8-K

 

 

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): February 7, 2019

 

 

RESTORBIO, INC.

(Exact name of registrant as specified in its charter)

 

 

 

Delaware   001-38359   81-3305277

(State or other jurisdiction

of incorporation)

 

(Commission

File Number)

 

(I.R.S. Employer

Identification No.)

500 Boylston Street, 12th Floor

Boston, MA 02116

(Address of principal executive offices, including zip code)

(857) 315-5521

(Registrant’s telephone number, including area code)

Not Applicable

(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))

Indicate by check mark whether the registrant is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (§ 230.405 of this chapter) or Rule 12b-2 of the Securities Exchange Act of 1934 (§ 240.12b-2 of this chapter).

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.

resTORbio, Inc. (the “Company”) from time to time presents and/or distributes to the investment community at various industry and other conferences slide presentations to provide updates and summaries of its business. A copy of its current corporate slide presentations (the “Presentations”) are attached to this Current Report on Form 8-K as Exhibits 99.1 and 99.2. The Company undertakes no obligation to update, supplement or amend the materials attached hereto as Exhibit 99.1 and 99.2.

Exhibit 99.2 to this Current Report Form 8-K shall not be deemed “filed” for 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 it be deemed incorporated by reference in any filing under the Securities Act of 1933, as amended, or the Exchange Act, except as expressly set forth by specific reference in such a filing.

 

Item 9.01.

Financial Statements and Exhibits.

(d) Exhibits

 

Exhibit
No.
  

Description

99.1    Corporate slide presentation of resTORbio, Inc., dated February 7, 2019.
99.2    Corporate slide presentation of resTORbio, Inc., dated February 7, 2019

*    *    *


SIGNATURES

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.

 

    resTORbio, Inc.
Date: February 7, 2019     By:  

/s/ Chen Schor

      Chen Schor
      President and Chief Executive Officer
EX-99.1

Slide 1

February 2019 Targeting the biology of aging to prevent and treat aging-related diseases Exhibit 99.1


Slide 2

This presentation may contain “forward-looking statements” within the meaning of the Private Securities Litigation Reform Act of 1995, including, but not limited to, statements regarding the safety, efficacy and regulatory and clinical progress of our product candidates, including RTB101 alone and in combination with everolimus or sirolimus. 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. The use of words such as “may,” “might,” “will,” “should,” “expect,” “plan,” “anticipate,” “believe,” “estimate,” “project,” “intend,” “future,” “potential,” or “continue,” and other similar expressions are intended to identify forward-looking statements. Forward-looking statements are neither historical facts nor assurances of future performance. Instead, they are based on our current beliefs, expectations and assumptions regarding the future of our business, future plans and strategies, our clinical results and other future conditions. All statements other than statements of historical facts contained in this presentation, including statements regarding future results of operations and financial position, business strategy, current and prospective product candidates, planned clinical trials and preclinical activities, including the initiation, timing, progress and results of our preclinical and clinical studies and our research and development programs, product approvals, research and development costs, current and prospective collaborations, timing and likelihood of success, including our ability to advance RTB101 alone and in combination with everolimus or sirolimus into, and successfully complete, clinical studies, timing of the end-of-Phase 2 meeting with the U.S. Food and Drug Administration, and the timing or likelihood of regulatory filings and approvals, expectations regarding market acceptance and size, plans for launch and commercialization, plans and objectives of management for future operations, and future results of anticipated product candidates, are forward-looking statements. New risks and uncertainties may emerge from time to time, and it is not possible to predict all risks and uncertainties. No representations or warranties (expressed or implied) are made about the accuracy of any such forward-looking statements. These statements are also subject to a number of material risks and uncertainties that are discussed in the section entitled "Risk Factors" in resTORbio’s annual report on Form 10-K for the fiscal year ended December 31, 2017, as well as discussions of potential risks, uncertainties, and other important factors in resTORbio's subsequent filings with the Securities and Exchange Commission. Any forward-looking statement speaks only as of the date on which it was made. Neither we, nor our affiliates, advisors, or representatives, undertake any obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law. Certain information contained in this presentation relates to or is based on studies, publications, surveys and other data obtained from third-party sources and the Company’s own internal estimates and research. While we believe these third-party sources to be reliable as of the date of this presentation, we have not independently verified, and we make no representation as to the adequacy, fairness, accuracy or completeness of, any information obtained from third-party sources. In addition, all of the market data included in this presentation involves a number of assumptions and limitations, and there can be no guarantee as to the accuracy or reliability of such assumptions. Finally, while we believe our own internal research is reliable, such research has not been verified by any independent source. Forward-looking statements Needs legal review


Slide 3

resTORbio highlights Targeting the biology of aging Lead clinical candidate, RTB101, is the most advanced selective TORC1 inhibitor TORC1 inhibition improves the function of aging organ systems including the immune, neurologic, and cardiovascular systems Positive results in Phase 2b to improve immune function and reduce the incidence of RTIs 30.6% reduction in the percentage of patients with laboratory-confirmed RTIs (p=0.025) 52.1% reduction in percentage of patients with severe laboratory-confirmed RTI symptoms (p=0.034) Successfully defined dose and patient population for Phase 3 program End-of-Phase 2 meeting with the FDA expected in 1Q19; Plan to initiate Phase 3 program in 1H19 RTIs are the 4th most common cause of hospitalization in people 65+; 2nd in 85+ (US) Data-driven approach to expand into additional aging-related indications Improving neurologic function: Plan to initiate Phase 1b/2a study in Parkinson’s disease in 1Q19 Building a pipeline targeting multiple mechanisms underlying aging RTI = respiratory tract infection


Slide 4

Aging is the biggest risk factor for most chronic diseases Why does this happen? Aging is not just due to random wear and tear Aging is biology that may be targeted with medicines Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2017 (GBD 2017) Results


Slide 5

Targeting the biology of aging AGING Accumulation of Senescence Cells TORC1 Activity NAD Depletion Epigenetic Changes DNA Damage Mitochondrial Dysfunction Stem Cell Exhaustion


Slide 6

The TORC1 pathway Source: Lamming, Dudley W., et al. (2013) Journal of Clinical Investigation123 (3): 980–989. TORC1 inhibition extended lifespan and healthspan in multiple species Mice Flies Worms Yeast TORC1 is an evolutionarily conserved pathway that regulates aging


Slide 7

Species Genetic Manipulation to Inhibit mTOR Yeast SCH9 (Akt/S6K homolog) insertional mutant 1 SCH9 (Akt/S6K homolog) deletion 1 SCH9 (Akt/S6K homolog) insertional mutant 2 SCH9 (Akt/S6K homolog) deletion 2 TOR1 deletion 3 TOR1 deletion 4 C. elegans TOR (let-363) RNAi 5 Raptor (daf-15) heterozygous 6 S6K (rsks-1) RNAi 7 S6K (rsks-1) deletion mutant 7 TOR (let-363) RNAi 7 S6K (rsks-1) RNAi 8 S6K (rsks-1) deletion mutant 8 TOR (let-363) RNAi 8 Raptor (daf-15) RNAi 9 RagGTPase (raga-1) RNAi 9 RagGTPase (raga-1) RNAi 9 Rheb (rheb-1) RNAi 9 D. melanogaster dTSC1 overexpression 10 dTSC2 overexpression 10 dTOR FRB domain (dominant negative) 10 dS6K dominant negative 10 DTOR mutant (hypomorph) 11 d4E-BP overexpression 12 d4E-BP weak activated 12 d4E-BP strong activated 12 M. musculus Loss of S6K1 13 Mtor+/-MIst8+/- genotype 14 Extensive genetic validation that TORC1 inhibition extends lifespan across species Corresponding citations can be found on slide 39


