A Phase 1/2 Study Of CFT1946, A Novel Bifunctional Degradation Activating Compound, or BiDAC Degrader, of Mutant BRAF V600 as Monotherapy and in Combination with Trametinib, in Mutant BRAF V600 Solid Tumors

Meredith McKean, MD1, Alexander I. Spira, MD, PhD2, Ezra Rosen, MD, PhD3, Vivek Subbiah, MD4, Victor Moreno, MD5, Valentina Gambardella, MD6, Maria Vieito, MD7, Omar Saavedra Santa Gadea, MD8, Sophie Cousin, MD9,

Philippe Alexandre Cassier, MD10, Riadh Lobbardi, PhD11, Oliver Schönborn-Kellenberger, MSc11, Eunju Hurh, PhD11, Mary M. Ruisi, MD11, Elizabeth I. Buchbinder, MD12

1Tennessee Oncology (Sarah Cannon Research Institute), Nashville, TN; 2Virginia Cancer Specialists, Fairfax, VA; 3Memorial Sloan Kettering Cancer Center, New York, NY; 4MD Anderson Cancer Center, Houston, TX; 5Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain; 6Institute of Hospital Clinico de Valencia, Valencia, Spain;

#356a

7Hospital Universitario Vall d'Hebron, Barcelona, Spain; 8Next Oncology, Barcelona, Spain; 9Institut Bergonié, Department of Medical Oncology, Bordeaux, France; 10Léon Bérard Center, Lyon, France; 11C4 Therapeutics, Inc., Watertown, MA; 12Dana-Farber Cancer Institute, Boston, MA

BACKGROUND

FIRST-IN-HUMAN STUDY DESIGN

  • BRAF is a protein kinase that acts as a signal transducer/amplifier in receptor tyrosine kinase (RTK) signaling pathways, specifically, the mitogen activated protein kinase (MAPK) pathway that promotes cell proliferation and survival when activated through extracellular signals1,2
  • Constitutively active mutated BRAF, specifically BRAF with valine 600 mutations (BRAF V600), is capable of uncontrolled signaling which signals as a monomer, resulting in hyperactivation of MEK, ERK, and dysregulation of cellular proliferation2

PRE-CLINICAL DATA: IN VIVO (continued)

Figure 5: CFT1946 Induces Tumor Regression in the A375 (homozygous BRAF V600E) Xenograft Mouse Model as a Single Agent and in Combination With the MEK Inhibitor, Trametinib, in a BRAF Inhibitor-Resistant Xenograft

5

KEY ELIGIBILITY CRITERIA6

KEY INCLUSION CRITERIA

KEY EXCLUSION CRITERIA

≥18 years of age at time of informed consent

Subject has had major surgery within 21 days prior

Documented evidence of a BRAF V600

to the planned first dose. Minor surgery is

BRAF V600 is a clinically

validated oncology target in

the follow tumor types1-3

Melanoma, colorectal

cancer (CRC), non-small

cell lung cancer (NSCLC),

and anaplastic thyroid

carcinoma (ATC)

Currently approved BRAF

inhibitors (BRAFi) result in

Figure 1: Utilizing a Degrader Approach to Overcome Limitations of BRAF Inhibition5

Mouse Model

A. CFT1946 Treatment of A375 Cell Line In Vivo Shows Dose-Dependent Tumor Regression Superior to Inhibitor

mutation obtained from tumor tissue or

permitted within

liquid biopsy

21 days prior to enrollment

Received ≥1 prior line of SoC therapy for

Subject with CNS involvement (primary tumor or

unresectable locally advanced or metastatic

metastatic disease), except if clinically stable

disease, NSCLC, CRC, ATC or other BRAF V600

Subject with known malignancy other than trial

mutation-positive tumors

indication that is progressing or has required

Adequate bone marrow, liver, renal, and

treatment within the past 3 years, except for

cardiac organ function

conditions that have undergone potentially

curative therapy

paradoxical RAF activation

as the mutant BRAF protein

is still able to dimerize with

wtBRAF, resulting in a

dimeric signaling complex4

(Figure 1)

volume (mm3)

