Corporate Presentation - August 2021 Listed on Tokyo Stock Exchange (Mothers) Code: 4594
←
→
Page content transcription
If your browser does not render page correctly, please read the page content below
Disclaimer The material that follows is a presentation of general background information about BrightPath Biotherapeutics Co., Ltd. (“BrightPath”) as of the date of this presentation. This material prepared solely for information purpose and is not to be construed as a solicitation or an offer to buy or sell any securities and should not be treated as giving investment advice to recipient. This material and any material distributed in connection with this presentation may contains forward-looking statements, beliefs or opinions regarding BrightPath’s future business, future position and result of operations, including estimates, forecasts, targets and plans for BrightPath. Without limitation, forward-looking statements often include words such as “target”, “plans”, “believes”, “hopes”, “continues”, “expects”, “aims”, “intends”, “ensures”, “will”, “may”, “should”, “would”, “could”, “anticipate”, “estimates”, “projects” or similar expressions or the negative thereof. Such forward-looking statements involve known and unknown risks, uncertainties and other factors which may cause our actual results, performance or achievements to be materially different from any future results, performance or achievements expressed or implied by such forward-looking statements. Such forward-looking statements are based on numerous assumptions regarding our present and future business strategies and the environment in which BrightPath will operate in the future. The important factors that could cause our actual results, performance or achievements to differ materially from those in the forward- looking statements include, among others, risks associated with product discovery and development, uncertainties related to the outcome of clinical trials, slower than expected rates of patient recruitment, unforeseen safety issues resulting from the administration of our products in patients, uncertainties related to product manufacturing, the lack of market acceptance of our products, our inability to manage growth, the competitive environment in relation to our business area and markets, our inability to attract and retain suitably qualified personnel, the unenforceability or lack of protection of our patents and proprietary rights, our relationships with affiliated entities, changes and developments in technology which may render our products obsolete, and other factors. These factors include, without limitation, those discussed in our public reports filed with the Tokyo Stock Exchange and the Financial Services Agency of Japan. Although BrightPath believes that the expectations and assumptions reflected in the forward-looking statements are reasonably based on information currently available to BrightPath's management, certain forward-looking statements are based upon assumptions of future events which may not prove to be accurate. BrightPath does not undertake to update any of the forward-looking statements contained in this presentation or any other forrard-looking statements it may make, except as required by law or stack exchange rule. No representation or warranty, either express or implied, by BrightPath is made as to the accuracy, fairness, or completeness of the information presented herein and no reliance should be placed on the accuracy, fairness, or completeness of such information. Various factors may cause actual results to differ materially from those described