  Meta-analysis of twelve randomized trials with control arm or adequate safety profile (includes nivolumab, pembrolizumab, and atezolizumab)  Clinically derived rationale points to significant opportunity to increase checkpoint therapy responses by targeting TGFβ-1   7.9-10.41MILLION US patients on CPI therapies  First-lineCPI therapy  Second-line(or later) CPI therapy  DID NOT respond   63%  2  78%  2 
 

 Strong Scientific Rationale for the Role of TGFβ Inhibition in Immuno-Oncology  29  “Bristol Myers Squibb Enters Agreement to Acquire Forbius TGF-beta Program”  August 2022.  “Merck to Acquire Tilos Therapeutics: Merck Gains Portfolio of Investigational Antibodies Modulating TGFβ”  $773 million total potential deal value  June 2019.  Selective inhibition of TGFβ-1 activation overcomes primary resistance to checkpoint blockade therapy by altering tumor immune landscape  Science Translational Medicine, March 25, 2020.  Constance J. Martin, et al.  Vol 12, Issue 536. DOI: 10.1126/scitranslmed.aay8456  Nature (online), February 14, 2018.  TGFβ attenuates tumour response to PD-L1 blockade by contributing to exclusion of T cells  Sanjeev Mariathasan1*, Shannon J. Turley1*, Dorothee Nickles1*, Alessandra Castiglioni1, Kobe Yuen1, Yulei Wang1,  Edward E. Kadel III1, Hartmut Koeppen1, Jillian L. Astarita1, Rafael Cubas1, Suchit Jhunjhunwala1, Romain Banchereau1,  Yagai Yang1, Yinghui Guan1, Cecile Chalouni1, James Ziai1, Yasin Şenbabaoğlu1, Stephen Santoro1, Daniel Sheinson1,   Jeffrey Hung1, Jennifer M. Giltnane1, Andrew A. Pierce1, Kathryn Mesh1, Steve Lianoglou1, Johannes Riegler1,  Richard A. D. Carano1, Pontus Eriksson2, Mattias Höglund2, Loan Somarriba3, Daniel L. Halligan3, Michiel S. van der Heijden4,  Yohann Loriot5, Jonathan E. Rosenberg6, Lawrence Fong7, Ira Mellman1, Daniel S. Chen1, Marjorie Green1, Christina Derleth1,  Gregg D. Fine1, Priti S. Hegde1, Richard Bourgon1 & Thomas Powles8  Willy Hugo, Jesse M. Zaretsky, Lu Sun, Douglas B. Johnson, Antoni Ribas, Roger S. Lo  Volume 165, Issue 1, 24 March 2016, Pages 35-44  Genomic and Transcriptomic Features of Response  to Anti-PD-1 Therapy in Metastatic Melanoma  July 24, 2020: https://doi.org/10.1038/ s41571-020-0403-1  Nature Reviews , July 24, 2020  NATURE REVIEWS | CLINICAL ONCOLOGY  TGFβ: biology in cancer progression  and immunotherapy  Rik Derynck1,2,3, Shannon J. Turley4 and Rosemary J. Akhurst2,3  
 

 SRK-181: Unique Latent TGFβ-1 Selective Approach Designedto Overcome Checkpoint Inhibitor Resistance  30  Traditional Target“Mature” growth factor  1. Wakefield LM, Winokur TS, Hollands RS, Christopherson K, Levinson AD, Sporn MB. Recombinant latent transforming growth factor beta 1 has a longer plasma half-life in rats than active transforming growth factor beta 1, and a different tissue distribution. J Clin Invest. 1990 Dec;86(6):1976-84. doi: 10.1172/JCI114932. PMID: 2254455; PMCID: PMC329834.  SRK-181: Latent TGFβ-1 Inhibitor  Targets TGFβ-1  Potential to overcome CPI resistance  SRK-181 inhibits the TGFβ-1 implicated in check point inhibitor resistance  Selective to β-1 isoform  Highly selective to β-1 isoform vs. 2 and 3  Increases therapeutic window and potentially avoids toxicities associated with non-selective TGFβ inhibition  Other programs target multiple isoforms of TGFβ  Context-independent  Inhibits all sources of TGFβ-1  SRK-181 targets all TGFβ-1 sources (LRRC33, GARP and LTBP1 and 3)  Some programs only target one source  Targets the latent form of TGFβ-1   Selectively targeting the latent form shuts off the growth factor before activation   Increases opportunity to inhibit TGFβ-1  Most other programs target the mature form of TGFβ-1 
 

 SRK-181-mIgG1 + Anti-PD1 Overcomes Immune Exclusion  31  Anti-PD1/ SRK-181-mIgG1  Anti-PD1  Overcoming immune exclusion  Tumor micro-environment  Preclinical data published in Science Translational Medicine. Martin CJ, et al. Sci Transl Med. 2020 Mar 25;12(536):eaay8456. https://scholarrock.com/platform/publications.  Data from MBT-2 syngeneic tumor model. Dose 10mg/kg QW for 4 weeks.   Overcome Exclusion  SRK-181-mIgG1 combination therapy led to influx and amplification of cytotoxic CD8+ cells in preclinical bladder tumor model  Immune Exclusion 
 

 DRAGON Phase 1 POC Trial to Evaluate SRK-181’s Ability to Overcome Primary Resistance to Checkpoint Inhibitors  32  * A cohort of 2000 mg Q2W (n=3) was also evaluated.  **The clear cell RCC and HNSCC cohorts will also explore the effects of SRK-181 in patients with relapsed response after anti-PD-1 treatment.  1. NCT04291079 on www.clinicaltrials.gov.  PART B  SRK-181 + pembrolizumab;  non-responders to prior anti-PD-1  SRK-181 + pembrolizumab  Non-small cell lung cancer  SRK-181 + pembrolizumab  Urothelial carcinoma  SRK-181 + pembrolizumab  Cutaneous melanoma  SRK-181 + pembrolizumab  Clear cell renal cell carcinoma**   SRK-181 + pembrolizumab  Head and neck squamous cell** carcinoma  COHORT  TREATMENT  PART A  SRK-181 80 mg (n=1)  SRK-181 240 mg (n=1)  SRK-181 800 mg (n=3)  SRK-181 1600 mg (n=3)  SRK-181 2400 mg (n=3)  SRK-181 3000 mg (n=3)*  SRK-181   all-comers  A1  A2  SRK-181+anti-PD-(L)1; non-responders to prior anti-PD-(L)1  SRK-181 240 mg (n=3)  SRK-181 800 mg (n=3)  SRK-181 1600 mg (n=3)  SRK-181 2400 mg (n=3) 
 