Slide 8

TORC1 inhibitors extend lifespan in mice even when started late in life and given intermittently Daily Dosing Intermittent Dosing Once Every 5 Days (1) (2) Harrison et al. (2009) Nature, 460:392-396 Arriola Apelo et al. (2016) Gerontol A Biol Sci Med Sci, 71: 876–88


Slide 9

TORC1 may become dysregulated and overactive in some aging organ systems TORC1 activity (pS6 in liver) Fed Fasted Young Mice Old Mice Decreasing TORC1 activity may upregulate protective pathways and may have benefits in aging-related diseases Fed Fasted Total S6 Feeding activates TORC1 leading to increased protein and lipid synthesis Fasting inhibits TORC1 leading to upregulation of protective pathways TORC1 activity remains aberrantly elevated during fasting , preventing upregulation of protective pathways Sengupta et al., Nature 2010


Slide 10

TORC1 inhibition may improve the function of multiple aging organ systems Improvement in physical activity Selman et al., Science, 2011 Harrison et al., Nature, 2009 Wilkinson et al., Aging Cell, 2014 Flynn et al., Aging Cell, 2013 Reversal of aging-related immune dysregulation Chen et al., Science Sig, 2009 Selman et al., Science, 2011 Neff et al., JCI, 2013 Hurez et al., Aging Cell, 2015 Reversal of aging-related cardiac dysfunction Flynn et al., Aging Cell, 2013 Dai et al., Aging Cell, 2014 Chiao et al., Aging, 2016 Improved Neurologic Function Tain et al., Nature Neuroscience, 2009 Malagelada et al., J Neurosci, 2010 Spilman et al., PLoS ONE, 2010 Halloran et al., Neuroscience, 2012 Majumder et al., Aging Cell, 2012 Neff et al., JCI, 2013


Slide 11

Additional Aging-Related Target Most advanced pipeline targeting aging-related diseases RTB101 Respiratory Tract Infections Parkinson’s Disease Program Indication Phase 1 Phase 2 Phase 3 Discovery Heart Failure with Preserved Ejection Fraction Preclinical Urinary Tract Infections *For heart failure with preserved ejection fraction, Parkinson’s Disease and certain other infections, we may be required to file an investigational new drug application, or IND, prior to initiating Phase 2 clinical trials. We expect to have the ability to initiate these Phase 2 clinical trials without the need to conduct prior Phase 1 trials. End-of-Phase 2 meeting planned for 1Q19 Initiate Phase 1b/2a in 1Q19* RTB101+ sirolimus Additional TORC1 Inhibitor Undisclosed RTB101 RTB101 RTB101 + rapalog Undisclosed Current Indications Potential Indications Discovery


Slide 12

TORC1 Pathway


Slide 13

mTOR Selective inhibition of TORC1 may have therapeutic benefit for the treatment of aging-related diseases Inhibition of TORC2 by genetic mutation decreases lifespan and causes hyperglycemia and hyperlipidemia in mice (Science, 2012; Aging Cell, 2014) S6K Ulk1 4EBP1 Atg Inhibition of TORC1 by genetic mutation extends lifespan (Science, 2012) Knock out of S6K extends lifespan and healthspan (Science, 2009) Overexpression extends lifespan (Cell, 2009) Transgenic overexpression extends lifespan (Nat Comm, 2013) TORC2 TORC1 4EBP1


Slide 14

S6K Ulk1 TORC1 everolimus or sirolimus 4EBP1 S6K Ulk1 4EBP1 TORC1 RTB101 + everolimus or sirolimus 4EBP1 S6K Ulk1 TORC1 4EBP1 RTB101 Inhibiting the phosphorylation of 1 target of TORC1 Inhibiting the phosphorylation of 2 targets of TORC1 Inhibiting the phosphorylation of 3 targets of TORC1 Spectrum of TORC1 inhibition with RTB101 and rapalog = indicates phosphorylation is inhibited


Slide 15

Improving Immune Function Respiratory Tract Infections (RTIs)


Slide 16

Results of Phase 2a trial 264 mostly healthy elderly people randomized to the following TORC1 inhibitor treatment arms (all doses were QD): Both RTB101 10mg QD and RTB101 10mg + everolimus 0.1mg QD significantly reduced the incidence of all infections as well as respiratory tract infections (RTIs) Reduction in RTIs: RTB101 10mg: 42% reduction (p=0.006) RTB101 10mg + everolimus 0.1mg: 36% reduction (p=0.01) Both RTB101 10mg and RTB101 10mg + everolimus 0.1mg upregulated antiviral gene expression in whole blood TORC1 inhibition QD = once daily Everolimus 0.1mg + RTB101 10mg RTB101 10mg Everolimus 0.5mg Everolimus 0.1mg Placebo


Slide 17

IMMUNOTHERAPY: RTB101 alone or in combination with everolimus RTB101 offers new approach to harnessing the immune system to target multiple pathogens Source: S. Jain et al., NEJM 2015 Indicates the annual number of pathogen-specific pneumonia hospitalizations per 10,000 adults ≥ 80 The majority of pathogens detected in elderly people hospitalized for pneumonia are viruses for which NO APPROVED THERAPIES are currently available Viruses with no FDA-approved therapies available * Incidence per 10,000 Persons 5 10 20 15 0 *Adenovirus L. pneumophila S. aureus M. pneumoniae *Coronavirus *Respiratory Syncytial Virus *Parainfluenza Virus *Human Meta-Pneumovirus S. Pneumoniae Influenza A or B *Human Rhinovirus


Slide 18

Phase 2a to Phase 2b: Population, primary endpoint and dosing duration were modified Self-reported RTIs Laboratory-Confirmed RTIs 6 weeks 16 weeks Healthy, 65 and older POPULATION: PRIMARY ENDPOINT: DOSING DURATION: Phase 2a Phase 2b Goal: Define patient population for Phase 3 program


Slide 19

Phase 2b design Primary Endpoint: Reduction in the percentage of patients with laboratory-confirmed RTIs through week 16 Population: Elderly subjects at increased risk of RTI-associated morbidity and mortality, defined as: ≥ 85 years of age 65-84 years of age with one or more comorbidities including: Asthma Chronic obstructive pulmonary disease (COPD) Type 2 diabetes mellitus (T2DM) Current smoker 16 weeks 8 weeks RTB101 5mg QD RTB101 10mg QD Placebo Follow-up Northern Hemisphere (n=473) RTB101 10mg + everolimus 0.1 mg QD RTB101 10mg QD Placebo 16 weeks 8 weeks Follow-up RTB101 10mg BID Southern Hemisphere (n=179) QD = once daily; BID = twice daily Interim Analysis


