Alpine Immune Sciences, Inc. in Various Jurisdictions

Total Page:16

File Type:pdf, Size:1020Kb

Alpine Immune Sciences, Inc. in Various Jurisdictions Leading the Next Evolution of Immunotherapies Jefferies 2019 Healthcare Conference June 2019 1 Forward Looking Statements This presentation contains forward-looking statements within the meaning of Section 27A of the Securities Act of 1933, Section 21E of the Securities Exchange Act of 1934 and the Private Securities Litigation Reform Act of 1995. These forward- looking statements are not based on historical fact and include statements regarding Alpine’s platform technology, potential therapies, potential milestone and royalty payments, future development plans, clinical and regulatory objectives and the timing thereof, expectations regarding the sufficiency of cash to fund operations into 2020, expectations regarding the plans of its collaborator, expectations of future collaborations, and expectations regarding the potential efficacy and commercial potential of Alpine’s and its collaborator’s product candidates. Forward-looking statements generally include statements that are predictive in nature and depend upon or refer to future events or conditions, and include words such as “may”, “will”, “should”, “would”, “expect”, “plan”, “intend”, and other similar expressions among others. These forward-looking statements are based on current assumptions involving risks, uncertainties, and other factors that may cause actual results, events, or developments to be materially different from those expressed or implied by such forward-looking statements. These risks and uncertainties, many of which are beyond our control, include, but are not limited to: Alpine’s discovery-stage and pre-clinical programs may not advance into the clinic or result in approved products on a timely or cost-effective basis or at all; Alpine may not achieve additional milestone payments pursuant to its collaborations; the impact of competition; adverse conditions in the general domestic and global economic markets; as well as the other risks identified in our filings with the Securities and Exchange Commission. These forward-looking statements speak only as of the date hereof, Alpine undertakes no obligation to update forward-looking statements, and readers are cautioned not to place undue reliance on such forward-looking statements. “Variant Immunoglobulin Domain”, “vIgD”, “Transmembrane Immunomodulatory Protein”, “TIP”, “Secreted Immunomodulatory Protein”, and “SIP” are registered trademarks or trademarks of Alpine Immune Sciences, Inc. in various jurisdictions. All rights reserved. 2 Leading the Next Evolution of Immunotherapies Seeking True Remissions in Inflammatory Diseases and Oncology ALPN-101 ALPN-202 Platform Technology First-in-class dual Dual PD-L1/CTLA-4 CD28/ICOS inhibitor for antagonist and CD28 agonist Strategic Partnerships inflammatory diseases for oncology Licensing Opportunities Enrolling Phase 1 IND-Enabling (Healthy Volunteers) (Submission Target 4Q19) 3 ALPN Pipeline 2Q19 PROGRAM DISCOVERY IND-ENABLING PHASE 1 COLLABORATOR Inflammatory Diseases ALPN-101 Dual CD28/ICOS Antagonist Healthy Volunteer Study Acute Graft-Versus-Host Disease Psoriatic Arthritis Immuno-Oncology ALPN-202 PD-L1/CTLA-4 Antagonist & CD28 Agonist Advanced Malignancies Engineered Cellular Therapies Secreted and Transmembrane Immunomodulatory Undisclosed Proteins (SIPs & TIPs) (Targets Undisclosed) 4 ALPN-101 Autoimmune/Inflammatory Diseases: Dual ICOS/CD28 Antagonist 5 Current Therapies Do Not Inhibition of Both CD28 and ICOS Induce Remission in Patients Pathways May be Required to Impact Targeting CD28 Alone is Insufficient… Many Serious Inflammatory Diseases CD28 blockade (abatacept) has been only modestly or not effective in: - Lupus (Arthritis Rheum 62:3077, 2010) During