 DRAGON Part A: Safety   PART A1 Monotherapy  Treatment-Emergent AEs Related to SRK-181, All Grades >10%   PART A2 Combination Treatment  Treatment-Emergent AEs Related to SRK-181 or Anti-PD(L)1, All Grades >10%  Dose (MG)  80N=1  240N=1  800N=3  1600N=4  2400N=3  3000N=3  2000N=4  AllN=19  Fatigue  0  1  0  0  1  0  1  3 (15.8%)  Decreased Appetite  1  0  1  0  0  0  0  2 (10.5%)  Nausea  1  0  0  0  0  0  1  2 (10.5%)  Dose (MG)  240N=3  800 N=3  1600N=6  2400N=3  AllN=15  Rash maculo-papular  1  1  1  2  5 (33.3%)  Pruritus  1  1  1  1  4 (26.7%)  Rash  0  1  0  2  3 (20.0%)  Diarrhea  0  0  2  0  2 (13.3%)  Pemphigoid  0  0  0  2  2 (13.3%)  All dose levels were administered q3w except 2000 mg, which was administered q2w.  Treatment-related Grade 3 AEs:  Alanine aminotransferase increased (1 patient)  Treatment-related SAEs:  None  No DLTs  were observed up to 3000 mg q3w and 2000 mg q2w  No Grade 4 or 5 treatment-related AEs occurred  Treatment-related Grade 3 AEs:  Pruritus (2 patients), blister, immune-mediated lung disease, pemphigoid, rash, rash maculo-popular and rash vesicular (1 patient each)  Treatment-related SAEs:  Blister, pruritus, and rash (all in 1 patient) and immune-mediated lung disease (1 patient)  No DLTs  were observed up to 2400 mg q3w  No Grade 4 or 5 treatment-related AEs occurred  Yap T et al. Safety and Efficacy Results of SRK-181, a latent TGFβ1 inhibitor, from a Phase 1 trial (DRAGON Trial); Presented at ESMO-TAT; March 7, 2023. *Clinical cutoff date: December 2, 2022. Response is assessed using RECIST v1.1 by PI; the scan is performed during screening, 6 weeks after first dose, every 9 weeks for the next 6 months of treatment, and every 12 weeks thereafter. SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  33 
 

 Duration of Treatment  DRAGON Part A: Preliminary Efficacy Data*  34  Part A1 (n=19)  Part A2 (n=15)  8 patients had a best response of stable disease (SD)   All 3 patients with ovarian cancer were stable for 6-10 months  Yap T et al. Safety and Efficacy Results of SRK-181, a latent TGFβ1 inhibitor, from a Phase 1 trial (DRAGON Trial); Presented at ESMO-TAT; March 7, 2023. *Clinical cutoff date: December 2, 2022. Response is assessed using RECIST v1.1 by PI; the scan is performed during screening, 6 weeks after first dose, every 9 weeks for the next 6 months of treatment, and every 12 weeks thereafter. SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  At 800 mg q3w, 1 partial response (PR) was observed in patient with anti-PD-1-resistant clear cell renal cell carcinoma (ccRCC)  9 patients had best response of SD  6 patients (green highlight) were stable beyond the 16-week cutoff   1 ongoing patient with head and neck cancer had a 29.4% tumor reduction  
 

 Preliminary Efficacy Data in Combination with Pembrolizumab: Best Response in Target Lesions  35  Best Response in Target Lesions  Best Response of PR  Best Response of SD  Best Response of PD  Ongoing  Percent Change from Baseline  800 mg q3w  800 mg q3w  1600 mg q3w  1600 mg q3w  1600 mg q3w  1600 mg q3w  2400 mg q3w  2400 mg q3w  2400 mg q3w  240 mg q3w  240 mg q3w  800 mg q3w  Head and Neck  Head and Neck  Head and Neck  Uveal Melanoma  NSCLC  Melanoma  Melanoma  Liver  RCC  RCC  TNBC  Squamous Cell Skin Carcinoma  Part A2  Yap T et al. Safety and Efficacy Results of SRK-181, a latent TGFβ1 inhibitor, from a Phase 1 trial (DRAGON Trial); Presented at ESMO-TAT; March 7, 2023. *Clinical cutoff date: December 2, 2022. Response is assessed using RECIST v1.1 by PI; the scan is performed during screening, 6 weeks after first dose, every 9 weeks for the next 6 months of treatment, and every 12 weeks thereafter. SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  Part B (as of 12/2/22)  20 patients dosed across multiple cohorts  Two confirmed PRs ongoing patients with anti-PD-1 resistant clear cell renal cell carcinoma   All dose levels were generally well tolerated SRK-181 dose of 1500 mg q3w or 1000 mg q2w in combination with anti‑PD-(L)1 for Part B  
 

 36  Summary of ccRCC Patients on Combination Treatment   (Part A2 and Part B)  11 ccRCC patients enrolled  n=2 in Part A2 (all discontinued from study) and n=9 in Part B (5 pts remain on study)  enrollment in Part B continues  3 confirmed PRs observed in ccRCC patients  Patients are anti-PD-1 resistant patient (no response on prior anti-PD-1 therapy and disease progress on the most recent prior anti-PD-1 therapy)   ORR: 27% (3/11)  Yap T et al. Safety and Efficacy Results of SRK-181, a latent TGFβ1 inhibitor, from a Phase 1 trial (DRAGON Trial); Presented at ESMO-TAT; March 7, 2023. *Clinical cutoff date: December 2, 2022. Response is assessed by investiogator using RECIST v1.1 by PI; the scan is performed during screening, 6 weeks after first dose, every 9 weeks for the next 6 months of treatment, and every 12 weeks thereafter. SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  As of clinical cutoff date of Dec 2, 2022  
 

 Summary of ccRCC Patients with PR in Dragon (Part A2 and Part B, Combination Treatment)  Responded Pts  SRK-181 Dose   (mg, Q3W)  Age (Year)/   Gender  Lines of   Prior Therapy  IMDC Score   at Screening  Metastatic Sites   at Screening  Pt Status  Duration of   Treatment (wks)  Best % Change in   SOD* from Baseline  Pt #1  Part A2, 800   56/M  Sunitinib  Nivolumab/Ipilimumab  Cabozantinib  Lenvatinib/Everolimus  Pembrolizumab/Axitinib  3   (Poor risk)  Lung/ Lymph Nodes/   Pleural/ Pancreas/ Bone  Off study  30  -57%  Pt #2  Part B, 1500  58/M  Nivolumab/Ipilimumab  Cabozantinib  3   (Poor risk)  Lung/ Lymph Nodes/Liver  Ongoing  32+  (by Dec 2, 2022)  -67%  Pt #3  Part B, 1500  63/M  Nivolumab/Ipilimumab  Nivolumab  Cabozantinib  2   (Intermediate   risk)  Lung/ Lymph Nodes  Ongoing  16+  (by Dec 2, 2022)  -50%  Pt#2: Screening  Common hepatic lymph node  4.0 × 3.0 (cm)  Pt#2: C7D8   Common hepatic lymph node  3.2 × 1.7 (cm)  Pt#1: Screening  Liver nodule segment II  1.7 × 1.4 (cm)  Pt#1: C8D1   Liver nodule segment II  0.0 × 0.0 (cm)  Yap T et al. Safety and Efficacy Results of SRK-181, a latent TGFβ1 inhibitor, from a Phase 1 trial (DRAGON Trial); Presented at ESMO-TAT; March 7, 2023. *Clinical cutoff date: December 2, 2022. Response is assessed using RECIST v1.1 by PI; the scan is performed during screening, 6 weeks after first dose, every 9 weeks for the next 6 months of treatment, and every 12 weeks thereafter. *SOD: sum of diameters in target lesions. SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  37 
 

 SRK-181: Encouraging Early Clinical Data Consistent with Hypothesis   38  TGFβ pathway evaluation (PD)  Target engagement (blood)  TGFβ-1 signaling (tumor p-SMAD2 & RNAseq)  Immunophenotyping, including immune exclusion status  Tumor immune contexture (e.g., tumor CD8+ T cells)  Immune cell contexture (tumor & blood MDSCs)  Immune response markers (e.g., IO gene signature)   Therapeutically relevant dose  Drug exposure needed for efficacy  Objective response  Anti-tumor response and survival benefits  TGFβ pathway evaluation (PD)  Target engagement (blood)  TGFβ-1 signaling (tumor p-SMAD2 & RNAseq)  Immunophenotyping, including immune exclusion status  Tumor immune contexture (e.g., tumor CD8+ T cells)  Immune cell contexture (tumor & blood MDSC’s)  Immune response markers (e.g., IO gene signature)  Therapeutically relevant dose  Dosing regimens achieved target steady state levels  Objective response  Anti-tumor response observed (partial responses)  Preclinical Data  Phase 1 DRAGON proof-of-concept trial 
 

