0001701541 false 0001701541 2022-01-10 2022-01-10 iso4217:USD xbrli:shares iso4217:USD xbrli:shares

 

 

 

UNITED STATES
SECURITIES AND EXCHANGE COMMISSION

Washington, D.C. 20549

 

 

 

FORM 8-K

 

 

 

CURRENT REPORT
Pursuant to Section 13 or Section 15(d)
of the Securities Exchange Act of 1934

 

Date of Report (Date of earliest event reported): January 10, 2022

 

 

 

Black Diamond Therapeutics, Inc.

(Exact name of registrant as specified in its charter)

 

 

 

Delaware 001-39200 81-4254660
(State or other jurisdiction (Commission (IRS Employer
of incorporation) File Number) Identification No.)

 

One Main Street, 10th Floor    
Cambridge, MA   02142
(Address of principal executive offices)   (Zip Code)

 

Registrant’s telephone number, including area code: 617-252-0848

 

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 to 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, $0.0001 par value per share BDTX 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 (§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 x

 

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 7.01Regulation FD Disclosure.

 

On January 10, 2022, Black Diamond Therapeutics, Inc., (the “Company”) issued a press release titled, “Black Diamond Therapeutics Announces Strategic Priorities and Expected Milestones for 2022” and updated its corporate presentation for use in meetings with investors, analysts and others. A copy of the press release and a copy of the corporate presentation are furnished as Exhibits 99.1 and 99.2, respectively, to this Current Report on the Form 8-K.

 

The information furnished under this Item 7.01, including Exhibits 99.1 and 99.2, 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 or the Exchange Act, except as expressly set forth by specific reference in such a filing.

 

Item 8.01.Other Events.

 

BDTX-1535

 

On January 10, 2022, the Company announced the submission of its Investigational New Drug application to the US Food and Drug Administration ("FDA") for BDTX-1535 and expects to initiate the Phase 1 study of BDTX-1535 in the first quarter of 2022, subject to allowance of the IND by the FDA. The Company expects to provide a clinical data update on BDTX-1535 in the second half of 2023.

 

BDTX-189

 

The Company also announced that, due to the rapid evolution of the treatment landscape in non-small cell lung cancer harboring either EGFR or HER2 Exon 20 insertion mutations, the Company has decided to enroll additional patients into the MasterKey-01 Phase 1 safety expansion cohort to obtain more clinical data and inform future development of BDTX-189. The Company expects to provide further guidance on the BDTX-189 program in 2022.

 

Financial Guidance

 

The Company announced that it ended 2021 with approximately $210 million in cash, cash equivalents and investments, which the Company believes is sufficient to fund its anticipated operating expenses and capital expenditure requirements into 2024.

 

 

 

 

Item 9.01. Financial Statements and Exhibits.

 

(d) Exhibits:

 

Exhibit
No.
  Description
99.1   Press Release issued by Black Diamond Therapeutics, Inc., dated January 10, 2022.
99.2   Corporate Presentation of Black Diamond Therapeutics, Inc., dated January 10, 2022.
104   Cover Page Interactive Data File (embedded within the Inline XBRL document contained in Exhibit 104).

 

 

 

 

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.

 

January 10, 2022 BLACK DIAMOND THERAPEUTICS, INC.
   
  By: /s/ Brent Hatzis-Schoch
    Name: Brent Hatzis-Schoch
    Title: Chief Operating Officer and General Counsel

 

 

 

Exhibit 99.1 

 

 

 

Black Diamond Therapeutics Announces Strategic Priorities and Expected Milestones for 2022

 

-      Company announces IND submission for BDTX-1535 for the treatment of GBM and NSCLC
including those with CNS tumors;

 

-      Company to enroll additional patients into MasterKey-01 Phase 1 safety expansion cohort to
obtain more clinical data and inform future development of BDTX-189;

 

-      Company anticipates initiation of IND-enabling studies for its CNS-penetrant Class I,II,III BRAF
program in 2022;

 

-      Existing resources to be prioritized in the near-term for advancement of MasterKey pipeline
programs BDTX-1535, BRAF, FGFR and the MAP discovery engine; cash runway extended to 2024

  

CAMBRIDGE, Mass. And NEW YORK, January 10, 2022 (GLOBE NEWSWIRE) – Black Diamond Therapeutics, Inc. (Nasdaq: BDTX), a precision oncology medicine company pioneering the discovery and development of MasterKey therapies, today announced a strategic pipeline update and outlined its expected upcoming milestones.

 

“MasterKey inhibitors target mutation families and are designed to address the unmet medical need of cancer patients with genetically defined cancers for whom precision therapies are not available,” said David Epstein, Ph.D., Chief Executive Officer of Black Diamond Therapeutics. “We are incredibly pleased to have submitted the IND ahead of schedule for BDTX-1535, a next generation CNS penetrant MasterKey inhibitor designed to target EGFR driver mutations found in certain patients with GBM and NSCLC. The oncogenic alterations of EGFR, particularly those associated with GBM, result in distinct conformations which impart unique pharmacology and drug resistance. BDTX-1535 is designed to exploit this mechanism as a critical point of attack. We believe Black Diamond is uniquely positioned to deliver a pipeline of truly differentiated MasterKey programs as we leverage our expertise in cancer genomics, onco-protein function and drug discovery.”