activation, many effector T cells lose CD28 dependence - Sjögren’s (Mod Rheumatol 26:891, 2016) - Myositis (Ann Rheum Dis 77:55, 2018) but upregulate ICOS – the most closely related IgSF member - GvHD (Transpl Immunol 43-44:54, 2017) - IBD (Gastroenterology 143:62, 2012) - Multiple Sclerosis (Mult Scler 23:686, 2017) T cell activation: ICOS blockade (prezalumab) has ICOS ↑ been only modestly effective in: Naïve T cell CD28 ↓ (some cells) CD28+ ICOS+ CD28lo ICOS+ CD28- ICOS+ CD28+ ICOS- - Lupus (Arthritis Rheumatol 70:1071, 2018) CD28 Activated / Effector / Memory / T cells ICOS FH Various populations ICOS+ / TFH cells have been implicated in: - GvHD (JCI Insight 1:e86660, 2016) - Arthritis (Nature 542:110, 2017) - Sjögren’s (Arthritis Rheumatol 70:774, 2018) - Myositis (Brain 127:1182, 2004) (Gastroenterology 126:829, 2004) - IBD 6 - Multiple Sclerosis (PLoS One 8:e57820, 2013) ALPN-101: a First-In-Class Dual CD28/ICOS Antagonist For Multiple Inflammatory Disease Indications CD28 and ICOS: Currently Available ALPN-101 (ICOSL vIgD-Fc) Key T cell Costimulatory Therapeutics Block Blocks Both CD28 & ICOS Pathways Only the CD28 Pathway Pathways APC APC APC CD80 CD80 CD86 CD80 CD86 ICOSL ICOSL CD86 ICOSL CTLA4-Ig ALPN-101 † ICOSL vIgD-Fc* CD28 ICOS CD28 ICOS CD28 ICOS T cell T cell T cell †Orencia 2018 sales = $2.7B (Source: BMS) *Effectorless IgG1 Fc 7 Two Mechanisms of Action for ALPN-101: Dual CD28/ICOS Antagonist ICD28/CD86 Inhibition ICOS/ICOSL Inhibition Stimulation of Jurkat Transfectants with Stimulation of Jurkat Transfectants with K562-OKT3 and plate-bound WT CD86-Fc K562-OKT3 and Plate-Bound WT ICOSL-Fc 100 ALPN-101 ALPN-101 Belatacept 60 Anti-ICOSL mAb 80 Abatacept Abatacept 40 60 40 20 %Inhibition ofluciferase signal 20 0 %Inhibition of luciferase signal -2 0 2 4 0 -4 -2 0 2 4 Log Transform of Test Article Log Transform of Test Article -20 Concentration (nM) Concentration (nM) 8 Superior and Single-Dose Preclinical Activity in Multiple Diseases Humanized Graft-Versus-Host Disease (GVHD)1 Collagen-Induced Arthritis2 Additional Data Supports Other Therapeutic Areas: 3 1 Dillon et al TCT #426, 2019 • Multiple Sclerosis (Experimental Autoimmune Encephalomyelitis) 2 Dillon et al. ACR # 71873, 2018 3 Dillon et al. ANA #S254, 2018 4 4. ALPN, unpublished • Inflammatory Bowel Disease, Sjögren’s Syndrome 9 Initial Dual Orphan and Non-Orphan Clinical Development Plans 2019 2020 2021 2022 2023 Phase 1 Healthy Volunteer Early PK/PD Phase 1(b) Acute Graft-Versus-Host Disease Safety, PK Phase 2a Psoriatic Arthritis KLH immune responses Ex vivo SEB cytokines Phase 2b / Pivotal Trials Alternative Indications Current non-clinical coverage: 3 months Current GMP supply sufficient through phase 2a / 2021 10 ALPN-101 Development and Lifecycle Vision Drives Potential Value Ophthalmology • Broad, diverse market potential Uveitis • Initial trials in rheumatology and heme/onc will enable most efficient development Neuroscience Pulmonology Multiple Sclerosis Interstitial Lung Disease • Larger Indications approachable with partners Gastroenterology Crohn’s Disease Ulcerative Colitis Rheumatology Rheumatology Sjögren’s Syndrome Juvenile Idiopathic Arthritis Rheumatoid Arthritis Systemic Lupus Erythematosus Rheumatology Rheumatology Systemic Sclerosis Inflammatory Myositis Psoriatic Arthritis Vasculitis Heme/Onc Heme/Onc Transplantation Acute GvHD Chronic GvHD Allograft Rejection GvHD Prophylaxis Rejection Prophylaxis 11 ALPN-202 Oncology: Conditional T-cell Agonist 12 ALPN-202: A First-in-Class Tri-Functional Immuno-Oncology Biologic Dual PD-L1/CTLA-4 Antagonist and PD-L1-Dependent CD28 Agonist Without Adequate T Cell Activation, ALPN-202 Mediates PD-L1-Dependent PD-L1/PD-1 Inhibits T Cell PD-(L)1 Inhibitors Fail to Elicit (T Cell) CD28 Agonism Along with PD-L1 and Activation Anti-Tumor Responses CTLA-4 Inhibition