 39  *(PD-1/PD-L1)  First in class monoclonal antibody targeting latent and context-independent binding to TGFβ1  Differentiated from other TGFβ inhibitors by its novel selectivity  Offers potential to avoid toxicity and dose-limiting challenges of non-selective TGFβ inhibition approaches  Differentiation  Emerging evidence implicates TGFβ1 as driving resistance to checkpoint inhibitor therapies  Potent and selective inhibitor of latent TGFβ1 activation in preclinical studies  Strong safety and preclinical efficacy data  Strong  Scientific  Rationale  DRAGON Part A demonstrated ability to escalate to high doses of SRK-181 at levels exceeding the anticipated efficacious drug exposure level  Advanced to DRAGON Part B: Evaluating SRK-181 in 5 parallel tumor-specific cohorts, with efficient path towards early POC for each  Early efficacy signals have been observed  Clear  Clinical  Pathway  PD-(L)1* becoming a standard of care therapy in many tumor types; the market for synergistic combination product would be vast  SRK-181 could potentially be used in many tumor types, potentially both in patients resistant to PD-(L)1 and in CPI naïve patients, as well as other therapeutic applications  High Unmet  Medical Need  & Large  Commercial Opportunity  SRK-181Summary 
 

 Next Horizon: Fibrosis 
 

 TGFβ is Established as Key Driver of Fibrosis Across Multiple Diseases   41  Nature Reviews , April 25, 2016  NATURE REVIEWS | NEPHROLOGY  TGF-β: the master regulator of fibrosis  Xiao-ming Meng1, David J. Nikolic-Paterson2 and Hui Yao Lan3  Nature Reviews. August 19, 2014  NATURE REVIEWS | RHEUMATOLOGY  Transforming growth factor β―at the centre  of systemic sclerosis  Robert Lafyatis  Int. J. Mol. Sci. August 27, 2018  Targeting TGF-β Signaling in Kidney Fibrosis  Yoshitaka Isaka  J. Am. Soc. Nephrol. December 3, 2017  Targeting Anti-TGF-β Therapy to Fibrotic Kidneys  with a Dual Specificity Antibody Approach  Steve McGaraughty,* Rachel A. Davis-Taber,† Chang Z. Zhu,* Todd B. Cole,*  Arthur L. Nikkel,* Meha Chhaya,† Kelly J. Doyle,* Lauren M. Olson,* Gregory M. Preston,†  Chrisine M. Grinnell,† Katherine M. Salte,* Anthony M. Giamis,* Yanping Luo,*  Victor Sun,† Andrew D. Goodearl,† Murali Gopalakrishnan,* and Susan E. Lacy†  J Pathol, July 25, 2021  TGF-β as a driver of fibrosis: physiological roles and therapeutic opportunities  Erine H Budi1, Johanna R Schaub1, Martin Decaris1, Scott Turner1, Rik Derynck2  J Receptors Sign Trans, Feb 13, 2020  Inevitable role of TGF-β in progression of nonalcoholic fatty liver disease  Bhagyalakshmi Nair and Lekshmi R. Nath  PNAS, February 24, 1986  Transforming growth factor type β: Rapid induction of fibrosis and  angiogenesis in vivo and stimulation of collagen formation in vitro  ANITA B. ROBERTS* MICHAEL B. SPORN*, RICHARD K. ASSOIAN*, JOSEPH M. SMITH*, NANETTE S. ROCHE*, LALAGE M. WAKEFIELD*, URSULA I. HEINE*, LANCE A. LIOTTA*, VINCENT FALANGA†, JOHN H. KEHRL‡, AND ANTHONY S. FAUCI‡  Proc Am Thorac Soc, July 3, 2006  Transforming Growth Factor β  A Central Modulator of Pulmonary and Airway Inflammation and Fibrosis  Dean Sheppard 
 

 GARP Presents TGFβ on Tregs  LRRC33 Presents TGFβ on Macrophages  LTBP1 & 3 Present TGFβ in Connective Tissue  Targeting Presenting Molecule/TGFβ-1 Complexes Provides Context Specificity   Targeting Latent TGFβ-1 Complexes Creates Multiple “Handles” For Selectivity  42  Latent TGF-β  GARP  Treg  Integrin expressing cell  avb8  Integrin  Latent TGF-β  LRRC33  Macrophage  Integrin expressing cell  avb8  Integrin  Latent TGF-β   binding protein   (LTBP)  Extracellular matrix  Fibrillin  Latent TGF-β  Epithelial  Cell  avb6  Integrin  Latent TGFβ-1 
 

 LTBP-49247 Reduced TGFβ Signaling and Fibrosis in Preclinical Models of Kidney Fibrosis  43  LTBP-49247 reduced a TGFβ PD biomarker in kidneys of Col4a3 KO mice (Alport Syndrome model)  LTBP-49247 reduced fibrosis in kidneys of Alport model  ** p < 0.01  One way ANOVA vs. IgG  HYP=hydroxyproline  Efficacy also seen in rat model of kidney fibrosis   No observed toxicity in mouse 13-week non-GLP repeat dose study  Favorable PK in cynomolgus monkeys (t1/2 ~28 days) suggests LTBP-49247 is amenable to clinical subcutaneous dosing with promising developability profile 
 

 Significant Opportunities to Address High Unmet Need Across Multiple Fibrotic Indications  44  Alport Syndrome (AS)   Focal Segmental Glomerulosclerosis (FSGS)   IgA Nephropathy (IgAN)  Primary Sclerosing Cholangitis (PSC)  Diffuse Cutaneous Systemic Sclerosis (dcSSc)  Idiopathic Pulmonary Fibrosis (IPF)  Collectively, significant commercial potential given large patient population with clear high unmet need given poor outcomes and lack of effective therapeutics  Significant impact to delay or stop progression to end-stage disease and organ transplant   Expansion opportunities via other indications given shared etiologies 
 

 Next Horizon: Iron-Restricted Anemia 
 

 BMP6/RGMc Pathway is a Well Validated Regulator of Systemic Iron Homeostasis   46  Fig: Muckenthaler, M.U., Rivella, S., Hentze, M.W. and Galy, B. (2017) A Red Carpet for Iron Metaboism. Cell, 168(3): 344-361  1: Kuns-Hashimoto R, et al. (2008) Selective binding of RGMc/hemojuvelin, a key protein in systemic iron metabolism, to BMP-2 and neogenin. Am J Physiol Cell Physiol 294(4):C994-C1003  2: Constante M, et al.. (2007) Repression of repulsive guidance molecule C during inflammation is independent of Hfe and involves tumor necrosis factor-alpha. Am J Pathol 170(2):497-504  3: Core A.B., et al. (2014) Hemojuvelin and bone morphogenetic protein (BMP) signaling in iron homeostasis. Front Pharmacol. 5:104.   4. Wang CY and Babitt JL. (2016) Hepcidin Regulation in the Anemia of Inflammation. Curr Opin Hematol 23(3): 189-197.  Elevation of proinflammatory cytokines drives increased hepcidin expression and results in anemia due to functional iron deficiency4  Anemia of Inflammation/ Chronic Disease  Hepcidin  Serum iron  Untreated  Hepcidin  Serum iron  Ab treated  Human mutations in HJV/RGMc establish it as a central player in hepcidin regulation1  Knockout phenotypes and tissue-specific expression pattern demonstrate that its predominant role is in iron homeostasis2  Member of repulsive guidance molecule (RGM) family (RGMa, RGMb, RGMc/HJV) that act as BMP co-receptors to modulate BMP signaling3  HJV/RGMc is a key player in the regulation of hepcidin expression 
 