Slide 20

Dosing regimens in Phase 2b estimated to result in different duration and spectrum of TORC1 inhibition S6K Ulk1 4EBP1 TORC1 4EBP1 RTB101 5mg QD Intermittent inhibition of two TORC1 targets RTB101 10mg QD 0 12 24 Estimated duration of TORC1 inhibition (hrs) RTB101 10mg BID Persistent inhibition of two TORC1 targets RTB101 10mg + everolimus 0.1mg QD Persistent inhibition of three TORC1 targets 0 12 24 0 12 24 QD = once daily; BID = twice daily


Slide 21

Odds ratio supports dose selection and potential efficacy of RTB101 10mg QD 21/61 26/60 0.618 0.109 34/176 50/180 0.601 0.025* 25/120 24/120 1.050 0.560 25/115 24/120 1.187 0.700 Active incidence1 Placebo incidence1 Odds ratio2 p-value3 1No. of patients in cohort with one or more laboratory-confirmed RTIs/No. of patients in cohort; 2Odds ratio represents the odds of experiencing one or more laboratory-confirmed RTIs in the active treatment group versus the placebo group; 3One-sided p-value; *p<0.05; QD = once daily; BID = twice daily Odds ratio (90% confidence interval) Odds ratio of experiencing lab-confirmed RTIs through Week 16 RTB101 5mg QD RTB101 10mg QD RTB101 10mg BID RTB101 10mg + everolimus 0.1mg 0.5 RTB101 10mg QD demonstrated a 30.6% reduction in the percentage of patients with laboratory-confirmed RTIs compared to placebo


Slide 22

RTB101 10mg QD showed consistent benefit in multiple pre-specified analyses of lab-confirmed RTIs Odds ratio of experiencing lab-confirmed RTIs through Week 16 – primary endpoint Odds ratio of experiencing severe lab-confirmed RTI symptoms through Week 16 Odds ratio of experiencing lab-confirmed RTIs through Week 24 Odds ratio (90% confidence interval) One-sided p-value; QD = once daily; BID = twice daily; Odds ratio represents the odds of experiencing one or more event in the active treatment group versus the placebo group RTB101 10mg QD RTB101 5mg QD RTB101 10mg BID RTB101 10mg + everolimus 0.1mg RTB101 5mg QD RTB101 10mg QD RTB101 10mg BID RTB101 10mg + everolimus 0.1mg RTB101 5mg QD RTB101 10mg QD RTB101 10mg BID RTB101 10mg + everolimus 0.1mg OR=0.601; p=0.025 OR=0.437; p=0.034 OR=0.623; p=0.030 RTB101 10mg QD associated with statistically significant reductions across three different analyses of laboratory-confirmed RTIs: Week 16, severe RTIs and Week 24


Slide 23

COPD and smokers were non-responding subgroups (pre-specified analyses) 85+: % subjects with 1 or more laboratory-confirmed RTIs Asthma T2DM COPD Smokers 65+: % subjects with 1 or more laboratory-confirmed RTIs Southern Hemisphere Northern Hemisphere Southern & Northern Hemispheres (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) (%) -66.7% (p=0.052) -66.8% (p=0.053) -66.7% (**p=0.007) -68.1% (p=0.001) -69.4% (p=0.016) -68.9% (p=0.0001) -40.5% (p=0.087) -15.4% (p=0.077) -25.3% (*p=0.028) 2.1% (p=0.168) 3.3% (p=0.147) 410.7% (p=0.874) 0.0% (p=0.324) 201.2% (p=0.833) = placebo cohort; = RTB101 10mg once daily cohort; One-sided p-value; QD = once daily; BID = twice daily 311.9% (p=0.936)


Slide 24

mTOR inhibition decreased airway inflammation in asthma and increased airway inflammation due to smoking Asthma Smoking mTOR inhibition with rapamycin (Rapa) significantly decreased airway inflammation in a preclinical asthma model in which mice were exposed to intranasal house dust mites (HDM)1 Disruption of mTOR selectively in bronchial epithelial cells (mBE-mtor-/-) significantly increased cigarette smoke (CS)-induced lung inflammation in a COPD model in which mice were exposed to cigarette smoke for 6 months2 1Mushaben E. M. et al., J Immunol 2011:187:5756-5763; 2 Wang Y et al., J Immunol 2018;200:2571-2580; *p<0.05, **p<0.01


Slide 25

A significant reduction in the incidence of laboratory-confirmed RTIs was observed in subjects 65+ (excluding smokers/COPD patients) 1No. of patients in cohort with one or more laboratory-confirmed RTIs/No. of patients in cohort; 2Odds ratio represents the odds of experiencing one or more laboratory-confirmed RTIs in the active treatment group versus the placebo group; 3One-sided p-value; ***p<0.0001; QD = once daily; BID = twice daily 15/41 21/42 0.560 0.1000 15/113 36/117 0.319 0.0007*** 14/83 15/75 0.771 0.4700 18/78 15/75 1.147 0.2700 Active incidence1 Placebo incidence1 Odds ratio2 p-value3 Odds ratio (90% confidence interval) Odds ratio of experiencing lab-confirmed RTIs through Week 16 RTB101 5mg QD RTB101 10mg QD RTB101 10mg BID RTB101 10mg + everolimus 0.1mg


Slide 26

Phase 2 clinical studies enrolling > 900 elderly subjects demonstrate potential efficacy of RTB101 10mg QD QD = once daily Study Patient population RTB101 10mg QD efficacy Phase 2a 264 healthy elderly 42% reduction in the rate of RTIs (p=0.006) Phase 2b 652 high-risk elderly 30.6% reduction in the percent of patients with lab-confirmed RTIs (p=0.025) 56.9% reduction in the percent of patients with lab-confirmed RTIs, excluding smokers/COPD patients (p=0.007)


Slide 27

RTB101 was well-tolerated in high-risk elderly patients through Week 24 Adverse events (AEs) were balanced between the RTB101 10mg QD and placebo cohorts 1 unrelated death occurred in the RTB101 10mg QD cohort (patient was hit by car while riding a bicycle), 1 unrelated death occurred in the RTB101 10mg BID cohort and 1 unrelated death occurred in the placebo cohort (both from unknown causes) RTB101 10mg QD Placebo Serious AEs (% of patients) 4.5% 7.8% Discontinued study drug due to an AE (% of patients) 5.1% 5.6% Number of severe AEs 12 25 QD = once daily; BID = twice daily


Slide 28

Market Opportunity in RTIs


Slide 29

Mortality from RTIs is higher than mortality from colorectal, pancreatic, breast or prostate cancer1 RTIs are the 4th most common cause for hospitalization in 65+ (2nd in 85+)2 RTIs cause the majority of asthma exacerbations in the elderly3 RTIs in the elderly represent a significant healthcare burden The majority of RTIs are caused by viruses for which there are no approved therapies4 Decreasing the incidence of RTIs in the elderly may significantly decrease health care costs Sources: 12017, NCHS, National Vital Statistics System, Mortality; 2015, American Cancer Society, Inc, Surveillance Research; 2Pfunter, A (2013) HCUP Statistical Brief #162; 3 BMJ, 1995, 310:1225-1228; BMJ, 1993, 307:982-986; 4 Am J Med. 2002. 112(6A):42S-49S