2000

1600

1200

Vehicle, PO BID Encorafenib, 35 mg/kg PO QD CFT1946, 0.3 mg/kg PO BID CFT1946, 3 mg/kg PO BID CFT1946, 10 mg/kg PO BID

STUDY ENDPOINTS6

PRIMARY ENDPOINTS

SECONDARY ENDPOINTS

Frequency and severity of AEs and SAEs of

Frequency and severity of AEs and SAEs of CFT1946

CFT1946 (Phase 1)

(Phase 2)

Incidence of DLTs (Phase 1)

Number of participants with changes between

Number of participants with changes between

baseline and post-baseline safety assessments

CFT1946 BACKGROUND5

  • CFT1946 is a novel, orally bioavailable, bifunctional degradation activating compound, or BiDAC degrader
    • CFT1946 selectively inhibits and degrades mutant BRAF V600 protein
    • Distinct from approved BRAFi, CFT1946 avoids paradoxical RAF activation as the degraded BRAF V600 mutant protein can no longer incorporate into a dimeric signaling complex (Figure 1)
  • CFT1946 is selective for the Figure 2: Mechanism of Action for CFT1946 Compound5
    mutant protein and spares wtBRAF V600
  • Mechanism of action

(Figure 2)

i. CFT1946 induces

A375 tumor

800

400

0

0

5

10

15

20

25

Treatment days

30

35

baseline and post-baseline safety assessments

(Phase 2)

(Phase 1)

Frequency of dose interruptions and dose reductions

Frequency of dose interruptions and dose

(Phase 2)

reductions (Phase 1)

Assessment of PK and PD

Frequency of AEs leading to discontinuation

PK-QTcF relationship

(Phase 1)

ORR (Phase 1 and 2)

ORR (Phase 2)

DCR

PFS

DOR

STUDY STATUS/ENROLLMENT6

The study opened to accrual in December 2022 and will be recruiting ~N=135patients from 11 sites*

ternary complex

formation with BRAF

and cereblon E3 ligase

(step 1)

ii. BRAF V600 is

ubiquitinated and

subsequently released

for degradation in

the proteasome

(steps 2-4)

CFT1946 has demonstrated

preclinical activity in BRAF

V600 mutant in vitro and

in vivo models, including

models resistant to BRAFi

(Figures 3-5)

PRE-CLINICAL DATA: IN VITRO

Figure 3: CFT1946 Is an On-Mechanism,CRBN-Based, Highly Selective BRAF V600X BiDAC Degrader5

A. CFT1946 Degrades

B.

CFT1946 is an

C.

Proteome Profiling

BRAF V600E in a Dose-

On-Mechanism

Demonstrates Selectivity of

Dependent Manner

BiDAC Degrader

CFT1946 for BRAF V600E

CFT1946 (300 nM, 24 h) in A375 Cells

Remaining%

dabrafenibvs.value-p

15

Emax = 26%

CFT1946 (100 nM) in A375 cells @ 24 h

100

10

75

BRAF V600E

50

pERK

DC50 = 14 nM

25

BRAF V600E

BRAFV600E

5

0

Vinculin

10

LOXL4

10

0

10

1

10

2

10

3

10

4

-Log

[CFT1946] (nM)

ARAF

*note: +/- refers to presence or absence of 100 nM CFT1946

RAF1

0

-4

-2

0

2

4

Log

Fold Change vs. dabrafenib

2

  • CFT1946 acts as both an inhibitor and a degrader of BRAF V600E as demonstrated in HiBiT assay in panel A. Phospho-ERK levels decrease in a CFT1946 dose-dependent manner. BRAF V600E levels decrease in a CFT1946 dose-dependent manner until "hook effect" concentrations are achieved. Panel B demonstrates that CFT1946 is on-mechanism for a CRBN-based BiDAC™ degrader and Panel C shows the selectivity of CFT1946 in A375 cells using global proteomic profiling.

