Presented at the SITC 38th Annual Meeting, 3-5 November 2023, San Diego, CA, USA, and online

Abstract 656

ELI-002 Immunotherapy Induces Broad Polyfunctional T cell Responses in Subjects with High

Relapse Risk KRAS Mutated Pancreatic Ductal Adenocarcinoma and Colorectal Cancer

James R. Perry1, Lochana M. Seenappa1, Haley VanWyk1, Amy M. Tavares1, Thian Kheoh1, Esther Welkowsky1, Christopher M. Haqq1, Peter C. DeMuth1,

and Lisa K. McNeil1

1 Elicio Therapeutics, Inc. 451 D St., Ste 501, Boston, MA 02210

Why Target mutated KRAS with Therapeutic Vaccination?

mKRAS T Cell Responses Correlate with Reduction in Risk of Relapse or Death13

Expansion of mKRAS-specific T cells by ELI-002 2P Immunization

1 Mutant KRAS Drives 25% of Solid Human Cancers

Prevalent among numerous tumor types1-2

Overall poor clinical prognosis3

Limited therapeutic options

2 Mutant KRAS is a Promising Tumor Antigen

Truncal: mutations occur early, expressed uniformly in all tumor cells

Driver: mKRAS signaling is required for tumor growth and survival

Highly prevalent: involved in ~25% of solid tumors1-2

Public neoantigen: not centrally tolerized, cognate TCRs present in naïve repertoire4-5

Promiscuous HLA presentation: potential off-the-shelf use in diverse patient population6-8

Proven Clinical MOA: mKRAS-specific T cells known to mediate anti- tumor efficacy4-5

Multi-targetingpotential: recognition of clonal and subclonal mKRAS variants to prevent escape9

KRAS mutant

NRAS mutant

5%

52%

93%

Colorectal Cancer (CRC)

Pancreatic Ductal

US Incidence: 151k

Adenocarcinoma

(PDAC)

US Incidence: ~56k

Strength of T Cell Response

86% Reduced Risk of Relapse or Death

Tumor Biomarker Response

(%)Survivalfree-Relapse

ResponseBiomarkerOverall

Baseline)of(%

300

100

Median RFS: not reached

P = 0.0017

200

75

✱✱

50

100

25

Median RFS: 3.91 months

Best

0

-100

Months 0

0

3

6

9

12

15

18

≥ Median

< Median

T Cell Response

≥ Median T Cell Response (n = 12)

P = 0.0134

Best Overall Tumor

Clearance

< Median T Cell Response (n = 10)

HR: 0.138 (0.031 - 0.610)

Biomarker Response

Reduction

Non-Responder

450

Fold-change

Ex Vivo Fluorospot

Ex Vivo ICS

and/orγIFN GrB SFC

1300

among

T cells

4

0.1 mg

50

200

Foldchange frombaseline

PBMC

2

0.5 mg

1.0

30

150

+

2.5 mg

Median:

1x10/

Cytokine%

CD8

10

13x

6

100

+

CD4or

0.5

5.0 mg

+

5

50

10.0 mg

0

Max

0

Max

0.0

Max

Baseline

Baseline

Baseline

Response

Response

Response

  • 87% of Patients generated direct ex vivo detectable mKRAS-specific T cell responses following ELI-0022P Immunization, with 100% Responders at the highest dose levels (5.0 and 10.0 mg)

Ex Vivo Responders

CD4 / CD8 Response

Specificity

Specificity

13

13

10

10

20

35

50

25

5

87

38

30

65

Responders

CD4 + CD8 T cells

7 antigens

G12R Responses

Non-responders

CD8 T cells

5-6 antigens

G12D Responses

ELI-002 2P: Advancing Innovation for mKRAS Cancer Vaccines

1 Technological Innovation: Amphiphile Lymph Node Targeting Platform10-11

2 Clinical Innovation: Treatment in High Relapse-Risk Adjuvant Setting

Smart trafficking to the lymph nodes after subcutaneous dosing generates

Targeting surgically debulked tumors enables T cells to address minimal residual

immune responses with increased magnitude, function, and durability.

disease to potentially eliminate remaining tumor cells and protect against

Takes advantage of potent lymph node immune mechanisms, including activation

recurrence.