Slide 30

US EU5 JP CN # Elderly People without COPD and who are non-smokers* 40M 53M 29M 77M Elderly (65-84 years old) with asthma: 3.2M 3.3M1 2.1M2 2.5M3 Very elderly (85+ years old): 6.5M 9.3M 5.5M 8.9M Estimated number of elderly that may benefit from RTB101 in key geographies *Based on references provided on slide 40; 1Based on estimated percentage of asthmatics in older adults in high-income countries. 2Based on percentage of asthmatics in the Japanese adult population. 3Based on percentage of adults age ≥60 on asthma medication in Jinan province; Likely underestimated due to low diagnosis rate of asthma


Slide 31

% Reduction in RTI Estimated % prescribed in patients (patient-weighted means) ≥85 65-84 with asthma 65-84 with comorbidities 25% 33% 36% 36% 33% 41% 44% 47% 40% 46% 48% 51% Survey of 100 physicians to determine potential usage in the target patient populations Medical Specialty Geriatrics 25 Primary Care 50 Pulmonologist 25 Practice characteristics Years practicing medicine Avg 19 (median 19.5, range 6-33) # pts ≥ 65 seen/month Avg 250 (median 220, range 80-600) % services billed to Medicare Avg 63% (median 65%, range 30-100%) *Respondent background (n=100): Physician survey*: Expected use in target populations


Slide 32

Ameliorating Neurodegenerative Diseases GBA Parkinson’s Disease


Slide 33

GBA mutation in Parkinson’s disease (PD) leads to a-synuclein aggregation and neuronal cell death Disease cascade: GBA is a gene encoding the lysosomal enzyme glucocerebrosidase (GCase) Mutant GCase may contribute to PD pathogenesis through a loss or gain of function: Loss of function: Decreased GCase activity leading to an accumulation of its lipid substrate glucosylceramide (GL1) that disrupts lysosomal function1 Gain of function: Accumulation of misfolded GCase aggregates that disrupt lysosomal function2 Disruption of lysosomal function prevents clearance of aggregated a-synuclein and leads to neuronal death Loss of Function Gain of Function 1Mazzulli, J. R., et al. (2011). Cell 146(1): 37-52; 2Cullen,V., et al. (2011). Annals of Neurology 69(6): 940-953. GBA = glucocerebrosidase; a-syn = alpha-synuclein Lysosomal Dysfunction Mutant GCase Accumulation of GL1 Accumulation of GCase Aggregates a-synuclein aggregation and neuronal death


Slide 34

resTORbio GBA PD program potential benefits to GBA PD patients (both loss or gain of function GBA mutations) Gain of Function 1Guri, Y., et al. (2017). Cancer Cell 32(6): 807-823; 2Decressac, M., et al. (2013). Proc Natl Acad Sci USA 110(19): E1817-1826; 3 Cullen,V., et al. (2011). Ann Neurol 69(6): 940-953; 4Kinghorn, K.J., et al. (2016). J Neurosci 36(46): 11654-11670; 5Roczniak-Ferguson, A., et al. (2012). Sci Signal 5(228): ra42. Lysosomal Dysfunction Accumulation of GCase Aggregates a-syn aggregation and neuronal death Loss of Function Lysosomal Dysfunction Accumulation of GL1 a-syn aggregation and neuronal death resTORbio Decreased GL1 synthesis through mTOR inhibition1 Clearance of GCase aggregates by activating ULK1 and inducing autophagy2,3,4 Increased lysosomal biogenesis by activating TFEB5 Clearance of a-synuclein aggregates by activating ULK1 and inducing autophagy2,3,4


Slide 35

Phase 1b/2a Parkinson’s disease trial design Study initiation planned for 1Q19 Cohort RTB 101 dose (mg) Sirolimus dose (mg) 1 300 0 2 0 2 3 300 2 4 300 4 5 300 6 Design Randomized, Placebo-Controlled Phase 1b/2a Study (4-week dosing) Mild PD patients (mH&Y I-II) with or without GBA mutations On standard of care PD drugs Once weekly dosing Study Size N=45 (2:1 randomization) Key Endpoints Primary endpoint: Safety and tolerability Secondary endpoint: Exposure in blood, plasma and CSF Exploratory endpoints: Biomarkers in plasma and CSF Clinical assessments, wearables or matching placebo


Slide 36

Near term milestones and financials Q1 2019 End of Phase 2 meeting with the FDA Q1 2019 Initiate Phase 1b/2a study in Parkinson’s disease H1 2019 Initiate Phase 3 program for reducing the incidence of RTIs Milestones Cash, cash equivalents and marketable securities were ~$115 million as of September 30, 2018 Financials


Slide 37

resTORbio highlights Targeting the biology of aging Lead clinical candidate, RTB101, is the most advanced selective TORC1 inhibitor TORC1 inhibition improves the function of aging organ systems including the immune, neurologic, and cardiovascular systems Positive results in Phase 2b to improve immune function and reduce the incidence of RTIs 30.6% reduction in the percentage of patients with laboratory-confirmed RTIs (p=0.025) 52.1% reduction in percentage of patients with severe laboratory-confirmed RTI symptoms (p=0.034) Successfully defined dose and patient population for Phase 3 program End-of-Phase 2 meeting with the FDA expected in 1Q19; Plan to initiate Phase 3 program in 1H19 RTIs are the 4th most common cause of hospitalization in people 65+; 2nd in 85+ (US) Data-driven approach to expand into additional aging-related indications Improving neurologic function: Plan to initiate Phase 1b/2a study in Parkinson’s disease in 1Q19 Building a pipeline targeting multiple mechanisms underlying aging RTI = respiratory tract infection


Slide 38

February 2019 Targeting the biology of aging to prevent and treat aging-related diseases


Slide 39

Extensive genetic validation that TORC1 Inhibition extends lifespan across species 1 Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD. Regulation of longevity and stress resistance by Sch9 in yeast. Science. 2001;292(5515):288–290. Fabrizio P, Pletcher SD, Minois N, Vaupel JW, Longo VD. Chronological aging-independent replicative life span regulation by Msn2/Msn4 and Sod2 in Saccharomyces cerevisiae. FEBS Lett. 2004; 557(1–3):136–142. Kaeberlein M, et al. Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients. Science. 2005;310(5751):1193–1196. Bonawitz ND, Chatenay-Lapointe M, Pan Y, Shadel GS. Reduced TOR signaling extends chronological life span via increased respiration and upregulation of mitochondrial gene expression. Cell Metab. 2007; 5(4):265–277. Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, Muller F. Genetics: influence of TOR kinase on lifespan in C. elegans. Nature. 2003;426(6967):620. Jia K, Chen D, Riddle DL. The TOR pathway inter- acts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development. 2004;131(16):3897–3906. Hansen M, Taubert S, Crawford D, Libina N, Lee SJ, Kenyon C. Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans. Aging Cell. 2007;6(1):95–110. Pan KZ, et al. Inhibition of mRNA translation extends lifespan in Caenorhabditis elegans. Aging Cell. 2007;6(1):111–119. Robida-Stubbs S, et al. TOR Signaling and rapamy- cin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metab. 2012;15(5):713–724. Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol. 2004;14(10):885–890. Luong N, et al. Activated FOXO-mediated insulin resistance is blocked by reduction of TOR activity. Cell Metab. 2006;4(2):133–142. Zid BM, et al. 4E-BP extends lifespan upon dietary restriction by enhancing mitochondrial activity in Drosophila. Cell. 2009;139(1):149–160. Selman C, et al. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science. 2009;326(5949):140–144. Lamming DW, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012; 335(6076):1638–1643.