PRE-CLINICAL DATA: IN VIVO

Figure 4: Dose Proportional PK and PD Profile in the BRAF V600E A375 Xenograft Mouse Model for CFT1946 Compound5

Dose Proportional PK and PD for CFT1946

A.

(ng/ml)plasmain

B.

concentrationCFT1946

100000

expressionproteinBRAF

vehicletorelative

1.5

10000

0.3 mg/kg PO

0.3 mg/kg PO

1000

3 mg/kg PO

1.0

3 mg/kg PO

100

10 mg/kg PO

0.5

10 mg/kg PO

10

1

0.0

0

12

24

36

48

0

12

24

36

48

Time (h)

Time (h)

C.

(ng/g)tumorin

D.

concentrationCFT1946

100000

expressionproteinpERK

vehicletorelative

1.5

10000

0.3 mg/kg PO

0.3 mg/kg PO

1000

3 mg/kg PO

1.0

3 mg/kg PO

100

10 mg/kg PO

0.5

10 mg/kg PO

10

1

0.0

0

12

24

36

48

0

12

24

36

48

Time (h)

Time (h)

  • Pharmacodynamic data demonstrates a dose-proportional loss of BRAF V600E protein and decreased signaling through the MAPK pathway as determined by loss of phospho-ERK (pERK). As CFT1946 degrades BRAF V600 mutant protein and results in decreases phospho-ERK levels, some MAPK pathway inhibition occurs prior to maximal loss of BRAF V600E protein.
  1. Combination Treatment of BRAFi-Resistant Xenograft Model With CFT1946
    and MEKi Shows Tumor Growth Inhibition/Regression

A375 + NRAS-Q61K Xenograft

)

2500

3

(mm

Vehicle (PO BID)

volume

2000

MEKi: Trametinib (0.1 mg/kg BID)

Encorafenib (35 mg/kg QD + MEKi)

tumor

1500

CFT1946 (10 mg/kg BID)

NRAS

1000

CFT1946 (30 mg/kg BID)

500

A375

CFT1946 (10 mg/kg BID) + MEKi

CFT1946 (30 mg/kg BID) + MEKi

0

0

7

14

21

Treatment days

  1. In the A375 mouse xenograft model, CFT1946 showed dose-dependent tumor growth inhibition (TGI) when administered PO at 0.3 mg/kg and 3 mg/kg BID, and immediate and sustained tumor regression at 10 mg/kg PO BID.
  2. In the BRAFi-resistantA375+NRAS-Q61K xenograft model, CFT1946 showed dose-dependent TGI when administered PO as a single agent at 10 mg/kg (TGI ~35%) and 30 mg/kg (TGI ~60%) BID.
    When CFT1946 at 10 and 30 mg/kg BID was combined with 0.1 mg/kg trametinib, combination treatment resulted in tumor regression up to Day 16 and Day 19, respectively, with slight tumor re-growth emerging at Day 19 and Day 21, respectively.

These preclinical data provide rationale for a first-in-human (FIH) study to

evaluate CFT1946 in BRAF V600 mutant solid tumors

PHASE 1/2: FIRST-IN-HUMAN CLINICAL STUDY DESIGN5,6

  • Open-label,multicenter, Phase 1/2 clinical trial with dose escalation and expansion phases*
  • Dose escalation Phase 1 comprises CFT1946 (Arm A) with a starting oral dose of 20 mg twice daily and CFT1946+trametinib (Arm B)
  • Dose expansion Phase 2 comprises CFT1946 (Arm A1) and CFT1946+trametinib (Arms B1 and B2)
    • Arms A1 and B1 will include BRAF V600 mutant melanoma and NSCLC with prior BRAFi therapy while Arm B2 will enroll BRAF V600 mutant NSCLC who are BRAFi-naïve
  • N=135 (approximately) across 11 US and European sites will be enrolled
  • Registered on ClinicalTrials.gov as NCT05668585, study is open for enrollment