 

“As a result of the rapid evolution of the treatment landscape in NSCLC harboring EGFR or HER2 Exon 20 insertion mutations, we have decided to obtain further clinical data from the BDTX-189 safety expansion cohort at the recommended Phase 2 dose in 2022 in order to inform the future development of our BDTX-189 program, gating the start of a Phase 2 trial. The revised strategy enables near-term prioritization of BDTX-1535 clinical development and further investment in our pipeline, while allowing us to obtain more clinical data on BDTX-189, and simultaneously extends our cash runway into 2024.”

 

 

 

 

Pipeline Updates and Expected Milestones:

BDTX-1535 

·Black Diamond announced ahead of schedule the submission of its Investigational New Drug (IND) application to the US Food and Drug Administration (FDA) for BDTX-1535 and expects to initiate the Phase 1 study of BDTX-1535 in the first quarter of 2022, subject to allowance of the IND by the FDA.
·BDTX-1535 is designed as a potent, selective, brain-penetrant and irreversible MasterKey inhibitor of epidermal growth factor receptor (EGFR) mutations expressed in glioblastoma multiforme (GBM) and of intrinsic and acquired resistance EGFR mutations to third generation EGFR inhibitors in NSCLC.
·In pre-clinical studies, Black Diamond has demonstrated that oncogenic alterations of EGFR, particularly those associated with GBM, result in distinct conformations which impart unique pharmacology and drug resistance. BDTX-1535 is designed to exploit this mechanism and has demonstrated anti-cancer activity and growth regressions across a panel of patient-derived xenograft models including intracranial tumor models.
·It is estimated that approximately 50% of GBM patients harbor an oncogenic EGFR alteration that has the potential to be addressed by BDTX-1535, representing a potential patient population of greater than 60,000 patients annually across the US, EU, Japan and China.
·It is estimated that across the US, EU, Japan and China there are approximately 20,000 patients who are diagnosed annually with non-small cell lung cancer (NSCLC) harboring an EGFR intrinsic or acquired resistance mutation.
·The Company expects to provide a clinical data update on BDTX-1535 in the second half of 2023.

 

BDTX-189

·BDTX-189 is designed as a MasterKey inhibitor targeting families of oncogenic mutations in EGFR and HER2.
·Clinical data obtained from the MasterKey-01 study to date have demonstrated a favorable safety profile for BDTX-189 and early signs of clinical activity in patients whose tumors are driven by MasterKey mutation families, including two confirmed partial responses in heavily pretreated patients who have remained on treatment for more than 10 months.
·Due to the rapid evolution of the treatment landscape in NSCLC harboring either EGFR or HER2 Exon 20 insertion mutations, the Company has decided to gate the initiation of the Phase 2 portion of the MasterKey-01 study and determine next steps in development based on further clinical data obtained from the safety expansion cohort at the recommended Phase 2 dose.
·The Company expects to provide further guidance on the BDTX-189 program in 2022.

 

BRAF Program

·Black Diamond is developing a CNS-penetrant BRAF inhibitor against a family of Class I, II, III canonical and non-canonical mutations. The Company’s lead BRAF compound is designed to be highly selective, potent and to avoid paradoxical activation.
·In cell-based assays, Black Diamond’s lead BRAF compound demonstrated potent inhibition of a wide spectrum of BRAF mutations and fusions and exhibited dose-dependent inhibition of protein kinase RNA-like endoplasmic reticulum kinase downstream signaling.

 

 

 

 

·In preclinical BRAF-driven tumor models, daily dosing of the lead compound demonstrated dose-dependent tumor growth inhibition, tumor regression and survival consistent with potent on-target and on-pathway inhibition.
·The Company’s lead BRAF compound demonstrated robust brain penetration properties and achieved intracranial tumor growth inhibition in pre-clinical studies.
·It is estimated that across the US, EU, and Japan there are approximately 190,000 patients with solid tumors who are diagnosed annually with BRAF oncogenic mutations.
·Black Diamond anticipates the initiation of IND-enabling studies for the BRAF program in 2022.

 

MAP Discovery Engine

·The Company continues to focus on building its Mutation-Allostery-Pharmacology (MAP) Discovery Engine to exploit mutant onco-protein conformations for the delivery of selective MasterKey therapies, with a focus on developing a MasterKey inhibitor that spares wild type fibroblast growth factor receptor 1 ( FGFR1) while inhibiting FGFR2 and FGFR3 mutant families, as well as other preclinical stage programs.
·The Company plans to integrate new molecular dynamic modeling tools from its strategic partnership with OpenEye Scientific.

 

Financial Guidance

·Black Diamond ended 2021 with approximately $210 million in cash, cash equivalents and investments, which the Company believes is sufficient to fund its anticipated operating expenses and capital expenditure requirements into 2024.