Activating the immune system before loss of HLA expression in the tumor

of innate and adaptive cells, antigen-spreading, and improved tumor T cell

trafficking / infiltration.

microenvironment in a chemotherapy-free window of opportunity.

Mutant KRAS peptides provide a validated antigen for application of the

Other oncology vaccines have typically been used in later lines of therapy for

Amphiphile platform.

advanced disease, after onset of tumor immune resistance.

Lymph node delivery of potent adjuvants minimizes systemic exposure to

In the adjuvant setting, tumor biomarkers (ctDNA, serum tumor antigen) are

improve safety.

early predictors of disease control or recurrence.

Amphiphile mKRAS Long

12

ELI-002 2P

Peptide Antigens

O

1

Amph-mKRAS G12D

O

2

Amph-mKRAS G12R

Albumin Binding Lipid

PEG Linker

G12D or G12R Peptide

Amphiphile TLR-9 Agonistic

O

NH

DNA Adjuvant

O

NH

Amph-CpG-7909

Albumin Binding Lipid

CpG-7909 DNA

Inclusion of 18-mer G12D and G12R mKRAS peptides allows for delivery of diverse HLA I and II - restricted epitopes for presentation on varied patient HLA molecules.

Amphiphile (Amph)-modificationof peptides promotes binding to endogenous albumin at the injection site to promote collection in lymphatic vessels for lymph node delivery, and prevents peptide uptake into local capillaries avoiding delivery to irrelevant or tolerogenic sites.

Amph-CpG-7909provides potent immune activation via TLR-9 stimulation of lymph node-resident professional antigen presenting dendritic and other key immune cells.

Tissue Injection Site

Lymph Node

Amphiphiles

Endogenous Albumin

Albumin-bound Amphiphiles

Albumin-bound Amphiphiles

Antigen Presenting Cell

T Cell

1 Subcutaneous

2

Albumin

3

Lymph node

4

Delivery to

injection

binding

targeting

immune cells

Conventional vaccine components (e.g. peptide antigens and molecular adjuvants) are rapidly absorbed into blood capillaries after administration leading to poor delivery to lymph nodes where protective immune responses are orchestrated.

Amph-modification promotes albumin binding to reprogram vaccines for enhanced lymph node delivery resulting in coordinated transport of antigen and adjuvant to immune cells. Improved uptake by Antigen Presenting Cells results in enhanced antigen-presentation and co-stimulation to cognate T cells.

Restricted delivery to lymph nodes minimizes systemic exposure to avoid toxic effects of potent adjuvants.

Patient #23: mKRAS-specific T cell Profile - Ex Vivo Fluorospot

250

G12R

1000

G12V

1000

WT KRAS

and/orIFNγ GrB SFC 1x10/

1x10/

and/orIFNγ GrB SFC

1x10/

200

and/orIFNγ GrB SFC

800

800

PBMC

150

PBMC

600

G12A

PBMC

600

6

6

G12S

6

100

400

400

G13D

50

200

200

0

0

0

Week

B

9

Week

B

9

Week

B

9

Week 9

Week 9

IFNγ+

IFNγ+

Elevated T cell responses to G12R

and G12D by Week 9 after prime

GrB+

GrB+

immunization series

99

IFNγ+ GrB+

99

IFNγ+ GrB+

No responses were observed to

99

the WT antigen

Patient #23: mKRAS-specific T cell Profile - Ex Vivo ICS Assay

8

G12R

CD8 T Cells: Week 9

G12R-Specific CD8: Week 9

cells

+

6

G12D

IFNγ+

mKRAS-specific, Cytokine+

Cytokine%

T

2

10

48

amongCD8

2+

12

12

+

4

42

TNFα+

24

24

16

41

7

3+

0

Naïve

Effector Memory

Week

B

9

Central Memory

TEMRA

3

G12R

CD4 T Cells: Week 9

G12R-Specific CD4: Week 9

+

Tcells

G12D

IFNγ+

mKRAS-specific, Cytokine+

Cytokine%

CD4among

2

38

2+

28

52

31

+

15

IL2+

20

4 14

44

α

+

1

TNF

51

0

3

3+

Week

B

9

Patient #23 mKRAS-specific Memory T cell Profile - IVS

CD4 T cells

2-4 antigens

G12R + G12D Responses

Responders

NR

1 antigen

Neither

(n = 20)