Tumor (If present) MHC/ PD-L1 Pep PD-L1 PD-L1 CD80CD86 ALPN-202 (CD80 vIgD-Fc*) Anti-PD-1 CD28 PD-1 CTLA-4 PD-1 CD28 TCR/ CD3 +++ CD28 PD-1 TCR/ T cell Inadequate T CD3 Activation J Exp Med 1995;182:459-65 T cell Science 2017;355:1423-1427 cell activation * CD80 vIgD is an ALPN-proprietary variant CD80 IgV domain, engineered by directed evolution. The Fc is an effectorless IgG1 Fc. Approximately ~70% of Patients Receiving PD-1/L1 Therapy Do Not Respond 13 Three Primary Mechanisms of Action of ALPN-202 1. PD-L1 – PD-1 Antagonism 3. PD-L1-Dependent Costimulation 3000 (CD28 Agonism) 25000 ALPN-202 No PD-L1 2000 WT CD80-Fc 20000 Durvalumab (anti-PD-L1) MFI Nivolumab (anti-PD-1) 15000 1000 ALPN-202 Fc Control Atezolizumab (anti-PD-L1) 10000 Fc Control IL-2 (pg/mL) 0 5000 1 10 100 1000 10000 1000001000000 [pM] 0 1 100 10000 1000000 2. CTLA-4 – B7 Antagonism 25000 With PD-L1 60000 20000 15000 40000 10000 IL-2 (pg/mL) MFI ALPN-202 5000 20000 Ipilimumab (anti-CTLA-4) 0 Fc Control 1 100 10000 1000000 0 [pM] 1 10 100 1000 10000 1000001000000 Primary T cells + K562 ± hPD-L1 14 [pM] ALPN-202 Exhibits Potent Efficacy in vivo, Superior to PD-L1 Inhibition PD-L1 inhibitor-superior, Efficacy as Monotherapy …with Evidence of Anti-Tumor Immunity ) 3 2000 Fc Control 0/11 mice tumor free ALPN-202-pretreated ALPN-202 1500 durvalumab (anti PD-L1) Naive p < 0.0001 ALPN-202 vs durvalumab 1000 2/11 mice tumor free 500 8/11 mice tumor free Median Tumor Volume (mm Volume Tumor Median 0 0 10 20 30 40 Mice dosed Day Superior Intra-Tumoral Inflammatory Signatures 15 ALPN-202 Phase 1 Development Plan 2020 2021 2022 2023 Dose Escalation: Any Advanced Malignancy MTD or maximal PD Expansion Cohorts: Selected Tumor Types ± Biomarker Phase 1b Combination(s) TBD Pilot toxicology studies completed; definitive studies in progress GMP DS completed 16 vIgD Platform 17 Turning Immunoglobulin Superfamily (IgSF) Proteins Into Novel Therapeutics • IgSF is the largest human protein superfamily – 765 The CD28 Protein Family 1 genes identified
Recommended publications
  • TNF‑Α‑Mediated Epithelial‑To‑Mesenchymal Transition Regulates Expression of Immune Checkpoint Molecules in Hepatocellular Carcinoma
    MOLECULAR MEDICINE REPORTS 21: 1849-1860, 2020 TNF‑α‑mediated epithelial‑to‑mesenchymal transition regulates expression of immune checkpoint molecules in hepatocellular carcinoma RITU SHRESTHA1,2, KIM R. BRIDLE1,2, DARRELL H.G. CRAWFORD1,2 and APARNA JAYACHANDRAN1,2 1University of Queensland, Faculty of Medicine and 2Liver Cancer Unit, Gallipoli Medical Research Institute, Greenslopes Private Hospital, Brisbane, QLD 4120, Australia Received June 12, 2019; Accepted January 31, 2020 DOI: 10.3892/mmr.2020.10991 Abstract. Hepatocellular carcinoma (HCC) is the ligand (PD‑L)1, PD‑L2, CD73 and B7‑H3. In contrast, reversal fastest growing cause of cancer-related deaths globally. of EMT suppressed the expression of PD‑L1, PD‑L2, CD73 Epithelial-to-mesenchymal transition (EMT) is a cellular and B7‑H3. In addition, high expression of TNF-α and PD‑L1 process that confers HCC tumor cells with the ability to evade in 422 patients with HCC was associated with poor overall the immune system. Immune escape in most tumors, including survival. The coordinate expression of TNF-α with PD‑L2 in HCC, is controlled by immune checkpoint molecules. The aim this patient cohort was associated with increased HCC recur- of the present study was to investigate the association between rence. In conclusion, the present study demonstrated a close EMT and immune checkpoint in HCC, and identify novel ther- association between immune modulator expression and EMT apeutic targets for HCC. An in vitro model of reversible EMT induction/reversal driven by TNF-α. was utilized based on cytokine tumor necrosis factor (TNF)-α treatment of HCC cell lines Hep3B and PLC/PRF/5.