in this material. The presentation contains information on drugs currently under development. Nothing contained herein should be considered any solicitation, promotion, advertisement, or medical advice. The information contained in this material may be changed without prior notice. In addition, BrightPath may alter, modify or otherwise change in any manner the contents of this presentation, in its own discretion without the obligation to notify anu person of such revision or changes. Neither BrightPath nor its sources or informants assume any responsibility or liability for any loss or damage that may be incurred by those who will rely on the information in this document. This presentation and its contents are proprietary confidential information and may not be reproduced, published or otherwise disseminated in whole or in part without BrightPath’s prior written consent. These materials are not intended for distribution to, or use by, any person or entity in any jurisdiction or country where such distribution or use would be contrary to local law or regulation. In the event of any inconsistency between the statement in the English-language presentation and the statement in the Japanese-language presentation, the statements in the Japanese-language version should prevail. 1
Development Focus Cancer immunotherapeutics as a main growth driver of pharmaceuticals Cancer Immunotherapeutics ($94.7bn) CAGR 20.2% source : EvaluatePharma, July 2020 3
Approved Cancer Immunotherapeutics Increasing numbers of immunotherapeutics have been approved globally, with the revenues of the marketed products forecast to reach $50bn (2024e) Enhancement Immunity Cycle and of activation Approved Immunotherapeutics ( ) : estimated sales for 2024, EvaluatePharma January 2020 Anti-CTLA-4 antibody Yervoy ($2.0bn) 4 Delivery of cancer- Blood vessel specific cells 5 Anti-PD-1 antibody T cell CAR-T Keytruda ($23.3bn) Kymriah ($1.0bn), Opdivo ($10.5bn) Yescarta ($2.0bn) Jemperli (new) Breyanzi(new) 3 Anti-PD-L1 antibody 6 Abecma (new) Tecentriq ($5.3bn ) T cell (2017-) Imfinzi ($2.2bn) Cancer Bavencio (new) antigen (2011-) Cancer cell Removal of inhibitory Induction of 2 Dendritic cell signals Cancer antigen antigen Anti-PD-1 antibody recognition 7 Keytruda ($23.3bn) Dendritic cell Immune Opdivo ($10.5bn) checkpoint Jemperli (new) vaccine Provenge (-) 1 Dead cancer cell Anti-PD-L1 antibody (2009) Tecentriq ($5.3bn ) Imfinzi ($2.2bn) Bavencio (new) Promotion of (2014-) antigen release Adapted from Chen & Mellman, Immunity 2013 Oncolytic virus T-VEC (-) (2015) 4
BrightPath Pipeline Focused on cancer immunotherapies Multiple modalities (vaccine, antibody, cell therapy) Clinical Stage 4 Adoptive cell transfer T cell 5 iPS-NKT (Investigator-initiated) BP2301 (HER2 CAR-T) 3 Enhancement 6 of antigen- T cell presentation Cancer antigen BP1401 Cancer cell TLR9 agonist 2 Dendritic cell Cancer antigen Induction of antigen 7 Blockade of inhibitory recognition Immune signals checkpoint GRN-1201 BP1200 (CD73) Four tumor associated Ag 1 Dead cancer cell BP1210 (TIM-3) BP1101 BP1202 (not disclosed) Personalized NeoAg Promotion of antigen BP1211 (PVR) BP1209 Next generation-NeoAg release BP1206 (HLA-DR) Adapted from Chen & Mellman, Immunity 2013 5
Near-term Events FY2020 FY2021 FY2022 Cancer vaccines GRN-1201 Tumor associated antigens P2 Stage 1 Read-out BP1101 Neoantigen FY2021 4Q ESMO 2021 BP1209 Neoantigen+Delivery 1005P ”A new platform of personalized neoantigen cancer vaccines directed by BP1401 TLR9 agonist checkpoint inhibitor antibodies to improve cancer immunity.” Cells iPS-NKT iPS cell-derived iNKT cells IND IND BP2301 HER2 CAR-T FY2022 1Q Antibodies ESMO 2021 BP1200 CD73 987P “A novel therapeutic antibody BP1210 TIM-3 against CD73 that ameliorates the tumor microenvironment and improves BP1202 (not disclosed) the efficacy of cancer immunotherapy” BP1206 HLA-DR BP1209 PVR 7