Slide 40

References for number of elderly people without COPD and who are non-smokers US: 4,038,000 elderly people estimated to be smokers in the US. U.S. NHIS 2017, https://www.cdc.gov/nchs/nhis/SHS/tables.htm (accessed Jan 5, 2019), Table A-12b. Prevalence of COPD in the elderly estimated at 14.2%. Hanania, N. et al, 2010 “COPD in the Elderly Patient” https://www.medscape.com/viewarticle/730813_2 (accessed Jan 5, 2019) 14.1% of current smokers were assumed to have COPD. Cunningham, T.J., et al., COPD, 2015. 12(3): p. 276-86. Size of U.S. elderly population estimated at 50,858,679 in 2017. U.S. Census Bureau. https://factfinder.census.gov/faces/nav/jsf/pages/index.xhtml? (accessed Jan 5, 2019) Europe: Smoking prevalence in the elderly in each European country. Eurostat database, http://appsso.eurostat.ec.europa.eu/nui/show.do?dataset=hlth_ehis_sk3e&lang=en (accessed Jan 5, 2019) COPD prevalence in the elderly estimated at 14.2%. Raherison, C. and P.O. Girodet, Epidemiology of COPD. Eur Respir Rev, 2009. 18(114): p. 213-21. 29.6% & 16.1% of COPD patients aged 65-75 & 75 and over, respectively estimated to be current smokers. Worth, H., et al., The 'real-life' COPD patient in Germany: The DACCORD study. Respir Med, 2016. 111: p. 64-71. Size of elderly population in each European country. UN Data, United Nations Statistics Division, http://data.un.org/Data.aspx?d=POP&f=tableCode%3A22 (accessed Jan 5, 2019) Japan: Smoking prevalence in people aged 60 and over in Japan (21.2% in men, 5.6% in women). Japan Tobacco Inc., JT’s Annual Survey Finds 18.2 % of Japanese Adults Are Smokers. 2017. https://www.jt.com/media/news/2017/pdf/20170727_E02.pdf (accessed Jan 5, 2019) COPD prevalence in people aged 60 and over in Japan (11.5% of men, 5.8% of women); 17% & 0% of male and female current smokers, respectively, estimated to have COPD. Takemura, H., et al., Prevalence of COPD in Japanese People on Medical Check-Up. Journal of Experimental Medicine, 2005. 207: p. 41-50. Size of elderly population estimated at 35,228,000 (15,294,000 men, 19,933,000 women). E-Stat, Portal Site of Official Statistics of Japan. https://www.e-stat.go.jp/en/stat-search/files?page=1&layout=datalist&toukei=00200524&tstat=000000090001&cycle=1&year=20180&month=12040606&tclass1=000001011678 (accessed Jan 5, 2019)China: China: Smoking prevalence in the elderly in China estimated at 22.7%. Li, Q., J. Hsia, and G. Yang, Prevalence of Smoking in China in 2010. New England Journal of Medicine, 2011. 364(25): p. 2469-2470. COPD prevalence in the elderly; COPD prevalence among current smokers. Fang, L., et al., Chronic obstructive pulmonary disease in China: a nationwide prevalence study. The Lancet Respiratory Medicine, 2018. 6(6): p. 421-430. Size of elderly population in China by age. https://www.populationpyramid.net/china/2016/ (accessed Jan 5, 2019)

EX-99.2

Slide 1

February 7, 2019 Targeting the biology of aging to prevent and treat aging-related diseases Exhibit 99.2


Slide 2

This presentation may contain “forward-looking statements” within the meaning of the Private Securities Litigation Reform Act of 1995, including, but not limited to, statements regarding the safety, efficacy and regulatory and clinical progress of our product candidates, including RTB101 alone and in combination with everolimus or sirolimus. 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. The use of words such as “may,” “might,” “will,” “should,” “expect,” “plan,” “anticipate,” “believe,” “estimate,” “project,” “intend,” “future,” “potential,” or “continue,” and other similar expressions are intended to identify forward-looking statements. Forward-looking statements are neither historical facts nor assurances of future performance. Instead, they are based on our current beliefs, expectations and assumptions regarding the future of our business, future plans and strategies, our clinical results and other future conditions. All statements other than statements of historical facts contained in this presentation, including statements regarding future results of operations and financial position, business strategy, current and prospective product candidates, planned clinical trials and preclinical activities, including the initiation, timing, progress and results of our preclinical and clinical studies and our research and development programs, product approvals, research and development costs, current and prospective collaborations, timing and likelihood of success, including our ability to advance RTB101 alone and in combination with everolimus or sirolimus into, and successfully complete, clinical studies, timing of the end-of-Phase 2 meeting with the U.S. Food and Drug Administration, and the timing or likelihood of regulatory filings and approvals, expectations regarding market acceptance and size, plans for launch and commercialization, plans and objectives of management for future operations, and future results of anticipated product candidates, are forward-looking statements. New risks and uncertainties may emerge from time to time, and it is not possible to predict all risks and uncertainties. No representations or warranties (expressed or implied) are made about the accuracy of any such forward-looking statements. These statements are also subject to a number of material risks and uncertainties that are discussed in the section entitled "Risk Factors" in resTORbio’s annual report on Form 10-K for the fiscal year ended December 31, 2017, as well as discussions of potential risks, uncertainties, and other important factors in resTORbio's subsequent filings with the Securities and Exchange Commission. Any forward-looking statement speaks only as of the date on which it was made. Neither we, nor our affiliates, advisors, or representatives, undertake any obligation to publicly update or revise any forward-looking statement, whether as a result of new information, future events or otherwise, except as required by law. Certain information contained in this presentation relates to or is based on studies, publications, surveys and other data obtained from third-party sources and the Company’s own internal estimates and research. While we believe these third-party sources to be reliable as of the date of this presentation, we have not independently verified, and we make no representation as to the adequacy, fairness, accuracy or completeness of, any information obtained from third-party sources. In addition, all of the market data included in this presentation involves a number of assumptions and limitations, and there can be no guarantee as to the accuracy or reliability of such assumptions. Finally, while we believe our own internal research is reliable, such research has not been verified by any independent source. Forward-looking statements


Slide 3

Aging is the biggest risk factor for most chronic diseases Why does this happen? Aging is not just due to random wear and tear Aging is biology that may be targeted with medicines Global Burden of Disease Collaborative Network. Global Burden of Disease Study 2017 (GBD 2017) Results


Slide 4

resTORbio is targeting the biology of aging AGING NAD Depletion Mitochondrial Dysfunction TORC1 Activity DNA Damage Accumulation of Senescence Cells Stem Cell Exhaustion Epigenetic Changes