Figure 6: CFT1946 Study Design5‡

PHASE 1

PHASE 2

Dose Escalation Monotherapy

Dose Expansion Monotherapy

Arm A:

Expansion Arm A1:

CFT1946 Monotherapy

V600 Solid Tumors (non-CNS)

CFT1946 Monotherapy

RP2D

(s/p BRAFi for lung, CRC, melanoma, ATC)

V600 Melanoma + NSCLC (s/p BRAFi)

3-6 patients/cohort, BLRM

N=~30

N=~40

Dose Expansion: combination

Dose Escalation: combination

Expansion Arm B1:

Arm B:

CFT1946 + trametinib

Dose determined

CFT1946 + trametinib

V600 Melanoma + NSCLC (s/p BRAFi)

to be safe by SRC

V600 Solid tumors (non-CNS) (s/p

N=~20

RP2D

BRAFi for lung, melanoma, ATC, CRC)

Expansion Arm B2:

3-6 patients/cohort, BLRM

CFT1946 + trametinib

N=~25

V600 NSCLC (BRAFi naÏve)

N=~20

*CFT1946 is administered orally in 28-day cycles until disease progression or intolerable toxicity.

Phase 2 will be initiated once the RP2D has been identified. Eligible subjects are ≥18 years-old with documented BRAF V600 mutant cancers who have received ≥1 prior therapy.

Phase 2 expansion study design will be updated based on Health Authority feedback.

in the US and Europe

  • Trial registration: NCT05668585
  • As of 05/15/2023, all 5 US sites have initiated recruitment
  • Contact information: clinicaltrials@C4therapeutics.com

United States

WA

ME

MT

ND

NH

MN

VT

OR

NY

MA

WI

CT

RI

ID

SD

MI

WY

PA

IA

NJ

NV

NE

IN

OH

DE

IL

UT

CO

KS

MO

WV

VA

MD

CA

KY

NC

TN

OK

SC

AZ

NM

AR

AL

MS

GA

TX

LA

AK

FL

HI

Europe

FR

ES

*Blue-colored US states and EU countries indicate clinical trial sites. 2 sites are located in France and 4 sites in Spain.

Abbreviations

AE, adverse event; ATC, anaplastic thyroid carcinoma; BID, twice daily; BiDACTM, bifunctional degradation activating compound; BLRM, Bayesian logistic regression model; BRAF, v-raf murine sarcoma viral oncogene homolog B1; BRAFi, BRAF inhibitor, Bx, biopsy; CNS, central nervous system; CRBN, cereblon; CRC, colorectal cancer; DCR, disease control rate; DLT, dose limiting toxicities; DOR, duration of response; ERK, extracellular signal-regulated kinases; FIH, first-in-human;HiBiT, high affinity bioluminescent tag; IMiD, immunomodulatory imide drug; MAPK, mitogen-activated protein kinase; MEK, mitogen-activated protein kinase kinase; MEKi, MEK inhibitor; MTD, maximum tolerated dose; NRAS, neuroblastoma ras; NSCLC, non-small cell lung cancer; ORR, overall response rate; pERK, phospho-ERK;PD, pharmacodynamics; PDX, patient-derived xenografts; PFS, progression-free survival; PK, pharmacokinetics; PO, by mouth; PoM, proof of mechanism; QD, once daily; QTcF, Fridericia heart-rate- corrected QT interval; RAF, rapidly accelerated fibrosarcoma; RP2D, recommended phase 2 dose; RTK, receptor tyrosine kinase; SAE, serious adverse event; s/p, status post; SoC, standard-of-care;SRC, Safety Review Committee; TGI, tumor growth inhibition; wt, wild-type.