(n = 3)

7

G12R

Amph-

ex vivo T cell

Average

CpG Dose

response

fold-

Number of mKRAS

antigens/patient

6

G12D

Level

(n, %)

change

5

G12V

0.1 mg

2/3 (67%)

30

4

G12C

0.5 mg

5/6 (83%)

82

3

G12A

2.5 mg

4/5 (80%)

113

2

G12S

5.0 mg

5/5 (100%)

19

1

G13D

10.0 mg

4/4 (100%)

26

0

No Response

Total

20/23 (87%)

56

Cohort 1 1 2 2 2 2 2 3 3 3 3 4 4 4 4 4 5 5 5 5 1 2 3

  • CD4 and CD8 T cell responses were observed, with 50% generating mixed CD4 + CD8 responses
  • mKRAS-specific T cells were polyfunctional (IFNγ, TNFα, IL-2), specific to both immunizing and non-immunizing mKRAS antigens

Increased mKRAS-specific Memory T cells Assessed by IVS

baselinefromchangeFold

100

Fold-change

IVS Fluorospot

cellsT

40

IVS ICS

4500

0.1 mg

150

SFCαTNFand/orγIFN

1x10/

Cytokine%

CD8among

60

50

PBMC

500

+

+

20

0.5 mg

20

400

8

2.5 mg

6

CD4 or

25

10

15

300

+

6

5.0 mg

10

200

4

10.0 mg

5

100

2

0

Max

0

Max

0

Max

Baseline

Baseline

Baseline

Response

Response

Response

  • 100% of patients induce mKRAS-specific memory T cell responses after in vitro stimulation

Increased Polyfunctionality of mKRAS-specific T cells after ELI-002 2P Immunization

Patient #23

T Cell Activation: Week 9

Cytolysis: Week 9

Proliferation: Week 9

  • Polyfunctional T cell Profile:
  • Increased activation,

cytolysis, and

CD8

0.71

CD4

7.81

CD4

5.02

proliferation after

vaccination

CD137

GrB

Ki67

ELI-002 2P Immunization Elicits Durable mKRAS-specific Immune Responses

2000

Patient 16

ELI-002 2P vaccination generates long-lasting mKRAS-

AMPLIFY 201: Trial Design12

Prior Therapy

Screening Period

Amph-Peptides 2P 1.4 mg + 0.1, 0.5, 2.5, 5 or 10 mg Amph-CpG-7909

Locoregional

mKRAS+

Prime

No Dosing

Booster

Follow-up

G12R+ or G12D+

Therapy:

Immunization

Period

Immunization

Period

Surgery

NED

+

Imaging Negative

Week S/B 0 1 2 3 4 5

6 7 8 9 17

20 21 22 23 24

25

105

Neoadjuvant /

MRD+

Dose

Adjuvant

ctDNA+ or

ctDNA

Chemotherapy

serum biomarker+

Serum biomarkers

PBMC

Patients

Baseline Characteristics: 20 Pancreatic (PDAC), 5 Colorectal (CRC) were evaluated for safety as of data cutoff: April 25, 2023

Safety

Safety:

No TEAEs ≥ Grade 3, no Dose Limiting Toxicities, no Cytokine Release Syndrome observed across all dose levels;

44% had Grade 1-2 TEAEs: e.g. injection site reaction, fatigue, headache, nausea12

AMPLIFY 201: Immunogenicity Methods

Immunogenicity of ELI-0022P was assessed using longitudinally collected peripheral blood from 23 evaluable patients to assess specificity, polyfunctionality,

antigen breadth, and phenotype of mKRAS-specific T cells.