Slide 5

The TORC1 pathway Source: Lamming, Dudley W., et al. (2013) Journal of Clinical Investigation123 (3): 980–989. TORC1 inhibition extended lifespan and healthspan in multiple species Mice Flies Worms Yeast TORC1 is an evolutionarily conserved pathway that regulates aging


Slide 6

Species Genetic Manipulation to Inhibit mTOR Yeast SCH9 (Akt/S6K homolog) insertional mutant 1 SCH9 (Akt/S6K homolog) deletion 1 SCH9 (Akt/S6K homolog) insertional mutant 2 SCH9 (Akt/S6K homolog) deletion 2 TOR1 deletion 3 TOR1 deletion 4 C. elegans TOR (let-363) RNAi 5 Raptor (daf-15) heterozygous 6 S6K (rsks-1) RNAi 7 S6K (rsks-1) deletion mutant 7 TOR (let-363) RNAi 7 S6K (rsks-1) RNAi 8 S6K (rsks-1) deletion mutant 8 TOR (let-363) RNAi 8 Raptor (daf-15) RNAi 9 RagGTPase (raga-1) RNAi 9 RagGTPase (raga-1) RNAi 9 Rheb (rheb-1) RNAi 9 D. melanogaster dTSC1 overexpression 10 dTSC2 overexpression 10 dTOR FRB domain (dominant negative) 10 dS6K dominant negative 10 DTOR mutant (hypomorph) 11 d4E-BP overexpression 12 d4E-BP weak activated 12 d4E-BP strong activated 12 M. musculus Loss of S6K1 13 Mtor+/-MIst8+/- genotype 14 Extensive genetic validation that TORC1 inhibition extends lifespan across species Corresponding citations can be found on slide 48


Slide 7

TORC1 inhibition may improve the function of multiple aging organ systems Improvement in physical activity Selman et al., Science, 2011 Harrison et al., Nature, 2009 Wilkinson et al., Aging Cell, 2014 Flynn et al., Aging Cell, 2013 Reversal of aging-related immune dysregulation Chen et al., Science Sig, 2009 Selman et al., Science, 2011 Neff et al., JCI, 2013 Hurez et al., Aging Cell, 2015 Reversal of aging-related cardiac dysfunction Flynn et al., Aging Cell, 2013 Dai et al., Aging Cell, 2014 Chiao et al., Aging, 2016 Improved Neurologic Function Tain et al., Nature Neuroscience, 2009 Malagelada et al., J Neurosci, 2010 Spilman et al., PLoS ONE, 2010 Halloran et al., Neuroscience, 2012 Majumder et al., Aging Cell, 2012 Neff et al., JCI, 2013


Slide 8

Additional Aging-Related Target Most advanced pipeline targeting aging-related diseases RTB101 Respiratory Tract Infections Parkinson’s Disease Program Indication Phase 1 Phase 2 Phase 3 Discovery Heart Failure with Preserved Ejection Fraction Preclinical Urinary Tract Infections *For heart failure with preserved ejection fraction, Parkinson’s Disease and certain other infections, we may be required to file an investigational new drug application, or IND, prior to initiating Phase 2 clinical trials. We expect to have the ability to initiate these Phase 2 clinical trials without the need to conduct prior Phase 1 trials. End-of-Phase 2 meeting planned for 1Q19 Initiate Phase 1b/2a in 1Q19* RTB101+ sirolimus Additional TORC1 Inhibitor Undisclosed RTB101 RTB101 RTB101 + rapalog Undisclosed Current Indications Potential Indications Discovery


Slide 9

TORC1: A Compelling Target for Neurodegenerative Diseases Matt Kaeberlein, PhD Department of Pathology University of Washington 9


Slide 10

Age is the greatest risk factor for neurodegenerative disease Age Risk of Death (per 100,000) http://www.spring.org.uk/ 10


Slide 11

The biology of aging drives neurodegenerative disease AGING Vascular dementia ALS Lewy body disease Alzheimer’s Parkinson’s Frontotemporal dementia 11


Slide 12

We now better understand the biology of aging 12


Slide 13

TORC1 connects aging with neurodegenerative disease TORC1 = mTOR Complex 1 (Adapted from Oncogene 36:2191 2017) mTOR Inhibitor 13


Slide 14

TORC1 is critical when we are young ↓ Autophagy TORC1 ↑ Protein synthesis ↑ Lipid Synthesis Environmental and Hormonal Growth signals ↑ Inflammation 14


Slide 15

TORC1 becomes hyperactivated as we get old ↓ Autophagy ↑ Protein synthesis ↑ Lipid Synthesis Environmental and Hormonal Growth signals ↑ Inflammation Proteo-toxicity Immune-senescence Metabolic dysfunction TORC1 TORC1 Inhibitor TORC1 inhibition may delay aging X X 15


Slide 16

TORC1 inhibition improves the function of aging organ systems in multiple mammalian species Inhibition of TORC1 increases lifespan and improves immunologic, neurologic and cardiac function in aging mice Inhibition of TORC1 reverses age-related decline in cardiac function in pet dogs Inhibition of TORC1 improves immune function in elderly people (resTORbio) 16


Slide 17

TORC1 inhibition has therapeutic benefit in multiple neurodegenerative diseases TORC1 inhibition delays or reverses Alzheimer’s disease in multiple mouse models TORC1 inhibition effective in multiple mouse and fly models of Parkinson’s disease Enhanced autophagy leading to clearance of aggregated such as amyloid-b and a-synuclein likely the primary mechanism ↓ Lipid Synthesis ↑ Autophagy TORC1 ↓ Protein synthesis ↓ Inflammation Neuroprotection 17


Slide 18

TORC1 is a Master Negative Regulator of Autophagy TORC1 TORC1 TFEB P ULK1 TFEB P Nucleus TFEB Autophagy and Lysosomal Genes ULK1 Vps34 complex Immature/Inactive Phagophore Atg complexes LC3-II Phagophore a-syn aggregate Autophagosome (Figure adapted from Neuron 93:1015, 2017) Lysosome Autolysosome 18


Slide 19

TORC1 inhibition may be of particular benefit in Parkinson’s Disease Nearly 200 papers published with “TORC1” (or “mTOR”) and “Parkinson’s” in the title/abstract Activation of autophagy is the favored mechanism of action Protects against a-synuclein toxicity Prevents neuron loss Improves motor function (J Neurosci 30:1166, 2010 ) Number of Dead/Dying Neurons/field Vehicle mTOR Inhibitor MPTP MPTP + mTOR Inhibitor 19


Slide 20

Classes of TORC1 Inhibitors Rapalogs: Allosteric inhibitors of TORC1 Chronic inhibition can also suppress TORC2 Inhibit only some targets downstream of TORC1 Approved for use in oncology indications and to prevent organ transplant rejection The class of TORC inhibitors used in most PD models Sirolimus (rapamycin) Catalytic inhibitors ATP competitive catalytic site mTOR inhibitors Inhibit all targets downstream of TORC1 May have advantages over rapalogs for PD RTB101 20


Slide 21

Rapamycin is an imperfect TORC1 inhibitor ↑ Autophagy TORC1 ↓ Protein synthesis ↓ Inflammation Neuroprotection Rapamycin RTB101 + Rapamycin does not consistently induce autophagy Chronic rapamycin treatment also inhibits TORC2 leading to side effects RTB101 induces autophagy at high concentrations that are difficult to achieve in the CNS Co-administration of rapamycin reduces the concentration of RTB101 needed to induce autophagy 21


Slide 22

Targeting Autophagy in Parkinson’s Disease Roy Alcalay, MD, MS Florence Irving Assistant Professor of Neurology Division of Movement Disorders Columbia University Medical Center


Slide 23

Disclosures Funding: Dr. Alcalay is funded by the Parkinson’s Foundation, the National Institutes of Health (NS080915, NS094607), the Smart Foundation and the Michael J. Fox Foundation. Dr. Alcalay receives consultation fees from Genzyme/Sanofi, Denali, Biogen and Roche.