 

About Black Diamond

Black Diamond Therapeutics is a precision oncology medicine company pioneering the development of novel MasterKey therapies. Black Diamond is addressing the significant unmet need for novel precision oncology therapies for patients with genetically defined cancers who have limited treatment options. Black Diamond is built upon a deep understanding of cancer genetics, onco-protein function, and drug discovery. The Company’s proprietary Mutation-Allostery-Pharmacology, or MAP drug discovery engine, is designed to allow Black Diamond to analyze population-level genetic sequencing tumor data to predict and validate oncogenic mutations that promote cancer across tumor types as MasterKey mutations. Black Diamond discovers and develops selective MasterKey therapies against these families of oncogenic mutations. Black Diamond was founded by David M. Epstein, Ph.D., and Elizabeth Buck, Ph.D. For more information, please visit www.blackdiamondtherapeutics.com.

 

 

 

 

Forward-Looking Statements

Statements contained in this press release regarding matters that are not historical facts are “forward-looking statements” within the meaning of the Private Securities Litigation Reform Act of 1995. Because such statements are subject to risks and uncertainties, actual results may differ materially from those expressed or implied by such forward-looking statements. Such statements include, but are not limited to, statements regarding: the continued development and advancement of BDTX-1535, including expectations for IND allowance and plans for initiating the Phase 1 trial of BDTX-1535, the continuation of the BDTX-189 safety expansion cohort and the resulting data, the continued development of the BRAF program, including the timing for initiating IND-enabling studies, the continued development of the FGFR program, including plans for nominating a development candidate, the continued development of the MAP discovery engine and the Company’s cash runway. Any forward-looking statements in this statement 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. Risks that contribute to the uncertain nature of the forward-looking statements include: the success, cost, and timing of the Company’s product candidate development activities and planned IND-enabling studies and clinical trials, the Company’s ability to execute on its strategy, regulatory developments in the United States, the Company’s ability to fund operations, and the impact that the current COVID-19 pandemic will have on the Company’s clinical trials and preclinical studies, supply chain, and operations, as well as those risks and uncertainties set forth in its Annual Report on Form 10-K for the year ended December 31, 2020, filed with the United States Securities and Exchange Commission and in its other filings filed with the United States Securities and Exchange Commission. All forward-looking statements contained in this press release speak only as of the date on which they were made. The Company undertakes no obligation to update such statements to reflect events that occur or circumstances that exist after the date on which they were made.

 

Contacts

For Investors:

Julie Seidel

investors@bdtx.com

 

For Media:

Kathy Vincent

(310) 403-8951

media@bdtx.com

 

 

 

Exhibit 99.2

 

GRAPHIC

1 Black Diamond Therapeutics, Inc. Pioneering the Development of MasterKey Therapies JANUARY 2022

GRAPHIC

2 Important Notice and Disclaimers This presentation contains “forward-looking statements” of Black Diamond Therapeutics, Inc.(“Black Diamond,” “we” or “our”) within the meaning of the Private Securities Litigation Reform Act of 1995. These forward-looking statements include, but are not limited to, express or implied statements regarding our ability to advance and expand the MAP drug discovery engine, the potential timing and advancement of our clinical trial and preclinical studies, including the initiation of our clinical trial for BDTX-1535 and the continuation of our clinical trial for BDTX-189, the timing and potential achievement of milestones to advance our product candidate pipeline, including initiation of additional investigational new drug (“IND”)-enabling studies, filing and allowance of INDs and product candidate nomination, and the potential commercial opportunities, including value and market, for our product candidates. Any forward-looking statements in this presentation are based on management’s current expectations and beliefs 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. Our actual future results may be materially different from what we expect due to factors largely outside our control, including the results of clinical trials, clinical trial patient enrollment, changes in regulatory requirements or decisions of regulatory authorities, commercialization plans and timelines if approved, the actions of our third party clinical research organizations, suppliers and manufacturers, and the impact that the current COVID-19 pandemic will have on our clinical trials, pre-clinical studies, and operations. Except as required by law, we assume no obligation to update these forward-looking statements publicly, or to update the reasons actual results could differ materially from those anticipated in the forward-looking statements, even if new information becomes available in the future. For a discussion of these and other risks and uncertainties, and other important factors, any of which could cause our actual results to differ from those contained in the forward-looking statements, see the section entitled “Risk Factors” in our 2020 annual report on Form 10-K, as well as discussions of potential risks, uncertainties, and other important factors in our other filings with the Securities and Exchange Commission. All information in this presentation is as of the date hereof, and we undertake no duty to update this information unless 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 our own internal estimates and research. While we believe these third-party sources to be reliable as of the date of this presentation, it has not independently verified, and makes 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.