2500

SFC

PBMC

2000

TNFα

1500

6

IFNγand/or / 1x10

1000

500

0

Week

B

9

4

+

cells

3

% Cytokine

T

+

among CD8

2

1

0

Week

B

9

G12R G12D

G12R G12D

% Cytokine Secretion in

SFC

1000

WT KRAS

Fluorospot: Week 9

PBMC

800

IFNγ+

TNFα

600

6

27

27

IFNγ+ TNFα+

and/orIFNγ

1x10/

46

200

TNFα+

400

0

Week

B

9

15

G12R

cells

+

G12D

After IVS stimulation, greatly

Cytokine%

CD4among

10

increased T cell responses to G12R

T

and G12D at Week 9 post prime

+

immunization series

5

No responses were detected for the

WT antigen

0

Week

B

9

SFC

1500

Patient 18

specific T cell responses

PBMC

1000

Patient 20

600

100% (4/4) of evaluable patients maintain elevated T

GrB

6

and/or

1x10

450

Patient 11

cell responses above baseline post-boost immunization

300

IFNγ

/

An increased post-boost T cell response was observed

150

in 75% (3/4) of evaluable patients compared to pre-

0

5

10

15

20

25

boost T cell levels

0

Weeks post-vaccination

T Cell Response

86% Reduced Risk of Relapse or Death

MESSAGES

MOA Correlated to:

Tumor Biomarker Response

Lymph node-targeted Therapeutic mKRAS-specific Cancer Vaccine ELI-002 2P:

Direct ex vivo mKRAS-specific T cell responses observed in 87% of patients

and IVS responses were observed in 100% of patients

HOME

50% of patients generated both CD4 and CD8 T cell responses

T cells exhibited robust functional quality: activation, cytokine production,

PBMCs from each patient were individually stimulated with overlapping peptides for each of the seven mKRAS antigens (G12R, G12D, G12V, G12C, G12A,

G12S and G13D) and the WT antigen, for evaluation of mKRAS-specific T cell responses using both direct ex vivo and in vitro stimulated assays.

T cell responses and polyfunctionality were determined by a direct ex vivo IFNγ/Granzyme B (GrB) Fluorospot and a 10-dayin vitro stimulated (IVS)

IFNγ/TNFα Fluorospot assay, where a positive immune response was defined as >2-fold over baseline and at least 50 SFC per million PBMCs.

Polyfunctionality and phenotype of patient T cells were further characterized using an ex vivo and IVS intracellular cytokine staining (ICS) assay, where

responder populations were defined as >2-fold over baseline and a frequency of at least 0.1% Cytokine+. The ICS assay included markers for CD3, CD4, CD8,

Memory (CCR7, CD45RA, CD45RO), cytokines (IFNγ, TNFα, IL2), cytolysis (GrB, Perforin, CD107a), activation markers (CD69, CD137, CD154), and proliferation

(Ki67).

References

1.

Bianken A, et al. Nature. 2012; 491(7424): 399-405

5.

Tran E, et al. NEJM. 2016; 375(23): 2255-2262

9.

Awad MM, et al. Cancer Cell. 2022; 40(9): 1010-

2.

Prior IA, et al. Cancer Research. 2012; 72(10):

6.

Bear AS, et al. Nat. Commun. 2021; 12(1): s41467-

1026

2457-2467

021-24562-2

10.

Liu H, et al. Nature. 2014; 507: 519-522

3.

Siegel RL, et al. Cancer J. Clin. 2021; 71(1): 7-33

7.

Carbone DP, et al. J Clin Oncol. 2005; 23(22): 5099-

11.

Moynihan KD, et al. Nature Medicine. 2016;

4.

Leidner R, et al. NEJM. 2022; 386(22): 2112-2119

5107

22(12): 1402-1410

8.

Palmer CD, et al. Br. J. Cancer 2020; 122(7): 971-977

12.

O'Reilly EM, et al. J Clin Oncol. 2023; 41(16): 2528

13.

Wainberg Z, et al. 2023 AACR Special Pancreatic.

Acknowledgements

2023

We are grateful to the patients who participated in the study, their families, and the investigators and staff at the participating institutions.

cytolytic capacity, proliferation, memory phenotype

100% (4/4) of patients evaluable for durability maintained elevated T cell

TAKE

responses above baseline

Phase 1, randomized Phase 2 Study of ELI-002 7P (NCT05726864) in PDAC patients: targeting G12D, R, V, C, A, S, G13D

Attachments

  • Original Link
  • Original Document
  • Permalink

Disclaimer

Elicio Therapeutics Inc. published this content on 03 November 2023 and is solely responsible for the information contained therein. Distributed by Public, unedited and unaltered, on 10 November 2023 14:37:53 UTC.