Slide 24

What is Parkinson’s Disease? Clinical manifestations: Four cardinal motor symptoms: Resting tremor Bradykinesia (slowed movements) Muscle rigidity Postural instability Pathobiology: Loss of >50% of the neurons that produce the neurotransmitter dopamine in a specific area of the brain (substantia nigra) Protein aggregation (Lewy bodies) Prevalence: Parkinson’s disease (PD) is the second most common neurodegenerative disease. Affects >1m Americans


Slide 25

PD: Non Motor Symptoms Many patients develop non-motor symptoms including: Autonomic dysfunction Sleep problems (RBD) Psychiatric symptoms: anxiety, depression Impaired sense of smell Over 80% of affected individuals develop cognitive impairment over time. There are a variety of symptomatic treatments. There are no FDA approved disease-modifying treatments.


Slide 26

PD Biology PD is defined by neuronal degeneration with protein (alpha-synuclein, a-syn) aggregation Multiple genes have been linked to PD Many of them are in the auto-phagosome pathway


Slide 27

Impaired Autophagy Genes in Neurodegenerative Diseases (Neuron 93:1015, 2017)


Slide 28

GBA/PD: A Genetically-linked form of PD A single GBA mutation (heterozygous mutation) is the most common genetic risk for PD 5-10% of PD patients carry a GBA mutation or variant (Gan-Or, 2015) Two mutations (homozygous mutations) in the GBA gene cause Gaucher disease, a lysosomal storage disorder Gaucher is caused because of significantly diminished glucocerebrosidase (GCase) activity


Slide 29

GBA: An Accelerated form of Idiopathic PD Clinical Similarities Motor signs are similar to idiopathic PD (iPD), but progress faster than idiopathic PD Cognitive impairment, loss of sense of smell, sleep disturbances and autonomic dysfunction are more common than in patients with idiopathic PD Good symptomatic motor response to symptomatic medication (L-dopa) like idiopathic PD No FDA approved interventions to slow disease progression


Slide 30

GBA: An Accelerated form of iPD Histologic and Biochemical Similarities GBA/PD associated with higher amounts of a-syn deposition (Lewy bodies) in brain (an accelerated form of PD) GCase (the GBA protein product) expression and activity decreases in human brains with age (Ann Clin Transl Neurol 2:433, 2015) Lewy bodies in GBA a-syn in Lewy bodies


Slide 31

TORC1 Inhibition Increases Autophagosomes and Decreases a-syn (PLoS one 5:e9313, 2010) LCE autophagosome marker staining (brown) Control Vehicle a-syn Transgenic Vehicle Mouse: Treatment: a-syn Transgenic mTOR inhibitor a-synuclein staining (brown)


Slide 32

TORC1 inhibitor/PD Clinical Trials: Why GBA/PD may be a good target for proof of concept/efficacy trials All disease modification strategies for PD to date have failed. Glucosylceramide (GCase substrate) is reduced by mTOR inhibition1 GBA/PD is more homogenous. Strongly associated with a-syn pathology. GBA/PD is faster progressing (earlier outcomes) 1Cancer Cell 32, 807 e812, 2017


Slide 33

Targeting the biology of aging to prevent and treat aging-related diseases


Slide 34

The TORC1 pathway Source: Lamming, Dudley W., et al. (2013) Journal of Clinical Investigation123 (3): 980–989. TORC1 inhibition extended lifespan and healthspan in multiple species Mice Flies Worms Yeast TORC1 is an evolutionarily conserved pathway that regulates aging


Slide 35

TORC1 inhibition may improve the function of multiple aging organ systems Improvement in physical activity Selman et al., Science, 2011 Harrison et al., Nature, 2009 Wilkinson et al., Aging Cell, 2014 Flynn et al., Aging Cell, 2013 Reversal of aging-related immune dysregulation Chen et al., Science Sig, 2009 Selman et al., Science, 2011 Neff et al., JCI, 2013 Hurez et al., Aging Cell, 2015 Reversal of aging-related cardiac dysfunction Flynn et al., Aging Cell, 2013 Dai et al., Aging Cell, 2014 Chiao et al., Aging, 2016 Improved Neurologic Function Tain et al., Nature Neuroscience, 2009 Malagelada et al., J Neurosci, 2010 Spilman et al., PLoS ONE, 2010 Halloran et al., Neuroscience, 2012 Majumder et al., Aging Cell, 2012 Neff et al., JCI, 2013 Improved Neurologic Function Tain et al., Nature Neuroscience, 2009 Malagelada et al., J Neurosci, 2010 Spilman et al., PLoS ONE, 2010 Halloran et al., Neuroscience, 2012 Majumder et al., Aging Cell, 2012 Neff et al., JCI, 2013


Slide 36

TORC1 inhibitors extend lifespan in mice even when started late in life and given intermittently Daily Dosing Intermittent Dosing Once Every 5 Days (1) (2) Harrison et al. (2009) Nature, 460:392-396 Arriola Apelo et al. (2016) Gerontol A Biol Sci Med Sci, 71: 876–88


Slide 37

mTOR Selective inhibition of TORC1 may have therapeutic benefit for the treatment of aging-related diseases Inhibition of TORC2 by genetic mutation decreases lifespan and causes hyperglycemia and hyperlipidemia in mice (Science, 2012; Aging Cell, 2014) S6K Ulk1 4EBP1 Atg Inhibition of TORC1 by genetic mutation extends lifespan (Science, 2012) Knock out of S6K extends lifespan and healthspan (Science, 2009) Overexpression extends lifespan (Cell, 2009) Transgenic overexpression extends lifespan (Nat Comm, 2013) TORC2 TORC1 4EBP1


Slide 38

TORC1 S6K 4EBP1 ULK1 TFEB Lipin1 Rapalogs lower the concentration of RTB101 needed to inhibit TORC1 in the brain and induce autophagy in animal models Rapalog sirolimus/ everolimus S6K High dose RTB101 4EBP1 ULK1 TFEB Lipin1 S6K TORC1 Lower dose RTB101 + sirolimus 4EBP1 ULK1 TFEB Lipin1 S6K TORC1 Increased autophagy Increased lysosomal biogenesis Decreased lipid synthesis Decreased protein synthesis


Slide 39

Potential mechanism underlying synergistic inhibition and autophagy activation by sirolimus + RTB101 Sirolimus may induce a conformation change in TORC1 that allows lower concentrations of RTB101 to inhibit TORC S6K 4EBP1 Sirolimus FKBP12 TORC RTB101