 HJV-35202: An Investigational High-Affinity Antibody Demonstrating Selective Inhibition of HJV/RGMc and Robust PK/PD in Cynos  47  Potent in vitro binding affinity (KD=3.9E-11)  Highly specific to RGMc/HJV with well understood mechanism  Sustained PD effect in single dose Cyno study  Specific to RGMc over other RGM family members  Nicholls S.B., et al. Poster: RGMc-selective antibodies modulate iron homeostasis in vivo, 12th International BMP Conference, Tokyo, October 2018  Scholar Rock, Data on File  High-affinity antibody  Specific to RGMc, with mechanism of specificity understood  Cross-reactive to human, mouse, rat and cyno  Sustained PD observed in healthy rats and cynos, with clear PK/PD relationship  Highly manufacturable framework with no sequence liabilities  Formulatable into a subcutaneous format (150 mg/mL)  HJV-35202 (mg/mL)  Serum Iron and UIBC (mg/dL)   PK: HJV-35202 (10mg/kg, IV)  PD: UIBC: HJV-35202 (10mg/kg, IV)  PDF: Serum Iron: HJV-35202 (10mg/kg, IV)  Octet Response Units  RGMa  RGMb  RGMc/HJV  RGMa  RGMb  RGMc/HJV  Key Attributes of HJV-35202:1,2 
 

 Significant Opportunities to Target Iron-Restricted Anemias Across Multiple Indications  48  Chronic Kidney Disease (CKD)  Anemia of Chronic Inflammation (AI)  Myelofibrosis (MF)  Targeting RGMc/HJV for anemia is well validated and relatively de-risked  High levels of hepcidin, the main regulator of systemic iron metabolism, are associated with anemia across various diseases  Safe and convenient RGMc inhibitor has promise of improving patient outcomes across multiple indications as stand alone or in combination with SoC   Significant and clear unmet need given lack of approved treatments or severe limitations of current treatments  Well defined patient population  Collectively, sizeable commercial opportunity given relatively large population  Potential for rapid POC with clear regulatory path  Opportunity to build an anemia franchise with initial POC and indication expansion in the future 
 

 Scholar Rock Summary  
 

 50  Key Investment Highlights  Revolutionary Platform  Overcome the Challenges  targeting the latent forms of growth factors  Discover and Develop  monoclonal antibodies with extraordinary selectivity  Positioned For Success  Upcoming Data Readouts  for both clinical programs  $205M financing in June 2022  Cash balance of $275M as of March 31, 2023; anticipated runway into 2025  Robust Clinical Pipeline  SRK-181 (Phase 1)  Potential to shift current treatment landscape for cancer patients with CPI resistance  Apitegromab (Phase 3)  Potential first-in-class  Significant market opportunity   Program on track  Clear path to approval  Discovery-stage Pipeline  Fibrosis and iron-restricted anemia  Strategic optionality  
 

 Appendix 
 

 TOPAZ Baseline Characteristics1,2  52  *Patients on average received ~2 years of nusinersen treatment at baseline and ~3 years of nusinersen treatment by the end of the TOPAZ study (12-months). SMN2 copy numbers were not available for all patients. †12-month baseline characteristics recorded in the table, 1-cohort 1 patient and 1- cohort 2 patient discontinued during 24M Extension Period A. All discontinuations were for reasons unrelated to study drug.  HFMSE, Hammersmith Functional Motor Scale–Expanded; max, maximum; min, minimum; RHS, Revised Hammersmith Scale; SMN, survival motor neuron.  1. Crawford T, et al. Neuromuscul Disord. 2022;32(Suppl1):S86-S87. P102. 2. Crawford T, et al. TOPAZ Extension: 24-month Efficacy and Safety of Apitegromab in Patients With Later-onset Spinal Muscular Atrophy (Type 2 and Type 3 SMA). Presented at CureSMA Annual Conference; June 16-19, 2022.  N (dosed)  Mean age (min, max)  Mean RHS (min, max)  Mean HFMSE (min, max)  Prior Nusinersen, Months  Mean (min, max)*  No. of patients with  2,3, or 4 SMN2 copies  Discontinuation(s)  Ambulatory  COHORT 1: Age 5-21  20 mg/kg  monotherapy  11  12  (7, 19)  48  (26, 63)     N/A  1, 4, 4  0  20 mg/kg  + nusinersen  12  13  (7, 21)  51  (43, 62)  20 (12, 28)  0, 9, 1  1†  Pooled  23  13  (7, 21)  50  (26, 63)  N/A  1, 13, 5  1†  Nonambulatory  COHORT 2: Age 5-21  20 mg/kg  + nusinersen  15  12  (8, 19)     23  (13, 39)  25 (12, 39)  0, 11, 2  0  COHORT 3: Age 2+  20 mg/kg  + nusinersen  10  4  (2, 6)     24  (14, 42)  24 (10, 34)  1, 8, 0  0  2 mg/kg  + nusinersen  10  4  (2, 6)  26  (12, 44)  1, 8, 1  0  Pooled  20  4  (2, 6)  25  (12, 44)  2, 16, 1  0 
 

 TOPAZ Patient Disposition Over 36 Months  a. Patients stratified based on previous treatment with approved SMN therapy.  b. Patients randomized to receive 2 or 20 mg/kg apitegromab.  *Includes patients who crossed over from 2 mg/kg to 20mg/kg starting week 68 through week 104  ** Excludes patients on monotherapy  SMN Rx=SMN therapy.  SMN Rx ≥ 5  ENROLLMENT n=58   YEAR 1: Thx Period n=57  YEARS 2-4: ExtensionN=50  ONYX  Study  NONAMBULATORY  N=20  Ages ≥ 2  Type 2 SMA   n=20  Apitegromab 20 mg + nusinersen*  SMN Therapy  Yes  No  n=10  n=10  Apitegromab 2 mg + nusinersen  Apitegromab 20 mg + nusinersen  NONAMBULATORY  N=15  Ages 5-21  Type 2 or Type 3 SMA  n=15  Apitegromab 20 mg + nusinersen  n=15  Apitegromab 20 mg + nusinersen  1 withdrew consent (COVID concern)  57 ENROLLED  7 discontinued   2 due to concerns with COVID-19  5 on monotherapy due to lack of benefit  >90% of patients on combination therapy remained in study**  TOPAZ patient retention  PRIMARY TREATMENT:  EXTENSION:  58 ENROLLED  57 completed primary treatment period and enrolled in the extension study   1 withdrew consent due to fatigue & weight gain  SMN Therapy  AMBULATORY   N=23  Ages 5-21  Type 3 SMA  1 withdrew consent (fatigue /weight gain)  1 withdrew consent (COVID concern)  Yes  No  n=11  n=12  Apitegromab 20 mg monotherapy  Apitegromab 20 mg + nusinersen  n=11  n=11  Apitegromab 20 mg monotherapy  Apitegromab 20 mg + nusinersen  4 withdrew consent (lack of benefit)  1 withdrew consent (lack of benefit)  53  SMN Rx < 5 
 