    [Show full text]
  • Superpixel Image Segmentation of VISTA Expression in Colorectal Cancer: Implications for Immunotherapy
    Superpixel Image Segmentation of VISTA Expression in Colorectal Cancer: Implications for Immunotherapy Dongling Wu Zucker School of Medicine at Hofstra Northwell Health Sean Hacking Zucker School of Medicine at Hofstra Northwell Health Taisia Vitkovski Zucker School of Medicine at Hofstra Northwell Health Mansoor Nasim ( [email protected] ) Zucker School of Medicine at Hofstra Northwell Health Research Article Keywords: Colorectal Carcinoma, v-domain immunoglobulin suppressor of T cell activation, VISTA, Immunohistochemistry, Superpixel, Clinical trials Posted Date: December 14th, 2020 DOI: https://doi.org/10.21203/rs.3.rs-120759/v1 License: This work is licensed under a Creative Commons Attribution 4.0 International License. Read Full License Page 1/24 Abstract Colorectal cancer is an overall bad player and accounts for 9% of all cancers. Today, advancements in immune checkpoint inhibition has provided therapeutics for many, but not all cancer patients. This issue is in part due to the tumoral microenvironment; which plays a signicant role in determining response to immune check point therapeutics. This study serves as the rst to evaluate a potent inhibitory checkpoint: V-domain immunoglobulin suppressor of T cell activation (VISTA) and its role in CRC. This was evaluated with both conventional light microscope and superpixel image segmentation. Here we found VISTA expression to be associated with low tumor budding, lower tumor stage, high tumor inltrating lymphocytes, mature stromal differentiation, BRAF mutation status and better disease-free survival in colorectal cancer. When comparing methodologies; superpixel image segmentation better stratied patients into prognostic groups. Anti-VISTA clinical trials are now open and recruiting for patient enrollment for patients with certain advanced solid tumors.
    [Show full text]
  • VISTA Is a Novel Broad-Spectrum Negative Checkpoint Regulator for Cancer Immunotherapy
    Cancer Immunology Cancer Immunology at the Crossroads: Experimental Immunotherapies Research VISTA Is a Novel Broad-Spectrum Negative Checkpoint Regulator for Cancer Immunotherapy J. Louise Lines1,2, Lorenzo F. Sempere3, Thomas Broughton4,5, Li Wang7, and Randolph Noelle1,2,4,5,6 Abstract In the past few years, the field of cancer immunotherapy has made great progress and is finally starting to change the way cancer is treated. We are now learning that multiple negative checkpoint regulators (NCR) restrict the ability of T-cell responses to effectively attack tumors. Releasing these brakes through antibody blockade, first with anti-CTLA4 and now followed by anti-PD1 and anti-PDL1, has emerged as an exciting strategy for cancer treatment. More recently, a new NCR has surfaced called V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA). This NCR is predominantly expressed on hematopoietic cells, and in multiple murine cancer models is found at particularly high levels on myeloid cells that infiltrated the tumors. Preclinical studies with VISTA blockade have shown promising improvement in antitumor T-cell responses, leading to impeded tumor growth and improved survival. Clinical trials support combined anti-PD1 and anti-CTLA4 as safe and effective against late-stage melanoma. In the future, treatment may involve combination therapy to target the multiple cell types and stages at which NCRs, including VISTA, act during adaptive immune responses. Cancer Immunol Res; 2(6); 510–7. Ó2014 AACR. Introduction Negative Checkpoint Regulators, New and Old The concept of immunosurveillance for cancer was pro- Molecules that promote or interfere with the mounting of posed by Burnet (1), who posited that transformed cells protective antitumor immunity are under intensive study.