GRAPHIC

Black Diamond Therapeutics Overview

GRAPHIC

4 Expanding Precision Medicine Through the Development of MasterKey Therapies Our proprietary MAP drug discovery engine is designed to: • Predict and validate novel oncogenic mutant families from population level tumor genomics • Pioneer mutant family conformation-based MasterKey drug design • Provide opportunities beyond oncology and small molecules MasterKey therapies designed to address overlooked mutation families; providing precision oncology to greater numbers of patients with genetically defined tumors Proof-of-principle mutation family selective inhibitors; clinical and late-preclinical pipeline of MasterKey inhibitors derived from our MAP drug discovery engine: EGFR, HER2, BRAF, and FGFR BDTX-1535, an irreversible, mutant selective, brain-penetrant EGFR inhibitor entering Phase 1 for the treatment of GBM and NSCLC driven by EGFR intrinsic & acquired resistance mutations BDTX-189, a mutant selective EGFR/HER2 inhibitor in Phase 1 Safety Expansion study in patients expressing targeted oncogenic mutations

GRAPHIC

5 Black Diamond’s MasterKey Approach Designed to Address Overlooked Mutation Families Classic/Current Approach: Targeting active site kinase domain mutations With expanding genetic profiling of cancer patients via Next Generation Sequencing (NGS) Mutation families yield significant market opportunities for populations lacking suitable precision therapies Black Diamond Approach: Targeting mutation families to expand the opportunity for precision oncology Less than 15% patients1 with metastatic cancer eligible for approved precision oncology medicines Targeting single mutations in individual tumor types 5 1 Haslam, A., et al. Annals Oncology Vol 32, Issue 7, p926-932; July 2021

GRAPHIC

6 Wholly-Owned Novel Classes of Precision Medicines Target Drug Candidate Indication Discovery Optimization IND-Enabling Phase 1 Phase 2 Phase 3 EGFR BDTX-1535 GBM, NSCLC ± CNS mets EGFR HER2 BDTX-189 NSCLC, breast BRAF Undisclosed Targeted solid tumors ± CNS mets FGFR Undisclosed Targeted solid tumors Safety Expansion Update (2022) Clinical Data (2023) Initiate IND-Enabling Studies (2022) Development Candidate Selection (2022) EGFR=Epidermal Growth Factor Receptor; HER2= Human Epidermal Growth Factor Receptor 2; FGFR=Fibroblast Growth Factor Receptor; IND=Investigational New Drug

GRAPHIC

MAP Drug Discovery Engine

GRAPHIC

8 Validate novel oncogenic driver mutations and targets Develop MasterKey inhibitors against mutation families Proprietary MAP scoring in silico MAP Drug Discovery Engine Unlocks Precision Medicine with a “MasterKey” Genomics Proteomics + Employ structural and dynamic methods to identify oncogenic mutation families Masterkey Therapy Oncogenicity Prediction Biological Validation Drug Discovery Conformation- based drug design

GRAPHIC

9 MAP Drug Discovery Engine: A Scaled Approach to Extract Oncogenic MasterKey Mutation Families Population level clinical mutation landscape Thousands Computational in-silico oncogenicity prediction Hundreds amino acid position prevalence amino acid position prevalence Oncogene amino acid position prevalence Experimentally validated oncogenic MasterKey mutations Tens Mutational Lollipop Plot

GRAPHIC

10 WILD TYPE MASTERKEY MUTATION FAMILIES MasterKey Inhibitors designed to exploit this vulnerability MasterKey mutations drive global conformation change Conformation Based Drug Design Enabled by MAP Drug Discovery Engine

GRAPHIC

BDTX-1535 Brain-Penetrant Inhibitor of GBM and NSCLC MasterKey EGFR Mutations

GRAPHIC

12 BDTX-1535: Oral, Brain Penetrant, Selective Inhibitor of Oncogenic EGFR MasterKey Mutations Designed to treat patients harboring EGFR oncogenic MasterKey mutations Irreversible Designed with selectivity versus WT-EGFR to deliver favorable safety profile Selective Designed to be brain penetrant to treat CNS tumors Brain Penetrant BDTX-1535 IND submitted for Phase 1 study in patients with either GBM or NSCLC with and without CNS tumors Clinical Stage CNS=Central Nervous System; WT=Wild-Type; GBM=Glioblastoma Multiforme; NSCLC=Non-Small Cell Lung Cancer

GRAPHIC

13 1 Van den Bent et al JCO 2009 2 Capuzzo et al Lancet 2010 GBM Mutations Time (months) p=0.003 PFS (%) GBM Trial1 Erlotinib in WT-EGFR GBM Patients Erlotinib in EGFR-vIII GBM Patients Current EGFR TKIs Do Not Extend PFS in Patients with GBM Mutations Erlotinib Placebo Time (months) PFS (%) HR=0.10 (0.04-0.25) log-rank p<0.0001 NSCLC Trial2 Kinase Domain Mutations EGFR TKIs Extend PFS in Patients with Kinase Domain Mutations 10-10 10-8 10-6 10-4 -0.4 0.0 0.4 0.8 1.2 1.6 2.0 [erlotinib] (M) P r o l i f e r a t i o n ( f o l d g r o w t h ) LoDi-EGFR (EGFR-Viii) KD-EGFR (E746-A750) 10-10 10-8 10-6 10-4 -0.4 0.0 0.4 0.8 1.2 1.6 2.0 [erlotinib] (M) P r o l i f e r a t i o n ( f o l d g r o w t h ) LoDi-EGFR (EGFR-Viii) KD-EGFR (E746-A750) EGFRvIII (GBM) EGFR Ex19del (NSCLC) Paradoxical Activation of EGFR in GBM Reversible EGFR Inhibitors Show Potentially Detrimental Pharmacology in EGFR Driven GBM TKIs=Tyrosine Kinase Inhibitors.