 Time (weeks)  No. of observations  Year 3  Year 2  Year 1  Year 3  Year 2  Year 1  PROMIS Fatigue (Proxy)  Year 2  Year 3  Year 1  Year 2  Year 3  Year 1  Pooled Nonambulatory Patients | Age 2 – 12 | All Doses | Over 36 MonthsImprovements in PRO Measures Consistent With Motor Function  54  Time (weeks)  No. of observations  IMPROVEMENT  PEDI-CAT Daily Activities  IMPROVEMENT  IMPROVEMENT   N = 29.; Baseline mean age=5.5 |Time on SMN Rx=24.6m   HFMSE  29  29  29  23  24  26  26  26  26  23  22  23  22  Change from baseline (±SE)  25  23  24  16  18  20  20  18  23  19  14  15  13  IMPROVEMENT  RULM  28  28  28  22  23  26  25  26  26  25  22  22  22  Change from baseline (±SE)  21  19  18  15  15  18  12  17  16  16  13  12  11  HFMSE=Hammersmith Functional Motor Scale Expanded; OC=observed case; PEDI-CAT=Pediatric Evaluation of Disability Inventory Computer Adaptive Test; PROMIS=Patient Reported Outcome Measurement Information System; RULM=Revised upper limb module; SE=standard error of the mean. SMN Rx=SMN therapy. Data on File. Scholar Rock, Inc. Cambridge, MA. Data cutoff date as of March 13, 2023. Error bars represent standard error (SE). Data cutoff date as of March 13, 2023. The updated PEDI-CAT analysis included additional records (2 at 12 months and 1 at 24 months) that were not available at the time of previous analysis. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established. 
 

 PEDI-CAT Daily Activities Over 36 Months Improvements Were Substantial and Sustained  55  For the 36-month evaluation, an observed case analysis was conducted, which pooled all the nonambulatory patients (Cohorts 2 and 3) and was based upon the available data for given timepoints. This analysis population included patients receiving either low dose (2 mg/kg) or high dose (20 mg/kg) apitegromab (inclusive of patients in Cohort 3 who switched from 2 mg/kg to 20 mg/kg in Year 2). Error bars represent SE. CI represents confidence interval. SMN Rx=SMN therapy. Data cutoff date as of March 13, 2023. The updated analysis included additional records (2 at 12 months and 1 at 24 months) that were not available at the time of previous analysis. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Baseline mean age=7.3 | Time on SMN Rx=24.1m   Baseline mean age=5.5 | Time on SMN Rx=24.6m   n=  35  21  25  23  17  95% CI=  (-0.0, 2.6)  (-0.1, 2.6)  (0.9, 3.5)  (-0.1, 4.5)  Mean Change from Baseline (+SE)  n=  29  16  20  19  15  95% CI=  (-0.1, 2.0)  (-0.5, 2.1)  (0.5, 3.2)  (-0.8, 4.2)  Pooled Nonambulatory Patients   Age 2-21 YearsAll Doses (N=35)  Age 2-12 YearsAll Doses (N=29) 
 

 PEDI-CAT Daily Activities Over 36 Months Improvements Were Substantial and Sustained  56  For the 36-month evaluation, an observed case analysis was conducted, which pooled all the nonambulatory patients (Cohorts 2 and 3) and was based upon the available data for given timepoints. This analysis population included patients receiving either low dose (2 mg/kg) or high dose (20 mg/kg) apitegromab (inclusive of patients in Cohort 3 who switched from 2 mg/kg to 20 mg/kg in Year 2). Error bars represent SE. CI represents confidence interval. SMN Rx=SMN therapy. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Baseline mean age=7.3 | Time on SMN Rx=24.1m   Baseline mean age=5.5 | Time on SMN Rx=24.6m   n=  35  21  25  23  17  95% CI=  (-0.0, 2.6)  (-0.1, 2.6)  (0.9, 3.5)  (-0.1, 4.5)  Mean Change from Baseline (+SE)  n=  29  16  20  19  15  95% CI=  (-0.1, 2.0)  (-0.5, 2.1)  (0.5, 3.2)  (-0.8, 4.2)  Pooled Nonambulatory Patients   Age 2-21 YearsAll Doses (N=35)  Age 2-12 YearsAll Doses (N=29) 
 

 PROMIS Fatigue (Proxy) Over 36 Months Improvements Were Substantial and Sustained  57  For the 36-month evaluation, an observed case analysis was conducted, which pooled all the nonambulatory patients (Cohorts 2 and 3) and was based upon the available data for given timepoints. This analysis population included patients receiving either low dose (2 mg/kg) or high dose (20 mg/kg) apitegromab (inclusive of patients in Cohort 3 who switched from 2 mg/kg to 20 mg/kg in Year 2). Error bars represent SE. CI represents confidence interval. SMN Rx=SMN therapy. Data cutoff date as of March 13, 2023. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Mean Change from Baseline (+SE)  Pooled Nonambulatory Patients   n=  35  19  17  20  14  95% CI=  (-4.3, 1.4)  (-5.7, 0.8)  (-6.0, -0.3)  (-8.7, -0.5)  n=  29  15  13  17  12  95% CI=  (-3.7. 2.8)  (-6.0, 0.2)  (-6.0. 0.1)  (-8.9, 0.4)  Baseline mean age=7.3 | Time on SMN Rx=24.1m   Baseline mean age=5.5 | Time on SMN Rx=24.6m   Age 2-21 YearsAll Doses (N=35)  Age 2-12 YearsAll Doses (N=29) 
 

 TOPAZ Topline 12-Month Data Showed Apitegromab’s Transformative Potential in Patients with Type 2/3 SMA  58  Crawford T et al. TOPAZ topline results; Presented at CureSMA, 2021 Virtual SMA Research & Clinical Care Meeting; June 9-11, 2021  * Pooled cohorts of nonambulatory patients treated with apitegromab 20 mg/kg and 2 mg/kg  **Nonambulatory patients who initiated background nusinersen at a young age of <5 years and treated with apitegromab 20 mg/kg dose. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  HFMSE change from baseline  Majority of nonambulatory patients* experienced HFMSE increases from apitegromabduring chronic maintenance phase of SMN therapy  Apitegromab led to HFMSE improvements in both nonambulatory cohortsincluding patients started on nusinersen at age ≥ 5  Initiated background nusinersen   Age < 5**  Age ≥ 5  Mean HFMSE Increase  +7.1 points  +0.6 points  ≥ 1-point Increase % (n/N)  88% (7/8)  64% (9/14)  ≥ 3-point Increase% (n/N)  63% (5/8)  29% (4/14) 
 

 TOPAZ Age 2-12 Analysis* in Pooled Nonambulatory Cohorts (20mg/kg) at 12 Months Mean Increase of Motor Function Outcomes by HFMSE was Significant   59  Mean HFMSE Increase  OF 4.4 POINTS  with majority experiencing ≥ 3-point increases on top of background SMN therapy  HFMSE Gains Also Notable  in subset of individuals in this analysis who had started background nusinersen at age ≥ 5:   75% (6/8) with ≥ 1-point increase  50% (4/8) with ≥ 3-point increase  Nonambulatory Types 2/3 SMA  (Apitegromab 20 mg/kg; Intent-to-Treat Population)  Age 2-12 years  (n=16†)  Mean HFMSE change from baseline, (95% CI)  +4.4 (1.3, 7.4)  Patients with ≥ 1-pt increase in HFMSE, n (%)  13 (81%)  Patients with ≥ 3-pt increase in HFMSE, n (%)  9 (56%)  TOPAZ results showed HFMSE improvement from baseline or RHS stabilization across all three pre-specified cohorts.1  No safety signals for apitegromab were identified through month 12 of TOPAZ; the five most frequently reported treatment-emergent adverse events were headache, pyrexia, upper respiratory tract infection, cough, and nasopharyngitis  *Exploratory, post hoc analysis. †For 12-month endpoint, if participants skipped three consecutive doses due to site restrictions caused by COVID-19, records after dose skipping were excluded from analysis. The last observation carried forward was used for other missing data.  1. Crawford T et al. TOPAZ topline results. Presented at Muscular Dystrophy Association, 2023 Clinical & Scientific Conference, March 22, 2023. CI, confidence interval; HFMSE, Hammersmith functional motor scale expanded; SMA, spinal muscular atrophy. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established. 
 