GRN-1201 Cancer vaccine comprised of four TAAs induces cytotoxic T cell to eradicate cancer cells Combination with immune checkpoint blockade antibodies increases antitumor effects 4 T cell 5 3 6 T cell Cancer antigen Antigen = Marker of Cancer Vaccine- Vaccine- Cancer cell Cancer cell induced Cancer cell induced T cell Immune checkpoint T cell molecules bind and send a suppressive signal to T cells 2 Dendritic cell Eliminate cancer cells Cancer antigen expressing antigens identical to vaccines 7 Immune checkpoint 1 Dead cancer cell Cancer cell Killing T cell Induction of antigen recognition GRN-1201 Four TAAs Anti-PD-1 mAb 8
GRN-1201 (cont’d) Address unmet medical needs in NSCLC First-line therapy PD-L1 expression ≧50% GRN-1201 added to first-line pembrolizumab therapy to increase responders Anti-PD-1 PD-L1 expression 22% Not effective in 55% ≧50% or not est. # of pts: 71,000 (US+ EU+ JP, 2025) * 52% PD-L1 expression 1-49% 30% Anti-PD-1 + Chemotherapy PD-L1 expression ≧1% or not Not effective in 45% First-line therapy Second-line therapy PD-L1 expression < 1% 48% Chemotherapy Anti-PD-1 Not effective 40% not in 80% eligible * Source: Datamonitor Healthcare® | Informa, 2020, Dietel et al. Lung Cancer 134(2019), BrightPath Biotherapeutics 9
GRN-1201 (cont’d) Ongoing Phase 2 trial of GRN-1201 in addition to pembrolizumab as first- line NSCLC therapy A Pilot, Open-Label, Multi-Center, Multi-Dose Study of GRN-1201 Added to Study title Pembrolizumab in Subjects with Non-Small Cell Lung Cancer with High PD-L1 Expression US clinical trial no. NCT03417882 Investigational GRN-1201: Four HLA-A2*1-restricted peptides product PD-L1 positive non-small-cell lung cancer Target PD-L1 positive (TPS*2 ≥ 50%) Primary endpoint Objective Response Rate Concomitant drug Pembrolizumab (Keytruda) 64 Sample size Simon's Two-Stage Design Method Open-label, multi-center *1 This 55% group consists of 50% of the American or European subjects and 40% of the Japanese subjects. *2 TPS: Tumor Proportion Score (percentage of cells expressing PD-L1 on their surface among all tumor cells) 10
GRN-1201 (cont’d) Preliminary analysis in the initial stage of Phase 2 supports the mechanistic hypothesis of GRN-1201, suggesting that the immune response may contribute to better clinical outcome 180 ELISPOT (IFNγ production 症例A cell count) - Case A Spot numbers/10 PBMC count Peptide A 160 Peptide B 140 Pepride C Change in tumor target lesions (as of May 2020) 120 Peptide D 100 PR SD (best target legion response) 6 80 30 60 40 20 20 Change in tumor target lesion (%) 0 10 0 19 43 (Weeks) ELISPOT (IFNγ production 症例B cell count) - Case B 0 Spot numbers/10 PBMC count 180 160 Peptide A Peptide B -10 140 Pepride C 120 Peptide D 6 -20 100 * 80 60 -30 40 20 n.a. -40 0 0 19 43 (Weeks) -50 ELISPOT (IFNγ production 症例C cell count) - Case C Spot numbers/10 PBMC count 180 -60 160 Peptide A Peptide B 140 Pepride C -70 120 Peptide D 100 0 10 20 30 40 50 60 6 80 Weeks after the initial administration 60 40 20 * Assessed PD after the observation point due to the development of new lesions 0 0 19 43 (Weeks) By Direct ELISPOT (24h) 11