GRAPHIC

14 Black Diamond Revealed the Potential for Unwanted Paradoxical Activation of GBM Mutations by Reversible EGFR TKIs The oncogenic conformation of mutant EGFR in GBM is a locked dimer 0 12 24 36 48 0 1 2 3 4 5 Time (hrs) F r a c t i o n o f C o n t r o l P h o s p h o r y l a t i o n Reversible TKIs can stimulate the activity of mutant EGFR in GBM PDX tumors expressing EGFRvIII Inhibitors against EGFR mutants in GBM should be potent, selective & irreversible to avoid paradoxical activation paradoxical activation Single dose in vivo PK/PD study PK=Pharmacokinetics; PD=Pharmacodynamics; PDX=Patient-Derived Xenografts

GRAPHIC

15 BDTX-1535 Addresses Unique Pharmacology of EGFR Mutations in GBM to Achieve Sustained Inhibition and Activity in Preclinical Models Kpuu Partition Coefficient Calculation: AUCbrain:blood x plasma Fu/brain Fu; QD=quaque die (once a day) 12 14 16 18 0 25 50 75 100 Days Post Implantation P r o b a b i l i t y o f S u r v i v a l GBM6 Vehicle Control N=10 BDTX-1535 50mpk BDTX-1535 (50mpk QD) vehicle Average oral unbound brain fraction (Kpuu) = 0.55 in Rats Control 4hr 8hr 10hr 12hr 24hr 0 20000 40000 60000 80000 p E G F R ( p 1 0 6 8 ) Single dose in vivo PK/PD study (50mpk) Complete & sustained inhibition of pEGFR/pERK Increased survival of intracranial PDX tumors

GRAPHIC

16 H 2 9 2 A 4 3 1 E G F R v I I I E G F R v I I E G F R v V I E G F R R 1 0 8 K E G F R R 2 2 2 C E G F R C 2 3 1 F E G F R A 2 8 9 T E G F R A 2 8 9 V E G F R C 5 9 5 S E G F R G 5 9 8 V E G F R S 6 4 5 C 0 20 40 60 80 100 120 A n t i P r o l i f e r a t i v e I C 5 0 ( n M ) H 2 9 2 A 4 3 1 D 7 6 1 Y E 7 0 9 A E 7 0 9 G G 7 1 9 A G 7 1 9 C G 7 1 9 D G 7 1 9 R G 7 1 9 S S 7 6 8 I E 7 4 6 _ A 7 5 0 d e l + S 7 6 8 I E G F R E x 1 9 d e l E x 1 9 d e l + C 7 9 7 S E G F R L 8 5 8 R E G F R L 8 5 8 R + C 7 9 7 S 0 20 40 60 80 100 120 A n t i P r o l i f e r a t i v e I C 5 0 ( n M ) BDTX-1535 Optimized to Address a Wide Range of Oncogenic EGFR MasterKey Mutations and Amplification in GBM and NSCLC Anti - proliferation Ba/F3 IC50 ( nM ) Anti - proliferation Ba/F3 IC 50 ( nM ) EGFR mutations Intrinsic resistance mutations Classical & acquired resistance mutations EGFR WT (EGFR amp) Average 59-fold selectivity vs EGFR WT Average 132-fold selectivity vs EGFR WT EGFR WT (EGFR amp) EGFR variants and mutations prevalent in GBM EGFR mutations of intrinsic resistance and acquired resistance in NSCLC Average IC50 = 3.8 nM Average IC50 = 3.5 nM IC50=half maximal inhibitory concentration

GRAPHIC

17 BDTX-1535 Designed to Potently Inhibit EGFR Intrinsic and Acquired Resistance Mutations in NSCLC Intrinsic Resistance Mutations Designed for potent & selective inhibition across mutant families Osimertinib Altered Covalent Interaction Altered P-loop Interaction C797S G719X Gefitinib Acquired Resistance Mutations Designed to covalently target C797 & C797S

GRAPHIC

18 BDTX-1535 retains irreversible binding against C797S mutant BDTX-1535 demonstrates dose-dependent tumor regression in EGFR Ex19del / C797S tumor model BDTX-1535 Achieves Dose-dependent Tumor Regression in EGFR Mouse Models, Including Acquired Resistance Mutation C797S Osimertinib BDTX-1535 Washout Washout Control 0 4.0 10.0 24.0 Control 0 0.5 1 2 0 20 40 60 80 100 % p E G F R ( E x 1 9 d e l + C 7 9 7 S ) Time after washout (hrs) 1 3 5 7 9 11 0 200 400 600 800 Days After Dosing Start M e a n T u m o r V o l u m e m m ^ 3 Ba/F3 EGFR Ex19del+C797S Vehicle BDTX-1535 40mpk Osimertinib 25mpk

GRAPHIC

19 EGFR AMP EGFR AMP EGFR vII EGFR AMP EGFR Ex19del EGFR C595F EGFR G719C EGFR AMP EGFR Ex19del EGFR Ex19del +C797S BDTX-1535 Promotes Regression Across Range of GBM & NSCLC PDX Tumors Expressing MasterKey EGFR Mutations & EGFR Amplification PDX Models -100 100 % Regression -10 10 AMP=Amplification