Disclosures

  1. consulting or advisory role: AstraZeneca, Pfizer, Astellas Pharma, Bicycle Therapeutics, Castle Biosciences, Eisai, IDEAYA Biosciences, iTeos Therapeutics, Moderna Therapeutics; research funding: Prelude Therapeutics, Genentech, Tizona Therapeutics, Inc., GlaxoSmithKline, IDEAYA Biosciences, Exelixis, Jacobio, Moderna Therapeutics, Regeneron, Epizyme, TopAlliance Biosciences, Ascentage Pharma Group, Oncorus, Ikena Oncology, Bicycle Therapeutics, Tmunity Therapeutics, Inc., Sapience Therapeutics, NBE Therapeutics, Dragonfly Therapeutics, Infinity Pharmaceuticals, Novartis, Plexxikon, Alpine Immune Sciences, Arcus Biosciences, Arvinas, Bayer, BioMed Valley Discoveries, BioNTech, EMD Serono, Erasca, Inc, Foghorn Therapeutics, Gilead Sciences, ImmVira, KeChow Pharma, Kezar Life Sciences, Kinnate Biopharma, MedImmune, Mereo BioPharma, Metabomed, Nektar, PACT Pharma, Pfizer, Pyramid Biosciences, Scholar Rock, Synthorx, Takeda, TeneoBio, Tempest Therapeutics, Xilio Therapeutics, AADI, Accutar Biotech, Astellas Pharma, G1 Therapeutics, OncoC4, Poseida Therapeutics. AIS: consulting or advisory role: Array BioPharma, Incyte, Amgen, Novartis, AstraZeneca/MedImmune, Mirati Therapeutics, Gritstone Bio, Jazz Pharmaceuticals, Merck, Bristol Myers Squibb, Takeda, Janssen Research & Development, Mersana, Blueprint Medicines, Daiichi Sankyo/Astra Zeneca, Regeneron, Lilly, Black Diamond Therapeutics, Sanofi; leadership: Next Oncology; stock and other ownership interests: Lilly; honoraria: CytomX Therapeutics, AstraZeneca/MedImmune, Merck, Takeda, Amgen, Janssen Oncology, Novartis, Bristol Myers Squibb, Bayer; research funding: Roche, AstraZeneca, Boehringer Ingelheim, Astellas Pharma, MedImmune, Novartis, NewLink Genetics, Incyte, AbbVie, Ignyta, LAM Therapeutics, Trovagene, Takeda, MacroGenics, CytomX Therapeutics, LAM Therapeutics, Astex Pharmaceuticals, Bristol Myers Squibb, Loxo, Arch Therapeutics, Gritstone Bio, Plexxikon, Amgen, Loxo, Daiichi Sankyo, ADC Therapeutics, Janssen Oncology, Mirati Therapeutics, Rubius Therapeutics, Synthekine, Mersana Therapeutics, Blueprint Medicines, Regeneron, Alkermes, Revolution Medicines, Medikine, Synthekine, Black Diamond Therapeutics, BluPrint Oncology, nalo therapeutics. ER: no disclosures. VS: consulting or advisory role: MedImmune, Helsinn Therapeutics, Relay Therapeutics, Pfizer, Loxo/Lilly; travel, accommodations, expenses: PharmaMar, Bayer, Novartis, Helsinn Therapeutics, Foundation Medicine; other relationship: Medscape; research funding: Novartis, GlaxoSmithKline, NanoCarrier, Northwest Biotherapeutics, Genentech/Roche, Berg, Bayer, Incyte, Fujifilm, PharmaMar, D3 Oncology Solutions, Pfizer, Amgen, AbbVie, MultiVir, Blueprint Medicines, Loxo, Vegenics, Takeda, Alfasigma, Agensys, Idera, Boston Biomedical, Inhibrx, Exelixis, Amgen, Turning Point Therapeutics, Relay Therapeutics. VM: employment: START; consulting or advisory role: Merck, Bristol Myers Squibb, Bayer, Janssen Oncology, Roche, Basilea, Affimed Therapeutics, AstraZeneca; speakers' bureau: Bayer, Pierre Fabre; travel, accommodations, expenses: Sanofi/Regeneron; expert testimony: Medscape/Bayer, Nanobiotix; other relationship: Bristol Myers Squibb, research funding: AbbVie, ACEA Biosciences, Adaptimmune, Amgen, AstraZeneca, Bayer, BeiGene, Bristol Myers Squibb, Boehringer Ingelheim, Celgene, Eisai, E-therapeutics, GlaxoSmithKline, Janssen, Menarini, Merck, Nanobiotix, Novartis, Pfizer, PharmaMar, PsiOxus Therapeutics, Puma Biotechnology, Regeneron, Rigontec, Roche, Sanofi, Sierra Oncology, Synthon, Taiho Pharmaceutical, Takeda, Tesaro, Transgene. VG: no disclosures.
    MV: consulting or advisory role: Roche, BMS GmbH & Co. KG; travel, accommodations, expenses: Roche; other relationships: Roche, BMS GmbH & Co. KG, Debiopharm
    Group, Incyte, Novartis, PharmaMar, Mundipharma, Taiho Oncology, SERVIER, HUTCHMED; research funding: Novartis, Roche/Genentech, Thermo Fisher Scientific,
    AstraZeneca, BeiGene, Taiho Oncology. OSSG: travel, accommodations, expenses: Affimed. SC: no disclosures. PAC: consulting or advisory role: OSE Immunotherapeutics,
    Bristol Myers Squibb/Celgene, Janssen Oncology, Boehringer Ingelheim; travel, accommodations, expenses: Roche, OSE Immunotherapeutics, Novartis; honoraria: Amgen;
    research funding: Novartis, Roche/Genentech, Lilly, Blueprint Medicines, Bayer, AstraZeneca, Celgene, Plexxikon, AbbVie, Bristol-Myers Squibb, Merck Sharp & Dohme,
    Taiho Pharmaceutical, Toray Industries, Transgene, Loxo, GlaxoSmithKline, Innate Pharma, Janssen, Boehringer Ingelheim. RL: employed by and equity holder in
    C4 Therapeutics. OSK: consulting or advisory role: C4 Therapeutics. EH: employed by and equity holder in C4 Therapeutics. MMR: employed by and equity holder in
    C4 Therapeutics. EIB: employment: Alexion Pharmaceuticals (an immediate family member), Takeda (an immediate family member); consulting or advisory role: Nektar,
    Instil Bio, Novartis, Xilio Therapeutics, Sanofi, Merck, Iovance Biotherapeutics; research funding: Bristol-Myers Squibb, Checkmate Pharmaceuticals, Novartis, Lilly, Genentech/Roche.