 TOPAZ 12-Month Data | Nonambulatory Type 2 High Dose CohortInitiated nusinersen age <51,2  60  Increases in HFMSE observed in patients already treated with chronic maintenance nusinersen  Improved: 88% (7/8)  ≥ 5-point increase: 63% (5/8)  > 10-point increase: 38% (3/8)  Continuous and durableimprovements observed through12-months of treatment  Apitegromab (20 mg/kg) + nusinersen  n=8*  Mean change from baseline in HFMSE  (95% CI)  +7.1   (1.8, 12.5)  # (%) patients achieving:  ≥ 1-pt increase in HFMSE  7/8 (88%)  ≥ 3-pt increase in HFMSE  5/8 (63%)  ≥ 5-pt increase in HFMSE  5/8 (63%)  Baseline characteristics: mean (min, max)  n=10  Age  3.8 (2, 6)  HFMSE score  23.5 (14, 42)  # of nusinersen maintenance doses  5.4 (3, 8)  *This was a primary intent-to-treat (ITT) analysis that, as prespecified, excluded 2 patients who missed 3 doses due to COVID-19 related site access restrictions. An all-patients sensitivity analysis that included those 2 patients had similar results as this primary ITT analysis.   Crawford T et al. TOPAZ topline results; Presented at CureSMA, 2021 Virtual SMA Research & Clinical Care Meeting; June 9-11, 2021. 2. Data on file; Scholar Rock. 2022.  Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established. 
 

 TOPAZ 12-Month Data | Nonambulatory Type 2/3 CohortInitiated nusinersen age ≥51,2  61  Majority of patients improved in HFMSE (despite initiating background nusinersen age ≥ 5)  ≥ 1-point increase: ~67%  ≥ 3-point increase: ~30%  Durability of effect observed through 12-months of treatment  *Intent-to-treat analysis excluded 1 patient (per prespecified approach) who missed 3 doses due to COVID-19 related site access restrictions; 1 patient who had inadvertently been enrolled who was receiving (and continued to receive) an acetylcholinesterase inhibitor was removed, which is not permitted per the trial protocol; 1. Crawford T et al. TOPAZ topline results; Presented at CureSMA, 2021 Virtual SMA Research & Clinical Care Meeting; June 9-11, 2021. 2. Data on file. Scholar Rock, Inc. Apitegromab is an investigational drug candidate being evaluated for the treatment of spinal muscular atrophy. Apitegromab has not been approved for any use by the US FDA or any other health authority, and its safety and efficacy have not been established.  Apitegromab (20 mg/kg) + nusinersen  Per Protocol Population*   (n=13)  Intent-to-Treat Population  (n=14)  Mean change from baseline in HFMSE   (95% CI)  +1.2   (-0.5, 2.9)  +0.6   (-1.4, 2.7)  # (%) patients achieving:  ≥ 1-pt increase in HFMSE  9/13 (69%)  9/14 (64%)  ≥ 3-pt increase in HFMSE  4/13 (31%)  4/14 (29%)  ≥ 5-pt increase in HFMSE  2/13 (15%)  2/14 (14%)  Baseline characteristics: mean (min, max)  n=15  Age  11.7 (8, 19)  HFMSE score  22.7 (13, 39)  # of nusinersen maintenance doses  5.1 (2, 9) 
 

 TOPAZ 12-Month HFMSE Changes and Duration of Prior Nusinersen  62  Patients enrolled were already in the chronic maintenance phase of nusinersen  Lack of clear correlation between 12-month HFMSE & duration of prior nusinersen exposure in patients aged 2 – 21 suggests motor function improvement mainly attributable to apitegromab  Post hoc analysis: Scatter plot of prior nusinersen treatment duration vs change in HFMSE from baseline; in nonambulatory Types 2 and 3 participants in TOPAZ, there was no clear relationship between duration of nusinersen treatment and change in motor function. Patients skipped 3 or more doses due to COVID-site restrictions excluded; apitegromab is an investigational product candidate under development. HFMSE, Hammersmith Functional Motor Scale–Expanded 
 

 1. Mercuri E, et.al. Nusinersen versus sham control in later-onset spinal muscular atrophy. N Engl J Med. 2018;378:625-635.  2. Efficacy and safety of risdiplam (RG7916) in patients with Type 2 or non-ambulant Type 3 spinal muscular atrophy (SMA) Roche/PTC Therapeutics  This third-party information is provided for background only and is not intended to convey or imply a comparison to the TOPAZ clinical trial results  Nusinersen CHERISH Trial in Later-Onset SMA1  In patients with later-onset SMA who were age >5 at screening:  Primary benefit of nusinersen: stabilization of motor function  Majority of patients do not experience HFMSE increases  Nonambulatory Type 2/3 SMA Majority of Patients Started on SMN Therapy After Age 5 Do Not Experience Motor Function Increases  63  Risdiplam SUNFISH Trial in Later-Onset SMA2  Low percentage of patients over the age of 5 achieved ≥3-point increase on MFM32 scale, even with risdiplam treatment  HFMSE secondary endpoint showed a mean 0.58-point improvement over placebo (not statistically significant)   Change  From Baseline  to 15 Months in HFMSE Score   Age, year  2  4  6  8  Nusinersen (N=66)  Control (N=34)  % of patients with > 3 change in   MFM32 total at Month 12  Change in MFM32 total score  2-5 Years  6-11 Years  12-17 Years  18-25 Years  Risdiplam  Placebo 
 

 Significance of Hammersmith Functional Motor Scale Expanded (HFMSE) and Revised Upper Limb Module (RULM)  64  HFMSE  Assesses the physical abilities of patients with Types 2/3 SMA  ABLE TO:  Touch Head Above Ear Level  whilst maintaining stable trunk and head  Roll From Supine to Prone  over the right side without pulling/ pushing on hands  33 Items  Graded on scale 0 to 2  0 = unable  1 = performed with modification or adaptation  2 = without modification or adaptation   Item scores are summed to give a total score   The higher the total score, the greater the patient’s motor function  Maximum score: 66  Examples of items:  One hand to head in sitting  Rolls supine to prone  Lying to sitting  Four–point kneeling  Supported standing  Stepping  Ascends 4 stairs with railing  RULM  Evaluates Motor Performance in Upper Limbs  ABLE TO:  Bring Token  to Cup  placed vertically at shoulder height  Bring Weight at Eye Level using two hands  19 Items  Graded on scale 0 to 2(Except for 1 activity with a binary score)  0 = unable  1 = able with modification  2 = able with no difficulty  Evaluated upper limb tasks correspond to ability to perform everyday activities  Maximum score: 37  Examples of items:  Putting a coin into a cup  Elevating a cup to mouth  Picking up a coin  Bringing hand to shoulder  Lifting up weighted objects  Opening a zip lock bag   Drawing a line on paper  O’Hagen et al. 2007; Glanzman et al. 2011; Hammersmith Functional Motor Scale Expanded for SMA (HFMSE) Manual, 2019  Mazzone et al. 2017; Pierzchlewicz et al. 2021; Revised Upper Limb Module for SMA Manual, 2014  
 