GRAPHIC

20 BDTX-1535: Focused, Biomarker-Driven First-in-Human Phase 1 Study Design Selected Patient Population • GBM Patients with EGFR alterations • NCSLC Patients with EGFR intrinsic resistance mutations • NCSLC Patients with acquired EGFR resistance mutations RP2D Dose Escalation EGFR MasterKey Mutations EGFR MasterKey Mutations RP2D=Recommended Phase 2 Dose

GRAPHIC

21 ~160,500 Sources: Epidemiology data from EvaluatePharma Addressable Patient Population (US / EU / Japan / China) Classical Acquired Resistance EGFRmut 11,700 Intrinsic Resistance EGFRmut 139,700 Classical EGFRmut 9,100 EGFRmut Newly Diagnosed GBM 36,400 24,700 EGFRmut Recurrent GBM ~61,100 Large Addressable Patient Population Harboring MasterKey Mutations Across GBM and NSCLC

GRAPHIC

22 BDTX-1535 is Well Positioned to Address Unmet Needs in EGFR Mutant GBM/NSCLC Potent & selective inhibition of EGFR mutations (Avg IC50 ~3nM) that drive intrinsic and acquired resistance to current generation TKIs • Irreversible inhibition of GBM mutations to avoid paradoxical activation • Irreversible binding to C797 and C797S acquired resistance in NSCLC • Regression across panel of PDX tumor models harboring EGFR mutations in GBM and NSCLC Robust brain penetration to treat patients with EGFR mutations and CNS tumors • Unbound brain fraction (Kpuu) = 0.55 in rat; activity demonstrated in intracranial GBM model Favorable drug like properties • Prolonged blood stability • Projected t1/2 of 15 hours for QD dosing t1/2=half-life; IC50 =half maximal inhibitory concentration

GRAPHIC

BDTX-189 Inhibitor Targeting MasterKey EGFR/HER2 Mutations

GRAPHIC

24 BDTX-189: MasterKey EGFR/HER2 Inhibitor, Early Proof-of-Concept Demonstrated FDA endorsed RP2D 800 mg QD non-fasting PK in-line with preclinical predictions RP2D Achieved Anti-tumor activity in NSCLC EGFR Ex20ins and in HER2 amplified solid tumors Activity Observed Well-tolerated, minimal evidence of systemic WT- EGFR related adverse events Favorable Safety BDTX-189 Continuing enrollment into Safety Expansion cohort to gate next steps in development MasterKey-01 Clinical Trial

GRAPHIC

25 BDTX-189 is a Potent and Selective Inhibitor Across EGFR/HER2 MasterKey Mutation Family EC domain 17 mutants JM domain 2 mutants Ex 20 ins 24 mutants Kinase domain 18 mutants A 4 3 1 H 2 9 2 E G F R A 7 6 3 - Y 7 6 4 i n s F Q E A E G F R A 7 6 7 - V 7 6 9 d u p A S V E G F R A 7 6 7 - V 7 6 9 d u p S V A E G F R S 7 6 8 - D 7 7 0 d u p S V D E G F R D 7 7 0 - N 7 7 1 i n s G E G F R D 7 7 0 - N 7 7 1 i n s G Y N E G F R D 7 7 0 - P 7 7 2 d u p D N P E G F R N 7 7 1 - H 7 7 3 d u p N P H E G F R P 7 7 2 - H 7 7 3 d u p P H E G F R H 7 7 3 - V 7 7 4 d u p H V E G F R H 7 7 3 - V 7 7 4 i n s A H E G F R H 7 7 3 d u p H H E R 2 A 7 7 5 - G 7 7 6 i n s S V M A H E R 2 A 7 7 5 - G 7 7 6 i n s V V M A H E R 2 P 7 8 0 - Y 7 8 1 i n s G S P H E R 2 V 7 7 7 - G 7 7 8 i n s V H E R 2 A 7 7 5 - G 7 7 6 i n s Y V M A H E R 2 G 7 7 8 - S 7 7 9 i n s G H E R 2 G 7 7 8 - S 7 7 9 i n s C P G H E R 2 G 7 7 6 i n s V C H E R 2 V 7 7 7 - G 7 7 8 i n s C H E R 2 G 5 8 R H E R 2 R 1 0 3 Q H E R 2 P 1 2 2 L H E R 2 V 2 1 9 I H E R 2 G 2 2 9 R H E R 2 G 2 9 2 C H E R 2 G 2 9 2 R H E R 2 A 2 9 3 T H E R 2 S 3 1 0 F H E R 2 S 3 1 0 Y H E R 2 A 5 1 6 T H E R 2 A 5 8 8 V H E R 2 G 6 0 3 C H E R 2 G 6 6 0 D H E R 2 R 6 7 8 Q H E R 2 Q 7 0 9 L H E R 2 E 7 1 7 V H E R 2 T 7 3 3 I H E R 2 L 7 5 5 S H E R 2 D 7 6 9 H H E R 2 D 7 6 9 Y H E R 2 G 7 7 6 V H E R 2 V 7 7 7 L H E R 2 V 8 4 2 I H E R 2 T 8 6 2 A H E R 2 L 8 6 9 R H E R 2 R 1 2 3 0 Q 1 10 100 1000 A n t i - p r o l i f e r a t i v e I C 5 0 ( n M ) WT-EGFR Average selectivity > 50-fold Exon 20 insertion Extracellular domain Juxtamembrane domain Kinase domain Average IC50 = 16.5 nM EC domain, extracellular domain; JM domain, juxtamembrane domain; Ex 20 ins, Exon 20 insertions Phenotypic Activity Against MasterKey Mutation Family & Selectivity vs WT-EGFR Oncogenic MasterKey Mutation Family EGFR/HER2