Acknowledgements

References

We would like to thank the site support staff, study sponsor,

1.

Ascierto PA, et al. J Transl

4.

Hoyer S, et al. Int J Mol Sci.

Copies of this poster

and collaborators as well as participating patients and their

Med. 2012;10:85.

2021;22(21):11951.

obtained through QR Code

families for their contributions to the study. This study is

2.

Zheng G, et al. BMC Cancer.

5.

C4 Therapeutics data on file.

are for personal use only

sponsored by C4 Therapeutics, Inc. Editorial support was

2015;15:779.

6.

NCT05668585.

and may not be reproduced

provided by Red Nucleus and funded by C4 Therapeutics, Inc.

3.

Chang CF, et al. Am J Cancer

www.clinicaltrials.gov.

without permission from

All authors contributed to and approved the presentation.

Res. 2022;12(11):5342-5350.

Accessed March 3, 2023.

ASCO and the author.

Presented at The American Society of Clinical Oncology Annual Meeting (ASCO 2023), June 2-6

Attachments

  • Original Link
  • Original Document
  • Permalink

Disclaimer

C4 Therapeutics Inc. published this content on 05 June 2023 and is solely responsible for the information contained therein. Distributed by Public, unedited and unaltered, on 05 June 2023 14:50:01 UTC.