 PEDI-CAT:  Measure of activities of daily living  PROMIS (Fatigue):  Measure of Patient Fatigue   ESBBT (Fatigability):  Measure of how fast a patient fatigues  1  2  3  PRO measurement tool4  Muscle endurance measurement tool6  4-point scale (1=unable to 4=easy) assessment of various activities, higher scores reflect improved abilities1,2  PEDI-CAT has been validated in SMA, but alone cannot identify small changes in function across all types of SMA3  Measures mild subjective feelings of tiredness to debilitating and sustained feelings of exhaustion, with lower scores reflecting less fatigue4,5  Has been utilized to assess fatigue and fatigability in the Cure SMA database, but has not been fully validated in SMA5  Part of a series of endurance shuttle tests that include: nine-hole peg test, box and block test, and walk test (ESNHPT, ESBBT, and ESWT)6  Patients are asked to move blocks individually from one box to another in one minute, with higher numbers of blocks suggesting higher muscle endurance6  The endurance shuttle tests have been validated for use in patients with SMA7  Measures pediatric abilities through 3 functional domains, daily activities, mobility, and social cognitive1  Activities of Daily Living and Fatigue: Assessed by Three Measures  65  ADL, activities of daily living; ESBBT, endurance shuttle box and block test; ESNHPT, endurance shuttle nine-hole peg test; ESWT, endurance shuttle walk test; PEDI-CAT, pediatric evaluation of disability inventory computer adaptive test; PROMIS, patient-reported outcomes measurement information system; PRO(s), patient-reported outcome(s); SMA, spinal muscular atrophy. 1. Cre Care. PEDI-CAT. Accessed April 26, 2022. https://www.pedicat.com/. 2. Data on file; Scholar Rock. 2022. 3. Pasternak A, et al. Muscle Nerve. 2016;54(6):1097-1107. 4. NIH. PROMIS. Accessed April 26, 2022. https://commonfund.nih.gov/promis/index. 5. Belter L, et al. Orphanet Journal of Rare Diseases. 2020;15:217. 6. Cure SMA. Best Practices for Physical Therapists and Clinical Evaluators in Spinal Muscular Atrophy (SMA). 2021. Available at: https://www.curesma.org/wp-content/uploads/2021/09/Clinical-Evaluators-Best-Practices-13-August-2021.pdf. 7. Bartels B, et al. Orphanet Journal of Rare Diseases. 2020;15:75.   PEDI-CAT, PROMIS, and ESBBT   Used to assess:  ADL  Fatigue  Muscle Endurance 
 

 Reported Impact of Scoliosis Surgery on Motor Abilities in SMA  66  Dunaway Young et al. 2020  Scoliosis Surgery  HFMSE Total Score  3-month post-surgery assessment  14/17  Lost >3 points on the HFMSE (mean change = - 12.1, SD = 8.9)   Functionally meaningful change   13/17  Minimal HFMSE changes within ± 2 points (mean change = - 0.7)  No change or stability  0/17  Improvement > 2 points post-surgery  Post-Surgery HFMSE scores Type 2/3 SMApeer-reviewed study  
 

 SRK-181: Transformative Potential as the Backbone For Next Era of Cancer Therapy  67  CPI Combination  (solid tumors)  Other Combinations (solid tumors)  & Hematologic  Other immunotherapies  Chemotherapy   XRT  Otherapplications  Myelofibrosis  Liquid tumors  1st line immunotherapy  First in class monoclonal antibody targeting latent and context-independent binding to TGFB-1  CURRENT FOCUS  Primary and AcquiredCPI resistance  Investigation in primary (pre-existing) CPI resistance offers path to early clinical POC for SRK-181 
 

 Regulatory T cell  Tumor Associated   Macrophage  Cancer Associated Fibroblasts (Stromal cells)  Tumor cells  SRK-181 Attributes  Targets latent TGFβ-1   Inhibits activation of latent TGFβ-1 across ALL compartments  Turns it off at the source  Context-Independent: SRK-181 Designed to Inhibit Latent TGFβ-1 Across All Compartments of the Tumor Microenvironment  68  Latent TGFβ-1  Key driver of tumor resistance to CPIs   Present in multiple compartments of the tumor microenvironment  TGFβ Latent growth factor  TGFβ source presentation  LRRC33  GARP  LTBPs  LTBPs  LRRC33: leucine-rich-repeat-containing protein family member 33 | LTBPs: latent transforming growth factor β binding proteins | GARP: glycoprotein A repetitions predominant 
 

 SRK-181 Therapeutic Hypothesis: Potential Advantages of Latent TGFβ-1 Inhibitor  69  SRK-181*  Bifunctional TGFβ/CPI   ALK5 Inhibitor  Nonselective TGFβ antibody  Selectivity for TGFβ-1: potential for wider therapeutic window and improved safety  Ability to combine with any anti-PD-(L)1        Ability to optimize dosing of each component of combination therapy  Activity spatially distinct from anti-PD-(L)1 in tissue     Inhibits all sources of TGFβ-1 contributing to CPI resistance (Context independent)  Target latent form (Blocks TGFβ-1 activation)  *SRK-181 is an investigational product candidate currently being evaluated in DRAGON phase 1 clinical trial. The efficacy and safety of SRK-181 have not been established.   X  X  X  X  X  X  X  X  X  X 
 

 TGFβ-1 Blockade with SRK-181-mIgG1 Rendered Preclinical Tumor Models Susceptible to Anti-PD1 Therapy  70  Days after treatment initiation  Tumor volume (mm3)  Anti-PD1/  SRK-181-mIgG1(3 mg/kg QW)  4/14  Anti-PD1/  SRK-181-mIgG1(10 mg/kg QW)  8/14  Preclinical data published in Science Translational Medicine. Martin CJ, et al. Sci Transl Med. 2020 Mar 25;12(536):eaay8456. https://scholarrock.com/platform/publications/.  *SRK-181-mIgG1 is the murine version of SRK-181; responder defined as tumor size <25% endpoint volume at study end.  Bladder Cancer  Breast Cancer (TGFβ-1/3 co-expressing)  0/13  Anti-PD1 (10 mg/kg BIW)  0/10  SRK-181-mIgG1* (10 mg/kg QW)  Anti-PD1/  SRK-181-mIgG1(10 mg/kg QW)  5/10  Responders 0/9  Control  0/9  Anti-PD1 (10 mg/kg BIW)  SRK-181-mIgG1* (10 mg/kg QW)  Responders 0/12  MBT-2 and EMT6 Models  
 

 SRK-181-mIgG1 Combination Treatment Led to Melanoma Tumor Regression and Survival Benefit   71  *P<0.01.   †P<0.05 Log-rank (Mantel-Cox test) vs anti-PD1.  *  *  †  Days after treatment initiation  Tumor Regression  Survival Benefit  Anti-PD1 + SRK-181-mlgG1 (30 mg/Kg/wk)  Anti-PD1 + SRK-181-mlgG1 (3 mg/Kg/wk)  Anti-PD1 + SRK-181-mlgG1 (10 mg/Kg/wk)  Anti-PD1 (10 mg/Kg/2xwk)  SRK-181-mlgG1 (30 mg/Kg/wk)  Control  Melanoma (Cloudman S91) model  Preclinical data published in Science Translational Medicine. Martin CJ, et al. Sci Transl Med. 2020 Mar 25;12(536):eaay8456. https://scholarrock.com/platform/publications.  Days after treatment initiation  Monotherapy  3/12  Anti-PD1  (10 mg/kg BIW)  Tumor volume (mm3)  SRK-181-mIgG1  (30 mg/kg QW)  Responders 0/12  Combination Therapy  Led to tumor regression and survival benefit  Anti-PD1/SRK-181-mIgG1 (30 mg/kg QW)  8/11  Days after treatment initiation  Anti-PD1/SRK-181-mIgG1 (10 mg/kg QW)  Tumor volume (mm3)  4/9 
 