    [Show full text]
  • Combining Immune Checkpoint Inhibitors: Established and Emerging Targets and Strategies to Improve Outcomes in Melanoma
    King’s Research Portal DOI: 10.3389/fimmu.2019.00453 Document Version Publisher's PDF, also known as Version of record Link to publication record in King's Research Portal Citation for published version (APA): Khair, D. O., Bax, H. J., Mele, S., Crescioli, S., Pellizzari, G., Khiabany, A., Nakamura, M., Harris, R. J., French, E., Hoffmann, R. M., Williams, I. P., Cheung, K. K. A., Thair, B., Beales, C. T., Touizer, E., Signell, A. W., Tasnova, N. L., Spicer, J. F., Josephs, D. H., ... Karagiannis, S. N. (2019). Combining Immune Checkpoint Inhibitors: Established and Emerging Targets and Strategies to Improve Outcomes in Melanoma. Frontiers in Immunology , (MAR), [453]. https://doi.org/10.3389/fimmu.2019.00453 Citing this paper Please note that where the full-text provided on King's Research Portal is the Author Accepted Manuscript or Post-Print version this may differ from the final Published version. If citing, it is advised that you check and use the publisher's definitive version for pagination, volume/issue, and date of publication details. And where the final published version is provided on the Research Portal, if citing you are again advised to check the publisher's website for any subsequent corrections. General rights Copyright and moral rights for the publications made accessible in the Research Portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognize and abide by the legal requirements associated with these rights. •Users may download and print one copy of any publication from the Research Portal for the purpose of private study or research.
    [Show full text]
  • Spatially Resolved and Quantitative Analysis of VISTA/PD-1H As a Novel Immunotherapy Target in Human Non–Small Cell Lung Cance
    Published OnlineFirst December 4, 2017; DOI: 10.1158/1078-0432.CCR-17-2542 Personalized Medicine and Imaging Clinical Cancer Research Spatially Resolved and Quantitative Analysis of VISTA/PD-1H as a Novel Immunotherapy Target in Human Non–Small Cell Lung Cancer Franz Villarroel-Espindola1, Xiaoqing Yu2, Ila Datar1, Nikita Mani1, Miguel Sanmamed3, Vamsidhar Velcheti4, Konstantinos Syrigos5, Maria Toki1, Hongyu Zhao2, Lieping Chen3, Roy S. Herbst6, and Kurt A. Schalper1,6 Abstract Purpose: Determine the localized expression pattern and clin- Expression in tumor and stromal cells was seen in 21% and ical significance of VISTA/PD-1H in human non–small cell lung 98% of cases, respectively. The levels of VISTA were positively þ þ cancer (NSCLC). associated with PD-L1, PD-1, CD8 TcellsandCD68 Experimental Design: Using multiplex quantitative immuno- macrophages. VISTA expression was higher in T-lymphocytes fluorescence (QIF), we performed localized measurements of than in macrophages; and in cytotoxic T cells than in T-helper VISTA, PD-1, and PD-L1 protein in 758 stage I–IV NSCLCs from cells. Elevated VISTA was associated with absence of EGFR 3 independent cohorts represented in tissue microarray format. mutations and lower mutational burden in lung adenocarci- þ The targets were selectively measured in cytokeratin tumor nomas. Presence of VISTA in tumor compartment predicted þ þ þ epithelial cells, CD3 T cells, CD4 T-helper cells, CD8 cytotoxic longer 5-year survival. þ þ T cells, CD20 B lymphocytes and CD68 tumor-associated Conclusions: VISTA is frequently expressed in human NSCLC macrophages. We determined the association between the targets, and shows association with increased tumor-infiltrating lympho- clinicopathological/molecular variables and survival.