iPS-NKT Novel allogeneic cell therapy using iPS cell-derived invariant NKT cells iNKT cells, unique subset of T cells, are implicated in the regulation of broad immune responses, but their limited frequency had been an obstacle to acquire sufficient numbers of iNKT cells from patients to induce effective antitumor immune responses of iNKT cell-based immunotherapy iPSC technology made it possible to reprogram human iNKT cells to pluripotency and subsequently re- differentiate into functional iNKT cells demonstrating anti-tumor efficacy The iPSC-derived iNKT cells are functionally recovered and available in an unlimited supply from iPSCs Adoptive cellular transfer Indirect iPS-NKT Indirect killing Cancer cell (Phase 1) killing NK cell killing Direct NK receptor T cell Cancer antigen Cancer cell T cell Dendritic cell NKT cell Lipid antigen Regulate pro- Activation Maturation tumor Mφ Dendritic cell Cancer antigen Immune iNKT cells regulate broad checkpoint antitumor immune responses Macrophage Dead cancer cell 12
iPS-NKT (cont’d) A first-in-human investigator-initiated trial in patient with head and neck cancers started in June 2020 A Phase I study of iPS-NKT cell intra-arterial infusion therapy in patients with recurrent or Study Title advanced head and neck cancer (First in human study) Recurrent or advanced head and neck squamous cell carcinoma, refractory or intolerant to Subjects standard treatment To exploratory evaluate safety, efficacy and tolerability of intra-tumor nutrient artery administration Purpose of iPS-NKT cells in patients with recurrent or advanced head and neck cancer after standard therapy, which is difficult to treat radically. Primary outcome Incidence of dose-limiting toxicity (DLT) at each dose Secondary efficacy endpoints Response rate (RECIST ver.1.1) Disease control rate (RECIST ver.1.1) Secondary outcome Secondary safety endpoints Occurrence of adverse events (type, frequency, severity, etc.) Changes in clinical laboratory values ・Peripheral blood concentration transition of iPS-NKT cell Exploratory ・Immune cell fraction outcomes ・Blood cytokine concentration Sample size 4-18 Study design Single site, open label, asymmetric, dose titration Trial site Chiba University Hospital Source: Chiba University Originated and developed by RIKEN and BrightPath has an option to obtain an exclusive license to develop, manufacture and sell and started collaborative research with RIKEN in April 2018 13
iPS-NKT (cont’d) Enhanced functionality of iNKT Cells iPS cell-derived NKT cells promote secretion of anti-tumor IFN-γ and reduce immunosuppressive IL-4 NKT line: iNKT cells before reprogramming into iPSC iPS-NKT: iNKT cells re-diffrentiated from iNKT cell- derived iPSC DC: co-culture with dendritic cells DC + Gal: co-culture with dendritic cells and antigens DC DC NKT DC DC NKT DC DC NKT DC DC NKT (α-GarCer) + only + only + only + only NKT Only: without dendritic cells and antigens Gar Gar Gar Gar Increased In vitro cytotoxicity vs. iNKT cells before reprogramming into iPSC Leukemia Non-small-cell lung Non-small-cell lung Colon cancer Colon cancer Pharyngeal cancer cancer cancer Killing activity (%) Killing activity (%) Yamada et al., Stem Cells. 2016 14
iPS-NKT (cont’d) In vivo Anti-tumor Activity iPS-NKT cells suppress the proliferation of cancer cells Tumor size (Fluorescence Intensity: Total Flux [photons/second]) Source: Yamada et al., Stem Cells. 2016 15