Slide 40

RTB101 and sirolimus synergize to induce autophagy at low concentrations RTB101 (nM) 87.5 130.58 114.37 156.80 170.61 173.28 181.56 196.15 174.61 158.67 216.07 43.8 91.48 71.47 123.06 118.25 166.88 154.73 189.63 194.12 190.70 214.89 21.9 31.89 25.16 81.50 100.98 125.12 137.82 212.58 197.37 166.87 218.33 10.9 0.02 4.25 29.25 41.45 88.97 138.95 155.32 184.65 146.93 179.15 5.47 -12.14 -14.12 -1.11 8.36 44.22 81.09 103.23 143.72 120.56 123.57 2.73 -12.10 -6.71 -0.19 -1.19 25.53 43.99 75.14 96.76 73.48 100.10 1.37 -7.40 -17.37 0.03 0.09 13.03 29.69 41.98 54.65 60.23 68.35 0.684 -23.25 -25.36 3.41 -2.42 5.87 16.31 26.84 52.55 33.51 31.80 0.342 -16.81 -28.70 -7.67 -5.83 5.18 14.95 9.42 33.10 21.35 43.68 0 -11.63 -20.72 -6.80 -6.46 -1.54 9.74 2.82 13.34 10.25 -4.02 0.00000 0.00214 0.00854 0.0342 0.137 0.547 2.19 8.75 35.0 140 Sirolimus (nM) % Autophagy induction >50%   <50%   Results shown are representative of 3 independent experiments Higher scores indicate greater autophagy


Slide 41

RTB101 synergizes with everolimus to clear mHtt protein aggregates and rescue striatal neurons 250nM Everolimus Green = Neurofilaments Red = Neuron cell bodies (DARPP-32) 50nM RTB101 300nM RTB101 10nM RTB101 + 250 nM Everolimus 30nM RTB101 + 250 nM Everolimus 50nM RTB101 + 250 nM Everolimus Days in Culture: Everolimus (nM): RTB101 (nM): 7 0 0 14 0 0 21 0 0 250 0 0 50 250 50 0 300 250 200 150 100 50 0 #mHtt Aggregates/area


Slide 42

Intermittent dosing of TORC1 inhibitors may have better safety and efficacy than daily dosing Beneficial effects of rapamycin on lifespan can be achieved with dosing once every 5-7 days with reduced side effects1 Rapamycin administered 3x/week (intermittent mTOR inhibition) is required for autophagic lysosomal reformation 1Arriola Apelo et al. (2016) Gerontol A Biol Sci Med Sci, 71: 876–88; 2Nature 465:942, 2010. mTOR Inhibition mTOR TORC1 inhibitor Nutrients Separation Maturation Lysosome Nature 465:942, 2010


Slide 43

Ameliorating Neurodegenerative Diseases Parkinson’s Disease


Slide 44

Phase 1b/2a Parkinson’s disease trial design Study initiation planned for 1Q19 Cohort RTB 101 dose (mg) Sirolimus dose (mg) 1 300 0 2 0 2 3 300 2 4 300 4 5 300 6 Design Randomized, Placebo-Controlled Phase 1b/2a Study (4-week dosing) Mild PD patients (mH&Y I-II) with or without GBA mutations On standard of care PD drugs Once weekly dosing Study Size N=45 (2:1 randomization) Key Endpoints Primary endpoint: Safety and tolerability Secondary endpoint: Exposure in blood, plasma and CSF Exploratory endpoints: Biomarkers in plasma and CSF Clinical assessments, wearables or matching placebo


Slide 45

Summary TORC1 may be an important therapeutic target for several neurodegenerative diseases in which misfolded proteins aggregate and cause neuronal toxicity TORC1 inhibition has shown therapeutic benefit in multiple preclinical PD models TORC1 inhibition may be of benefit in PD by inducing autophagy and thereby clearing toxic proteins in neurons Combinations of TORC1 inhibitors (RTB101 and sirolimus) administered intermittently may provide the best approach to activating brain autophagy Planning to initiate a Ph1b/2a study in PD with RTB101 + sirolimus in 1Q19 RTB101+ sirolimus may be of particular benefit to patients with GBA-PD


Slide 46

Targeting the biology of aging with TORC1 Inhibitors TORC1 inhibition extended lifespan and improved: Immune Function Neurologic Function ü Clinical trials enrolling > 900 subjects Initiation of Phase 1a/2b planned in 1Q19 Translation to humans with RTB101 Mice Flies Worms Yeast


Slide 47

February 7, 2019 Targeting the biology of aging to prevent and treat aging-related diseases


Slide 48

Extensive genetic validation that TORC1 Inhibition extends lifespan across species 1 Fabrizio P, Pozza F, Pletcher SD, Gendron CM, Longo VD. Regulation of longevity and stress resistance by Sch9 in yeast. Science. 2001;292(5515):288–290. Fabrizio P, Pletcher SD, Minois N, Vaupel JW, Longo VD. Chronological aging-independent replicative life span regulation by Msn2/Msn4 and Sod2 in Saccharomyces cerevisiae. FEBS Lett. 2004; 557(1–3):136–142. Kaeberlein M, et al. Regulation of yeast replicative life span by TOR and Sch9 in response to nutrients. Science. 2005;310(5751):1193–1196. Bonawitz ND, Chatenay-Lapointe M, Pan Y, Shadel GS. Reduced TOR signaling extends chronological life span via increased respiration and upregulation of mitochondrial gene expression. Cell Metab. 2007; 5(4):265–277. Vellai T, Takacs-Vellai K, Zhang Y, Kovacs AL, Orosz L, Muller F. Genetics: influence of TOR kinase on lifespan in C. elegans. Nature. 2003;426(6967):620. Jia K, Chen D, Riddle DL. The TOR pathway inter- acts with the insulin signaling pathway to regulate C. elegans larval development, metabolism and life span. Development. 2004;131(16):3897–3906. Hansen M, Taubert S, Crawford D, Libina N, Lee SJ, Kenyon C. Lifespan extension by conditions that inhibit translation in Caenorhabditis elegans. Aging Cell. 2007;6(1):95–110. Pan KZ, et al. Inhibition of mRNA translation extends lifespan in Caenorhabditis elegans. Aging Cell. 2007;6(1):111–119. Robida-Stubbs S, et al. TOR Signaling and rapamy- cin influence longevity by regulating SKN-1/Nrf and DAF-16/FoxO. Cell Metab. 2012;15(5):713–724. Kapahi P, Zid BM, Harper T, Koslover D, Sapin V, Benzer S. Regulation of lifespan in Drosophila by modulation of genes in the TOR signaling pathway. Curr Biol. 2004;14(10):885–890. Luong N, et al. Activated FOXO-mediated insulin resistance is blocked by reduction of TOR activity. Cell Metab. 2006;4(2):133–142. Zid BM, et al. 4E-BP extends lifespan upon dietary restriction by enhancing mitochondrial activity in Drosophila. Cell. 2009;139(1):149–160. Selman C, et al. Ribosomal protein S6 kinase 1 signaling regulates mammalian life span. Science. 2009;326(5949):140–144. Lamming DW, et al. Rapamycin-induced insulin resistance is mediated by mTORC2 loss and uncoupled from longevity. Science. 2012; 335(6076):1638–1643.