    [Show full text]
  • Spatially Resolved and Quantitative Analysis of VISTA/PD-1H As a Novel
    Author Manuscript Published OnlineFirst on December 4, 2017; DOI: 10.1158/1078-0432.CCR-17-2542 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 Spatially resolved and quantitative analysis of VISTA/PD-1H as a novel immunotherapy target in 2 human non-small cell lung cancer 3 Franz Villarroel-Espindolaa, Xiaoqing Yub, Ila Datara, Nikita Mania, Miguel Sanmamedc, Vamsidhar 4 Velchetid, Konstantinos Syrigose, Maria Tokia, Hongyu Zhaob, Lieping Chenc, Roy S. Herbstf and Kurt A. 5 Schalpera,f 6 aDepartment of Pathology, Yale School of Medicine, New Haven, CT, 06520 USA. 7 bDepartment of Public Health, Yale School of Medicine, New Haven, CT, 06520 USA. 8 cImmunobiology, Yale School of Medicine, New Haven, CT, 06520 USA. 9 dSolid Tumor Oncology, Cleveland Clinic Foundation, Cleveland, OH, 44195 USA. 10 eOncology Unit GPP, Athens School of Medicine, Greece. 11 fMedical Oncology and Yale Cancer Center, New Haven, CT, 06520 USA. 12 Running title: Role of VISTA/PD-1H in NSCLC. 13 Keywords: NSCLC, VISTA/PD-1H, PD-1 pathway, TILs 14 Correspondence to: 15 Kurt A. Schalper, M.D./Ph.D. 16 Office Address: 333 Cedar St. FMP117, New Haven, CT 06520-8023 17 Email: [email protected] 18 Financial support: 19 Lung Cancer Research Foundation (LCRF), Yale SPORE in Lung Cancer (P50CA196530), Department of 20 Defense-Lung Cancer Research Program Career Development Award (LC150383), Stand Up to Cancer – 21 American Cancer Society Lung Cancer Dream Team Translational Research Grant (Grant Number: SU2C- 22 AACR-DT17-15). Stand Up To Cancer is a program of the Entertainment Industry Foundation.
    [Show full text]
  • Evaluating the Potential for B7-H4 As an Immunoregulatory Target
    Author Manuscript Published OnlineFirst on March 21, 2017; DOI: 10.1158/1078-0432.CCR-15-2440 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 1 2 Molecular Pathways: Evaluating the Potential for B7-H4 as an Immunoregulatory Target 3 Heather L. MacGregor1,2 and Pamela S. Ohashi1-3* 4 5 6 7 1Campbell Family Institute for Breast Cancer Research, Princess Margaret Cancer Centre, 8 Toronto, Ontario, Canada 9 2Department of Immunology, University of Toronto, Toronto, Ontario, Canada 10 3Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada 11 12 13 Running Title: Evaluating B7-H4 as a Potential Immunoregulatory Target 14 Keywords: B7-H4; immunosuppression; co-inhibitory molecule; immunotherapy; tumor 15 microenvironment 16 17 Funding sources: This work was supported by grants from the Canadian Institute for Health 18 Research awarded to P.S. Ohashi. 19 20 *Corresponding Author: Pamela S. Ohashi, Princess Margaret Cancer Centre, 610 University 21 Avenue, Suite 8-407, Toronto, Ontario M5G 2M9, Canada. Phone: 416-946-2357; Fax: 416-204- 22 2276; e-mail: [email protected] 23 Disclosures: The authors have no financial conflicts of interest. 1 Downloaded from clincancerres.aacrjournals.org on September 27, 2021. © 2017 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 21, 2017; DOI: 10.1158/1078-0432.CCR-15-2440 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. 24 (Abstract: 152 words; Body: 2937 words; 2 figures, 1 table) 25 Abstract: With the clinical success of CTLA-4 and PD-1 blockade in treating malignancies, 26 there is tremendous interest in finding new ways to augment anti-tumor responses by targeting 27 other inhibitory molecules.
    [Show full text]
  • Activation of the Receptor Tyrosine Kinase AXL Regulates the Immune Microenvironment in Glioblastoma Hirokazu Sadahiro1,2, Kyung-Don Kang1, Justin T
    Published OnlineFirst March 12, 2018; DOI: 10.1158/0008-5472.CAN-17-2433 Cancer Tumor Biology and Immunology Research Activation of the Receptor Tyrosine Kinase AXL Regulates the Immune Microenvironment in Glioblastoma Hirokazu Sadahiro1,2, Kyung-Don Kang1, Justin T. Gibson3, Mutsuko Minata1, Hai Yu1,4, Junfeng Shi5, Rishi Chhipa6, Zhihong Chen7, Songjian Lu8, Yannick Simoni9, Takuya Furuta10, Hemragul Sabit11, Suojun Zhang1,12, Soniya Bastola1, Shinobu Yamaguchi1, Hebaallah Alsheikh1, Svetlana Komarova1, Jun Wang1, Sung-Hak Kim13, Dolores Hambardzumyan7, Xinghua Lu8,EvanW.Newell9, Biplab DasGupta6, Mitsutoshi Nakada11, L. James Lee5, Burt Nabors1, Lyse A. Norian3,14, and Ichiro Nakano1,14 Abstract Glioblastoma (GBM) is a lethal disease with no effective ther- We also found that treatment of GSC-derived mouse GBM tumors apies available. We previously observed upregulation of the TAM with nivolumab, a blocking antibody against the immune check- (Tyro-3, Axl, and Mer) receptor tyrosine kinase family member AXL point protein PD-1, increased intratumoral macrophages/micro- in mesenchymal GBM and showed that knockdown of AXL glia and activation of AXL. Combinatorial therapy with nivolumab induced apoptosis of mesenchymal, but not proneural, glioma plus BGB324 effectively prolonged the survival of mice bearing sphere cultures (GSC). In this study, we report that BGB324, a GBM tumors. Clinically, expression of AXL or PROS1 was associ- novel small molecule inhibitor of AXL, prolongs the survival of ated with poor prognosis for patients with GBM. Our results immunocompromised mice bearing GSC-derived mesenchymal suggest that the PROS1–AXL pathway regulates intrinsic mesen- GBM-like tumors. We show that protein S (PROS1), a known chymal signaling and the extrinsic immune microenvironment, ligand of other TAM receptors, was secreted by tumor-associated contributing to the growth of aggressive GBM tumors.