iPS-NKT (cont’d) Established iPS master cell bank as renewable starting cell source and robust cGMP process from iPSC to NKT cells to mass produce homogeneous NKT cells Pre-diagnosis Post-diagnosis Autologous cell therapy Patient Apheresis Production Administration of single dose Diagnosis Allogeneic cell therapy 100+dosage Healthy donor T cell Production Master cell bank Substantial reduction of allogeneic cell 10,000+dosage turnaround time from diagnosis therapy to administration Healthy donor T cell Reprogramming Master cell Production Administration of bank multiple doses Reprogramming Process iPSC iPS cell-derived expansion NKT cell NKT cell-derived iPS cell iPS single-cell clone Master cell bank Reprogramming Re-differentiation & Multiplication into iPS cells NKT cell Cryopreserved stock (for immediate use) 16
iPS-NKT (cont’d) Benchmarking CAR-transfected iPSC-derived effector-cell products for expansion strategy (IND filing year) Hematology CAR Solid tumor CAR Un- Company CD19 BCMA CD38 EGFR GPC3 MICA modified Fate Tx 2018 2020 2021 2022 iPSNK Century Tx 2021 2022 Cytovia Tx 2021 2022 2022 2022 iPSNKT BrightPath 2020 (RIKEN) 17
BP2301 Autologous HER2 CAR-T cell therapy for solid tumors HER2 is a clinically validated target and HER2 CAR-T has third party’s precedent successful clinical outcome in sarcoma* BP2301 demonstrates a stem cell memory-rich phenotype, which is expected to improve the anti-tumor efficacy Flow of CAR-T cell therapy Adoptive cell transfer HER2 CAR-T CAR gene transfer (piggyBac) CAR-T cell T cell Multiplication Kill cancer cells CAR-T cell Cancer cells Antigen Signal transduction Antigen binding site domain Cancer antigen CAR-T cells Cancer cell T cell Blood Drawing Administration * HEROS2 study of HER2 CAR-T in patients with sarcoma conducted by Baylor College of Medicine showed one complete response and a response rate of 18% (AACR2019) although most clinical studies of CAR-T for solid tumors have not been able to demonstrate CR. 18
BP2301 (cont’d) piggyBac transposon-based CAR gene transfer enables CAR-T cells to maintain stem cell memory-like phenotype with self-renewing, long-term persisting ability and durable antitumor effects T cell differentiation and phenotypes Persistence of memory-rich CAR-T cells Cell death Self-renewal Long-term persistence Effector function SCM: ステム・セル・メモリー CM: セントラル・メモリー SCM: Stem Cell Memory EM: エフェクター・メモリー CM: Central Memory EFF: エフェクター EM: Effector Memory EFF: Effector Memory-rich CAR-T Effector-rich CAR-T 19
BP2301 (cont’d) BP2301 cells exhibits dominant of stem cell memory phenotype without early T cell exhaustion TEFF TSCM PD-1 72.5 % 0.3 % CD45RA PD-1 TEM TCM CCR7 CD3 SCM: Stem Cell Memory CM: Central Memory EM: Effector Memory EFF: Effector Source: Shinshu University, BrightPath 20
BP2301 (cont’d) Memory-rich BP2301 exhibits long-term functional persistence and exhaustion resistance BP2301 exhibits robust serial killing Tumor: Osteosarcoma cells(U2-OS) E:T =1:1 Tumor alone Round 1 +HER2 CAR-T cells Tumor alone Round 2 +HER2 CAR-T cells Tumor alone Round 3 +HER2 CAR-T cells Source: Shinshu University, BrightPath 21
BP2301 (cont’d) BP2301 mediates robust antitumor function in vivo Vehicle CD19.CAR HER2.CAR Vehicle Tumor size (Fluorescence Intensity: Total Flux Day 6 Day14 [photons/second]) Day21 CD19.CAR Day28 Day35 HER2.CAR Day42 Source: Shinshu University, BrightPath Number of days from cancer cell transplant 22
BP2301 (cont’d) BP2301 demonstrates long-term functional persistence in vivo BP2301 sustains antitumor function in re-challenge tumor mouse model 1 x 107 Re-challenge tumor Primary tumor Vehicle CD19.CAR HER2.CAR Rechallenge Day0 Rechallenge Day7 Rechallenge Day14 Source: Shinshu University, BrightPath 23