 Selectivity of SRK-181 Offers Potential to Overcome Toxicity and Dose-limiting Challenges of Non-selective TGFβ Pathway Approaches   72  Preclinical data published in Science Translational Medicine. Martin CJ, et al. Sci Transl Med 2020 Mar 25;12(536): eaay8456.   *Source: Anderton MJ, et al. Induction of heart valve lesions by small-molecule ALK5 inhibitors. Toxicol Pathol. 2011;39: 916-924.; and Stauber AJ, et al. Nonclinical safety evaluation of a transforming growth factor β Receptor I kinase inhibitor in Fischer 344 rats and beagle dogs. J Clin Pract. 2014: 4:3.  Microscopic Observations in Heart  Valvulopathy  Atrium—Mixed cell infiltrate  Myocardium—Degeneration/necrosis  Myocardium—Hemorrhage  Myocardium—Mixed cell infiltrate, base  Coronary artery—Necrosis with inflammation  Cardiomyocyte—Necrosis/inflammatory cell infiltrate  CONTROL  Vehicle  iv, qwk x 4  LY2109761  300 mg/kg  po, qd x 8  PanTGFβAb  30 mg/kg  Iv, 1 dose  10 mg/kg  iv, qwk x 4  30 mg/kg  iv, qwk x 4  100 mg/kg  iv, qwk x 4  SRK-181  Selective TGFβ-1 Toxicity: Minimal  Non-selective TGFβ  Toxicity: Minimal, slight and moderate  Unremarkable  Minimal  Slight  Moderate  Toxicology:  Repeat Dose Pilot Toxicology Study  Adult female Sprague Dawley rats  Cardiac findings were exhibited  in animals dosed with pan-TGFβ antibody or LY2109761 (inhibitor of ALK5, common TGFβ receptor kinase) as expected based on published data†  NO CARDIOTOXICITIES (valvulopathy)were noted with SRK-181   NOAEL for SRK-181: 100 mg/kg QW(highest dose evaluated)  4-week GLP toxicology studies  RATS  NOAEL for SRK-181: 200 mg/kg QW (highest dose evaluated)  NON-HUMAN PRIMATES  NOAEL for SRK-181: 300 mg/kg(highest dose evaluated)  Not test article related 
 

 Bladder urothelial carcinoma  Had & neck squamous carcinoma  Kidney clear cell carcinoma  Kidney papillary cell carcinoma  Liver hepatocellular carcinoma  Lung adenocarcinoma  Skin cutaneous melanoma  Stomach adenocarcinoma  Acute myeloid leukemia  Adrenocortical cancer  Brain lower grade glioma  Breast invasive carcinoma  Cervical & endocervical cancer  Cholangiocarcinoma  Colon adenocarcinoma  Diffuse large B-cell carcinoma  Esophageal carcinoma  Glioblastoma multiforme  Kidney chromophobe  Lung squamous cell carcinoma  Mesothelioma  Ovarian serous cystadenocarcinoma  Pancreatic adenocarcinoma  Pheochromocytoma & paraganglioma  Prostate adenocarcinoma  Emerging Evidence Implicates TGFβ-1 as Driving Primary Resistance to Checkpoint Inhibitors  73  Human Tumor Analyses Reveal TGFβ-1 as Most Likely Driver of TGFβ Signaling Pathway in Cancers  †Priti H, et al. Top 10 challenges in cancer immunotherapy. Immunity. 2020 Jan 14:52(1):17-35. https://doi.org/10.1016/j.immuni.2019.12.011.  *Source: National Cancer Institute - Cancer Genome Atlas Program.  NSCLC  TMB  Low  High  Substantial % of Solid Tumors Exhibit Immune Exclusion  Immune Excluded  Immune Phenotype  Inflamed  Immune Desert  Tumor Type  Melanoma  RCC  UBC  TNBC  Gastric  CRC MSS  Pancreatic  SCLC  HR+BC  Prostate  Immune Excluded  Inflamed  Immune Desert  Cancer Genome Atlas RNAseq Analysis of >10,000 Samples Spanning 33 Tumor Types*  TGFB1  TGFB2  TGFB3  Scale  % of patient samples (+)  for TGF-β isoform  100  80  60  40  20  0 
 

 Biomarker Strategies Employed in DRAGON Trial  74  Multiple tissue-based and circulating biomarker analyses to be evaluated in DRAGON study  Higher resolution histochemical characterization of tumor immune contexture (e.g. CD8+)  Classification of inflamed, excluded or immune desert tumors and tumor nests  Ability of SRK-181 to overcome tumor immune exclusion  Analysis of immune response markers (e.g. PD-L1)  Changes to intra-tumoral and/or circulating immune cell contexture (MDSC)  Show evidence of the SRK-181 target engagement  e.g. circulating TGFβ-1 levels  TGFβ pathway modulation:  e.g. Histochemical analysis of pSMAD  e.g. RNA-based TGFβ gene signatures and pathway analyses  Paired biopsies from the head and neck cohort allow for a potential to accelerate the development path  Immunophenotyping  Assessment of immune landscape  TGFβ-1 pathway evaluation  Assessment of signaling pathway 
 

 Clear Evidence of Target EngagementPharmacodynamic Biomarker Results for Part A: Circulatory TGFβ-1  75  Yap T et al. SRK-181, a latent TGFβ1 inhibitor: safety, efficacy, and biomarker results from the dose escalation portion of a phase I trial (DRAGON trial) in patients with advanced solid tumors (Poster 780); Presented at SITC; Nov. 10-11, 2022. Circulatory TGFβ-1 and PF4 levels were quantitated by using validated ELISA kits from R&D System.12 Because platelet activation during sample processing can lead to elevated TGFβ-1 levels, samples with elevated PF4, a platelet activation biomarker, were excluded from the analysis based on a preliminary cutoff value.  Pre-infusion.  SRK-181 is an investigational drug candidate that is being evaluated for the treatment of cancer. SRK-181 has not been approved by the US FDA or any other health authority, and its safety and efficacy have not been established.  Binding to latent TGFβ-1 delays maturity state allowing TGFβ-1 to accumulate in system  Combination treatment with pembrolizumab did not appear to impact circulatory TGFβ-1 levels  C1D2  C1D8  C1D15  C2D1*  C3D1*  C3D2  C3D8  C3D15  C4D1*  C5D1*  Baseline  Circulatory TGFβ-1  (Fold-Change from Baseline)  2  3  4  6  5  Median Circulatory TGFβ-1 Increased Post-treatment with SRK-181 (Q3W, All Patients) 
 

 Preclinical Data Provide Scientific Rationale to Evaluate Peripheral Samples for Evidence of SRK-181 Activity  76  MBT-2 bladder tumor model IgG, anti-PD-1 and SRK-181-mIgG1 dosed d1, d7  Analysis on day 10  Circulating MDSC Levels Correlate  with Tumor Volume  Both tumoral and circulatory MDSC are being evaluated in the DRAGON study  Immunophenotyping  Assessment of immune landscape  Measurement of MDSCs in circulation may provide indirect evidence of drug action on the tumor  Myeloid-derived suppressor cells (MDSCs) have immune suppressive functions  SRK-181 plus anti-PD1 combination drive MDSC levels down significantly in the tumor microenvironment  Reductions in circulating MDSC levels correlate with reduced tumor volume following SRK-181 and anti-PD1 treatment in MBT-2 tumor model  Anti-PD-1+  SRK-181   1 mpk   3 mpk   10 mpk  IgG Ctrl  SRK-181  Anti-PD-1