    [Show full text]
  • Francis Collins' Notes on the Press Release
    '-\ \\ \ EMBARGOED UNTIL 10 am EST, February 12, 2001 International Human Genome Sequencing Consortium Publishes Sequence and Analysis of the Human Genome Washington, D.C., Feb. 12, 2'1-The Human Genome Project international consortium today it has ptlblished a draft sequence and initial analysis of the human genome-the genetic blueprint for a human being. The paper will be published in the Feb. 15 issue of the journal Nature. The draft sequence, which covers more than 90 percent of the human genome, represents the exact order of DNA's four chemical bases, commonly abbreviated A, T, C and G, arrayed along the human chromosomes. This text underlies and shapes ever man trait-from eye color and height, to health, diseases, an 7 The Consortium's initial analysis of this text describes some of the insights that can be gleaned from the genome and represents scientists' first glimpse r\JJ.. of the human DNA terrain- its quirks and curiosities, as well as the extraordinary trove of information about human development, physiology, medicine, and evolution. The results reported in Nature paper represents the next milestone since the , consortium's announcement on June 26 that it had a fiist ..assembly gf tho human goRome. Tlie fi:fst assembly aH:fl6tlnced in June -.vas ,analt:Jgotts to ha...-ing a eolleetit:Jn of all thQ letters that percent of "tl..t_ "i the text for the "Book of Life." This week's paper representra;c;ompilation of _u these . into .the first draft of a readable GoA '"•, 1> p • small gaps still remaining to be filled in this text, but 11'-CAN"'\ scientists are already getting of what the genome topography looks like it has to tell.
    [Show full text]
  • Changes in the Adipose Tissue Expression of CD86 Costimulatory
    Sindhu et al., J Glycomics Lipidomics 2015, 5:4 DOI: 10.4172/2153-0637.1000135 Journal of Glycomics & Lipidomics Research Article Open Access Changes in the Adipose Tissue Expression of CD86 Costimulatory Ligand and CD163 Scavenger Receptor in Obesity and Type-2 Diabetes: Implication for Metabolic Disease Sardar Sindhu1*, Reeby Thomas1, Shihab Kochumon1, Eman Alshawaf2, Amal Hasan1, Munera Alghanim1, Kazem Behbehanil2 and Rasheed Ahmad1* 1Immunology & Innovative Cell Therapy Unit, Dasman Diabetes Institute (DDI), Dasman 15462, Kuwait 2Pancreatic Islet Biology and Transplantation Unit, Dasman Diabetes Institute (DDI), Dasman 15462, Kuwait Abstract Background: The CD86 costimulatory ligand and CD163 scavenger receptor are expressed on monocytes/macrophages. However, modulations in the expression of these immune regulatory receptors in metabolic disease remain unclear. We, therefore, compared the adipose tissue expression of CD86/CD163 and signature inflammatory cytokines/chemokines in non-diabetic and diabetic individuals. Methods: Subcutaneous adipose tissue biopsies were collected from 57 non-diabetics and 46 diabetics classified based on body mass index as obese, overweight and lean and the expression of CD86/CD163 was assessed by quantitative RT-PCR, immunohistochemistry/ confocal microscopy. Results: The data show that CD86 and CD163 gene expression was elevated in non-diabetic obese individuals as compared with their lean counterparts (P [CD86] = 0.0035; P [CD163] = 0.0028). As expected, CD86 and CD163 protein expression was also found to be elevated in the adipose tissue samples of obese individuals. The CD86 gene expression in non-diabetic and diabetic individuals correlated positively (P<0.05) with that of TNF-α (r [non-diabetic] = 0.26; r [diabetic] = 0.49), IL-18 (r [non-diabetic] = 0.67; r [diabetic] = 0.63), and IL-8 (r [non-diabetic] = 0.38; r [diabetic] = 0.33).