BP1101 A fully-personalized neoantigen vaccine, induces immunity against antigens derived from cancer-specific genetic mutations (neoantigens), which are completely unique to every patient T cell migration Blood vessel T cell T cells infiltrate cancer tissue Lymph node T cells recognize cancer antigens presented on the dendritic cells and induce multiplication and T cells recognize cancer activation antigens on the cancer cell T cell surface Cancer antigen Cancer cell Cancer Cancer antigens phagocytized by Dendritic cell dendritic cells appear on the cell surface T cells kill cancer Immune Kill cancer cells, release cells checkpoint 50 Cutaneous cancer antigens squamous-cell Dead cancer cell Merkel-cell Non-colorectal 40 (MMRd) Melanoma Objective Response Rate (%) Colorectal (MMRd) 30 Anal Tumor mutation burden (neoantigen burden) Renal-cell Cervical 20 Hepatocellular Urothelial Mesothelioma NSCLC(squamous) NSCLC(non-squamous) Head and neck correlates with the clinical response to anti- checkpoint therapy, suggesting that neoantigens Response rate Endometrial Small-cell lung Sarcoma Ovarian 10 Esophagogastric Glioblastoma Prostate Uveal Adrenocortical Breast Pancreatic Germ-cell Colorectal (MMRp) are cancer immunity targets 0 1 10 20 30 40 50 Median No. of Coding Somatic Mutations per MB Cancer somatic mutation burden (neoantigen burden) Yarchoan et al, NEJM (2017) 24
BP1209 Antibody-vaccine conjugate with improved vaccine delivery to dendritic cells and immune induction Anti-dendritic cell marker antibody Neoantigen peptide vaccine (BP1101) ESMO 2021 1005P ”A new platform of personalized neoantigen cancer vaccines directed by checkpoint inhibitor antibodies to improve cancer immunity.” Source: BrightPath 25
BP1209 (cont’d) Improves anti-tumor immunity by increasing cDC1’s uptake of peptide vaccine and migration to lymph node Dendritic cells’ uptake of vaccine Immune induction in a mouse model and transition to lymph nodes (B6 +MC38 neoantigen) 1.2 cDC1 cDC2 1500 Amount of vaccine transferred to the lymph nodes (relative) IFNg+/ CD8+ cell (%) 0.8 1000 0.4 500 cDC1 cDC2 cDC1 cDC2 0.0 0 Adj Adj+peptide Adj+peptide+CD40Ab e h) h) Antibody-binding h) h) Antigen peptide lin 2 Eq 2 Eq - - - ig ig ig ig antigen peptide - 2 Eq 2 Eq Sa (h (h (FAM-labeled) (h (h c c 40 40 -F nF Non-binding D D gk - - 2 Eq 2 Eq - k- C +C antigen peptide Anti-DC marker mAb - - 200 dp g + dp (μg/head) c Fc A -F A -n gk gk PolyICLC dp Anti-DC marker mAb - 200 - 200 - 8 dp A (μg/head) (ug/head) A PolyICLC 8 - (μg/head) Source: BrightPath 26
BP1209 (cont’d) Tumor-bearing mouse study demonstrated increased anti-tumor immunity Vaccine (OVA) Day0 3 10 33 E.G7 (EL4/OVA) OVA expressed lymph Tumor cell transplant 2800 2400 2000 saline Tumor size (mm3) 1600 adjuvant only 1200 Administration 800 Administration vaccine ← BP1101 type 400 vaccine-binding 0 dendritic cell marker ← BP1209 type 0 7 14 21 antibody Days after cancer cell transplant Source: BrightPath 27
BP1401 Lipid nanoparticle of Type-A TLR9 agonist Suppress tumor growth by changing tumor immune-microenvironment LNP enables intravenous administration expecting safely brought systemic effects, which is differentiated from intratumoral injection-based predecessors in a clinical stage Promote cross presentation of Class I antigen Blood vessel T cell Enhancement of antigen- presentation Lymph node Recruit T cells to BP1401 tumor site TLR9 agonist T cell Cancer antigen Cancer cell Cancer Cancer antigen Immune checkpoint Macrophage Dead cancer cell Myeloid cell, etc. 28