GRAPHIC

26 MasterKey-01 Dose Escalation Complete; Safety Expansion Enrolling Recommended Phase 2 Dose (RP2D) • 800 mg QD non-fasting • Well tolerated, no DLTs at RP2D • Most common AE grade 1 diarrhea, no AE ≥ grade 2 • Low rate of rash Safety Summary • Safety dataset dose escalation QD cohorts n=59* • n=18 at 800 mg QD fasting • n=13 at 800 mg QD non-fasting Phase 1 QD Dose Escalation RP2D Safety Expansion Currently Enrolling Fasting Non-Fasting FDA Endorsed RP2D: 800mg QD Non- fasting 25mg QD 50 100 200 400 800 1200 1000 800 *Includes 18 patients from food-effect cohort; DLT=dose limiting toxicity; AE=adverse events Safety Expansion Cohort • Gate future development as single agent and in combination

GRAPHIC

27 Targeted selectivity E r l o t i n i b P o z i o t i n i b A f a t i n i b O s i m e r t i n i b C L N - 0 8 1 / T A S 6 4 1 7 M o b o c e r t i n i b / T A K 7 8 8 B D T X - 1 8 9 0.1 1 10 100 F o l d S e l e c t i v i t y ( E G F R W T / M u t a n t ) > Poor selectivity Improved Selectivity Drives Improved Safety – Potential to Limit Overlapping Toxicities in Combination Treatment related AEs by max grade observed in >10% of patients 800 mg QD, Non-fasting (N=15) * Mutations assessed: HER2 S310F, HER2 P95, HER2 R678Q, HER2 L755S, HER2 V777L, HER2 YVMA, HER2 GSP, EGFRvIII, EGFR NPH, EGFR ASV BDTX-189 designed for superior selectivity profile Improved Safety - Limited skin and manageable gastro-intestinal toxicity at RP2D *Rash = Dermatitis acneiform and Rash NCT04209465 is an ongoing study and is undergoing active data entry and cleaning. Includes AEs with start date prior to Oct. 30, 2021

GRAPHIC

28 Radiologic Scans NSCLC EGFR Ex 20 Ins: 51-year-old Woman with 53% Tumor Reduction at Week 12 and Improved Diffuse Lung Disease SLD=Sum of longest diameter; PR=partial response; PD=progressive disease Genomic alteration: EGFR Ex20ins (SVD), HER2-amp Prior therapy: • Poziotinib with PR and toxicity followed by PD • Carboplatin and pemetrexed Response to BDTX-189: • BDTX-189, 800 mg QD non-fasting • Controlled, low-grade diarrhea • Off oxygen starting at ~9 weeks • On study with PR, beyond 14+ Cycles (10 months) Baseline Week 6 NCT04209465 is an ongoing study and is undergoing active data entry and cleaning. Includes data prior to 30 Oct 2021 Target Lesions Over Time

GRAPHIC

29 Early PoC Achieved for BDTX-189 in Solid Tumors with HER2-Amp with Deep and Durable Confirmed PR Observed Radiologic Scans Genomic alteration: HER2-amplification Prior therapy: • Carbo/taxol, FOLFIRINOX, Everolimus Response to BDTX-189: • BDTX-189 800 mg QD fasting • Had brief episodes of Grade 1 diarrhea • Off study due to PD after 17+ Cycles (12 months) 100% regression Target Lesions Over Time Cancer of Unknown Primarya (HER2 Amplification): 73-year-old Woman with Deep and Durable Confirmed PR aCUP presumed by the investigator of this patient to be of breast cancer, pancreato-biliary or upper gastrointestinal origin. Baseline 7 months NCT04209465 is an ongoing study and is undergoing active data entry and cleaning. Includes data prior to Oct. 30, 2021

GRAPHIC

BRAF Program Oral, Brain Penetrant, MasterKey Inhibitor

GRAPHIC

31 BDTX-BRAF: Oral, Brain Penetrant, Inhibitor of Oncogenic BRAF Mutations BDTX-BRAF Designed for potent and selective inhibition of Class I, II, and III BRAF mutations MasterKey Family Inhibits dimerized RAF kinases and avoids paradoxical activation Avoids Paradoxical Activation Designed to be brain penetrant to treat CNS tumors Brain Penetrant Initiation of IND-enabling studies expected in 2022 Status