    [Show full text]
  • VISTA Regulates the Development of Protective Antitumor Immunity
    Cancer Microenvironment and Immunology Research VISTA Regulates the Development of Protective Antitumor Immunity Isabelle Le Mercier1, Wenna Chen1, Janet L. Lines3,4, Maria Day2, Jiannan Li1, Petra Sergent1, Randolph J. Noelle1,3,4, and Li Wang1 Abstract V-domain Ig suppressor of T-cell activation (VISTA) is a novel negative checkpoint ligand that is homologous to PD-L1 and suppresses T-cell activation. This study demonstrates the multiple mechanisms whereby VISTA relieves negative regulation by hematopoietic cells and enhances protective antitumor immunity. VISTA is highly þ þ expressed on myeloid cells and Foxp3 CD4 regulatory cells, but not on tumor cells within the tumor microenvironment (TME). VISTA monoclonal antibody (mAb) treatment increased the number of tumor- specific T cells in the periphery and enhanced the infiltration, proliferation, and effector function of tumor- reactive T cells within the TME. VISTA blockade altered the suppressive feature of the TME by decreasing the presence of monocytic myeloid-derived suppressor cells and increasing the presence of activated dendritic cells within the tumor microenvironment. In addition, VISTA blockade impaired the suppressive function and reduced þ þ the emergence of tumor-specific Foxp3 CD4 regulatory T cells. Consequently, VISTA mAb administration as a monotherapy significantly suppressed the growth of both transplantable and inducible melanoma. Initial studies explored a combinatorial regimen using VISTA blockade and a peptide-based cancer vaccine with TLR agonists as adjuvants. VISTA blockade synergized with the vaccine to effectively impair the growth of established tumors. Our study therefore establishes a foundation for designing VISTA-targeted approaches either as a monotherapy or in combination with additional immune-targeted strategies for cancer immunotherapy.
    [Show full text]
  • High Expression of Immune Checkpoints Is Associated with the TIL Load, Mutation Rate and Patient Survival in Colorectal Cancer
    INTERNATIONAL JOURNAL OF ONCOLOGY 57: 237-248, 2020 High expression of immune checkpoints is associated with the TIL load, mutation rate and patient survival in colorectal cancer MARA KITSOU1, GEORGIOS D. AYIOMAMITIS2 and APOSTOLOS ZARAVINOS3 1Department of Life Sciences, School of Sciences, European University Cyprus, 1516 Nicosia, Cyprus; 2First Department of Surgery, Tzaneio General Hospital, 18536 Piraeus, Greece; 3College of Medicine, Member of QU Health, Qatar University, P.O. Box 2713 Doha, Qatar Received February 10, 2020; Accepted April 13, 2020 DOI: 10.3892/ijo.2020.5062 Abstract. Adoptive cell therapy with the use of tumor- ‘central memory T-cell’ and ‘exhausted T-cell’ gene signa- infiltrating lymphocytes (TILs) is a very promising tures were significantly lower among the READ tumors. The immunotherapeutic approach for the treatment of patients expression of PD‑1, PD‑L1, PD‑L2, CTLA‑4 and IDO1 was with colorectal cancer (CRC). However, within the tumor significantly higher among COAD patients with a high muta- microenvironment, co-inhibitory immune checkpoints can tion rate (>34 mutations/Mb) compared to those with a lower inactivate TILs. The aim of the present study was to examine rate. Somatic mutations in PD‑1, PD‑L1, CTLA‑4 and other the association between the TIL load, the mutation rate and checkpoint molecules did not seem to affect their expression the clinical outcome in the immune landscape of patients with levels. On the whole, the data of the present study highlight CRC. RNA-seq and whole exome seq data of 453 colon adeno- the association of immune checkpoint molecules with the TIL carcinomas (COAD) and rectal adenocarcinomas (READ), load, patient survival and a high mutation rate in CRC.
    [Show full text]