BP1401(cont’d) BP1401 promotes T-cells’ infiltration to and eradication of tumors Intratumoral administration of BP1401 Predecessors under clinical development are all intratumorally administrated 8 6 40 4000 ** ** N o n -tre a te d ** R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n 6 30 4 Is o ty p e -A b Non-treated D35LNP0.5 3000 4 * 20 T u m o r s iz e (m m ) 3 C D 8 a -A b 2 2 10 D 35LNP+CD8Ab 0 0 0 2000 D 3 5 L N P 0 .5 % 10 2 .5 2 .0 50 ** ** ** R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n Cxcl11 Cxcr3 8 2 .0 40 1 .5 1000 6 1 .5 30 * s 4 1 .0 1 .0 20 0 .5 ** ** 2 0 .5 10 0 ** ** ** 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 0 0 .0 0 .0 0 Intravenous administration of BP1401 6 5 6 Ifna6 Ifnb1 Ifng R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n N o n -tre a te d 4 4000 4 4 D 3 5 o n ly 3 Interferons ** ** ** ** T u m o r s iz e (m m ) D 3 5 L N P 3 .0 % 3 2 2 2 3000 D 3 5 L N P 0 .5 % 1 0 0 0 2000 6 3 3 4 * i.v. Cxcl9 Cxcl10 Cxcl11 Cxcr3 R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n R e la tiv e E x p r e s s io n 3 4 2 2 1000 Chemokines ** 2 2 ** 1 1 ** ** 1 0 0 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 0 0 0 0 Intravenous administration of BP1401 in combo with anti-PD-1 mAb 800 Is o ty p e A b g tu m o r v o lu m e (m m ) D 3 5 L N P 0 .5 % 3 600 A n ti P D - 1 A b 800 600 * 200 150 400 C o m b in a tio n 600 * 150 400 100 c e lls /m g c e lls /m g c e lls /m g c e lls /m g 400 100 200 200 50 * 200 50 ** 0 0 0 0 0 0 5 10 15 20 day Lipid nanoparticles of Type-A CpG D35 suppress tumor growth by changing tumor immune-microenvironment and activate CD8 T cells in mice Journal of Controlled Release 313 (2019) 106-119 29
BP1200 A novel antibody targeting CD73 Adenosine generation in tumors causes T cell exhaustion and dysfunction CD73 is highly expressed in various tumors and produce adenosine, leading to poor prognosis Dendritic T cell cell CD8+ Production of adenosine in the tumor T cell environment CD4+ T cell T cell Cancer antigen Cancer cell Tumor Dendritic cell Cancer antigen Immune Suppress tumor cytotoxicity Promote differentiation into suppressor T cells checkpoint Dead cancer cell 30
BP1200 (cont’d) BP1200 blocks the function of adenosine-generating enzyme (CD73) Superior to benchmarks in clinical stages BP1200 promotes T cell division and cytokine secretion, which leads to enhance anti-tumor immunity • がん免疫を亢進し、抗腫瘍効果を発揮することが期待される Induction of secretion of Inhibition of adenosine-generating enzyme Promotion of T cell division cytokine (IFN-γ) ● BP1200 〇 Control antibody ESMO 2021 987P “A novel therapeutic antibody against CD73 that ameliorates the tumor Source: BrightPath Biotherapeutics microenvironment and improves the efficacy of cancer immunotherapy” 31
BP1210 Targeting TIM-3 as a novel immune checkpoint following PD-1/L1 BP1210 blocks TIM-3 and inhibits exhaustion of T cell to enhance anti-tumor immunity BrightPath’s target immune checkpoints T cell exhaustion is associated with increased TIM-3 expression T cell T cell Dendritic cell Naive T cell Anti- BP1210 CTLA-4 BP1210 Stem-like memory PD-1lo T cell Exhausted (Exhausted) PD-1med Anti- LAG3+ PD-1 TIM-3+ CD39+ Hyper-exhausted (Hyper-exhausted) Cancer cell PD-1hi Antigen presenting cell LAG3+ TIM-3+ CD39+ Red: Clinically validated, regulatory-approved target Bold: BrightPath’s target Granzyme Cytotoxic T cell 32
BP1210 (cont’d) BP1210 promotes T cell proliferation and cytokine secretion Superior to benchmarks in clinical stages SEB stimulation of PBMC (in combination with PD-1 antibody) T cell count IL-2 production IFN-γ production (day6) (day 6) (day 2) 2.0×10 5 6000 15000 (pg/ml) IFN-γ (pg/ml) cell count 1.5×10 5 (pg/ml) IL-2 (pg/ml) 4000 10000 TT細胞数 1.0×10 5 IFN-γ濃度 IL2濃度 2000 5000 5.0×10 4 0 0 0 体 体 体 抗 抗 s社 抗 s社 s社 0 0 0 ル ル ル 21 21 21 it i it i it i ー ー ー P1 P1 P1 ar ar ar ロ ロ ロ B B B ov ov ov ト ト ト ン ン ン N N N コ コ コ 33