GRAPHIC

32 BDTX-BRAF: Designed with Potential Best-In-Class Target Product Profile Potent Inhibition of All Three Class I / II / III BRAF Mutations Brain Penetration High incidence of CNS metastasis among tumors express BRAF mutations (lung, melanoma) High prevalence of BRAF mutations in primary CNS malignancies (glioma) Avoidance of Paradoxical Activation Class I Mutants (V600) Class II Mutants Class III Mutants PP RAS Independent BRAF Monomer RAS Independent BRAF Mutant Homodimers RAS Dependent BRAF Mutant Heterodimers Impaired Kinase Activity BRAF mut BRAF mut RAS GDP RAS GDP RAS GTP BRAF V600E CRAF BRAF mut Designed for • Superior breadth & depth of activity • Lack of cutaneous toxicity -11 -10 -9 -8 -7 -6 -5 0 1 2 3 4 WT BRAF, NRASQ61R (SK-MEL-2) Concentration Log[M] p - E R K N o r m a l i z e d t o D M S O c o n t r o l Vemurafenib Encorafenib BDTX-A paradoxical activation

GRAPHIC

33 0 5 10 15 20 0 500 1000 1500 2000 Days (post treatment) T u m o r V o l u m e ( m m 3 ) Vehicle BDTX-BRAF (10 mpk) Belvarafenib (15 mpk) BDTX-BRAF (3 mpk) 0 3 6 9 12 0 500 1000 1500 2000 Days (post treatment) T u m o r V o l u m e ( m m 3 ) Encorafenib (60 mpk) Vehicle BDTX-BRAF (3 mpk) 0 5 10 15 0 70 140 210 280 350 Days (post treatment) T u m o r V o l u m e ( m m 3 ) Vehicle Encorafenib (60 mpk) BDTX-BRAF (5 mpk) BRAF Class II Mutant Tumor Model BRAF Class III Mutant Tumor Model BRAF Class I Mutant Tumor Model BDTX-BRAF Exhibits Strong Anti-Tumor Activity Across All BRAF Mutation Classes in In Vivo Models

GRAPHIC

34 Day 0 Day 15 BDTX-BRAF prolongs survival in BRAF-V600E intracranial tumor model Vehicle BDTX-BRAF 3 mpk Belvarafenib 15 mpk BDTX-BRAF Is Brain Penetrant and Exhibits Robust Activity in Treating CNS Disease in in vivo models BDTX-BRAF 10 mpk

GRAPHIC

35 BDTX BRAF: Potential Best-in-Class, Masterkey Inhibitor For A Greater Number Of Patients With Overlooked Oncogenic Mutations Sources: EvaluatePharma Epi and GENIE/TCGA 47,500 NRAS Melanoma 51,600 BRAF Class II/III alt. Solid Tumors 91,810 BRAF V600 Solid Tumors Addressable US / EU / JP Patient Population ~190,910 BDTX Is Growing The Addressable Patient Population By De-Orphaning of Overlooked Mutations # of Known Oncogenic Mutations # of BDTX MAP Engine De-orphaned Oncogenic Mutations Growing +

GRAPHIC

Corporate Overview

GRAPHIC

37 Deep Oncology and Small Molecule Drug Discovery and Development Experience David M. Epstein, Ph.D. President & CEO Liz Buck, Ph.D. Chief Scientific Officer Brent Hatzis-Schoch, J.D. COO and General Counsel Leadership Team Board of Directors Ali Behbahani, M.D. General Partner, NEA Brad Bolzon, Ph.D. Managing Director, Versant Ventures Kapil Dhingra, M.D. Managing Member, KAPital Consulting David M. Epstein, Ph.D. CEO, Black Diamond Therapeutics Bob Ingram – Chairman General Partner, Hatteras Ventures Sam Kulkarni, Ph.D. CEO, CRISPR Therapeutics AG Alex Mayweg, Ph.D. Managing Director, Versant Ventures GarryMenzel CEO, TCR2 Rajeev Shah Managing Director, RA Capital Mark Velleca, M.D., Ph.D. Venture Partner, Hatteras Ventures Karsten Witt, M.D. Interim Chief Medical Officer Fang Ni, Pharm.D. Chief Business Officer and Chief Financial Officer

GRAPHIC

38 Expanding Precision Medicine Through the Development of MasterKey Therapies Our proprietary MAP drug discovery engine is designed to: • Predict and validate novel oncogenic mutant families from population level tumor genomics • Pioneer mutant family conformation-based MasterKey drug design • Provides opportunities beyond oncology and small molecules MasterKey therapies designed to address overlooked mutation families; providing precision oncology to greater numbers of patients with genetically defined tumors Proof-of-principle mutation family selective inhibitors; clinical and late-preclinical pipeline of MasterKey inhibitors derived from our MAP drug discovery engine: EGFR, HER2, BRAF, and FGFR BDTX-1535, an irreversible, mutant selective, brain-penetrant EGFR inhibitor entering Phase 1 for the treatment of GBM and NSCLC driven by EGFR intrinsic & acquired resistance mutations BDTX-189, a mutant selective EGFR/HER2 inhibitor in Phase 1 Safety Expansion study in patients expressing targeted oncogenic mutations

GRAPHIC

Thank You