BP1210 (cont’d) BP1210 strongly inhibits the binding of ligand PtdSer to TIM-3 • Superior to benchmarks in clinical stages BP1210 promotes T cell proliferation and cytokine secretion • Superior to benchmarks in clinical stages Binding inhibition of PtdSer SEB stimulation of PBMC (in combination with PD-1 antibody) T cell count IL-2 production IFN-γ production (day6) (day 6) (day 2) 2.0×10 5 6000 15000 (pg/ml) IFN-γ (pg/ml) cell count 1.5×10 5 (pg/ml) IL-2 (pg/ml) 4000 10000 TT細胞数 1.0×10 5 IFN-γ濃度 IL2濃度 2000 5000 5.0×10 4 0 0 0 体 体 体 抗 抗 s社 抗 s社 s社 0 0 0 ル ル ル 21 21 21 it i it i it i ー ー ー P1 P1 P1 ar ar ar ロ ロ ロ B B B ov ov ov ト ト ト ン ン ン N N N コ コ コ Source: BrightPath Biotherapeutics 34
BP1211 Anti-PVR antibody to inhibit binding TIGIT to PVR (CD155) and prevent T cell exhaustion BrightPath targeting immunocheckpoints Inhibition of TIGIT binding BP1211 BP1210 (PVR) antibody concentration (log, μg/mL) Red: Clinically validated, regulatory-approved target Bold: BrightPath target Source: BrightPath 35
BP1206 Anti-HLA-DR antibody targeting HLA-DR on blood cancer cells Antitumor effects in vitro dependent on HLA-DR expression 100 60,000 Cancer cell killing 80 HLA-DR expression (MFI) Viable Cancer Cells (%) 40,000 60 40 20,000 20 0 0 MUTZ-5 (ALL) L428 (HL) GRANTA-519 (DLBCL) JJN-3 (MM) Rmos (BL) OCI-LY-19 (DLBCL) Pfeiffer (DLBCL) Daudi (BL) Raji (BL) Mino (MCL) KG-1 (AML) KMS-11 (MM) KMS-26 (MM) NU-DHL-1 (DLBCL) Nalm-6 (ALL) Kasumi-1 (AML) BP1206 induces HMGB1(DAMP) release from dying cancer cells Control BP1206 Source: BrightPath HMGB1 in tumor 36
BP1206: (cont’d) BP1206 exhibits robust antitumor effects in vivo NOG mouse Day -12 0 42 Hodgkin’s lymphoma cell L428 s.c. engraftment (n=10) 10 mg/kg i.p. Q4D x 4 times 1200 1400 PBS Isptype (10mg/kg) 3 complete BP1206 (1mg/kg) 1000 eliminations 1200 BP1206 (5mg/kg) BP1206 (10mg/kg) BP1206 10mg/kg tu m o r w e ig h t ( m g ) 1000 800 Mouse # 47 Tumor size( mm3) 800 600 BP1206 10mg/kg BP1206 Mouse # 52 600 400 400 BP1206 10mg/kg Mouse # 53 200 200 0 0 Is o ty p e 1 m g /k g 5 m g /k g 1 0 m g /k g -7 0 7 14 21 28 35 42 Days post tumor engraftment Source: BrightPath 37
Company Overview
Company Profile Company BrightPath Biotherapeutics Co., Ltd. (Tokyo Stock Exchange Mothers: 4594) name Headquarters: 2-2-4 Kojimachi, Chiyoda-ku, Tokyo Location Kawasaki Research Laboratories: 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa Cell Technology Laboratories: 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa Established May 8, 2003 Business Development of novel cancer immunotherapy description Stated capital 6,459 million yen (as of the end of March 2021) Number of 44 (as of the end of March 2021) employees Chief Executive Officer Kenichi Nagai Chief Science Officer Norihiro Nakamura Director (part-time) Akira Yamada (Professor at Kurume University) Management Director (outside, independent ) Hirotaka Takeuchi (Professor at Harvard Business School) team Auditor (outside) Tsutomu Kishino Auditor (outside, independent) Taketoshi Abe Auditor (outside) Yoshiyasu Yamaguchi (Partner, TMI Associates) 39
Business Locations Headquarters: Kojimachi Central Building 7F, 2-2-4 Kojimachi, Chiyoda-ku, Tokyo, Japan Kawasaki Research Laboratories Cell Technology Laboratories Life Innovation Center, 3-25-22 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, Japan Haneda Airport Tonomachi International Strategic Zone King SkyFront 神奈川県川崎市川崎区殿町3丁目25 ライフイノベーションセンター412 電話 044-440-3310 FAX 044-440-3311 40
We pioneer immunotherapy, to enable a world where cancer patients can defeat cancer on their own.
You can also read