Adcs and Solid Tumours the Payload Revolution That Drives Therapeutic Opportunities in Unmet Need

Total Page:16

File Type:pdf, Size:1020Kb

Adcs and Solid Tumours the Payload Revolution That Drives Therapeutic Opportunities in Unmet Need Therapeutics ADCs and solid tumours the payload revolution that drives therapeutic opportunities in unmet need The fight against cancer is ongoing. Despite progress in many indications, mortality rates for some of the most difficult to treat solid tumour types have not improved substantially since the early 1970s. By Dr Jenny Thirlway en-year survival statistics for oesophagus 90 ADCs in clinical trials as of January 20202. The and lung cancer have shown an increase of global market for ADCs is anticipated to reach Tless than 10% and pancreatic cancer has $7.5 billion by 2025. shown no improvement in survival rates at all1. First generation ADCs suffered from a lack of Even for cancers where survival rates have potency, as existing chemotherapeutic agents with improved, such as malignant melanoma, breast known toxicity profiles such as methotrexate, vin- and uterine, there is still a need for more effica- blastine and doxorubicin were conjugated in an cious treatments which importantly, do not come effort to increase their specificity. The cBR96-dox- with debilitating side effects. orubicin conjugate showed promise in pre-clinical Antibody Drug Conjugate (ADC) technology studies and progressed to a Phase II clinical trial but has been the focus of intense interest as a means to was insufficiently efficacious. The effectiveness of an provide selective tumour killing with increased effi- ADC depends on efficiency of internalisation and cacy and fewer side-effects than standard of care the failure of this conjugate was largely attributed to chemotherapies. ADCs comprise a monoclonal under-potency of the doxorubicin payload. It is esti- antibody (or antibody fragment) that targets a mated that 4-12 million doxorubicin molecules are tumour-associated antigen, conjugated via a chem- required to kill a cell. Antigen expression levels are ical linker to a highly cytotoxic entity. Binding of generally less than one million copies per cell mak- the antibody to the cell surface triggers internalisa- ing it difficult to achieve a critical concentration of tion, and processing within endosomes or lyso- toxin3. Lessons from the story of cBR96-doxoru- somes releases the potent cell-killing molecule. bicin established the use of more potent cytotoxic Combining the targeting power of an antibody payloads to counteract issues with payload delivery. with a potent cytotoxic agent makes it possible to The first ADC to be approved, Mylotarg® for eradicate cancer cells more effectively and selec- treatment of acute myelogenous leukaemia (AML), tively, while reducing the side-effects which under- is conjugated to calicheamicin. Mylotarg® was mine patient quality of life. Currently, seven ADCs originally approved by the FDA in 2000 and sub- have been approved (four for haematological can- sequently withdrawn in 2010 following an unsuc- cers and three for solid tumours) and the clinical cessful confirmatory Phase III trial4. Calicheamicin and commercial potential of ADC technology is is a potent anti-tumour antibiotic that associates reflected in the burgeoning pipeline, with around with the minor groove of DNA causing double 24 Drug Discovery World Spring 2020 Therapeutics Mechanism of action of ADCs strand DNA breaks. The failure of Mylotarg® was bind to the ‘plus’ end of growing microtubules partly attributed to an unstable linker that released blocking polymerisation of tubulin dimers. Two out payload prematurely. It was reapproved by the of three approvals of ADCs in solid tumours FDA in September 2017 and by the EMA in April employ tubulin inhibitors as the payload. Kadcyla® 2018 on an altered dosing regimen. Another was approved by the FDA in February 2013 for the calicheamicin-based ADC, Besponsa®, was also treatment of Her2 positive metastatic breast cancer approved in 2017 for the treatment of relapsed or and is comprised of the anti-Her2 antibody refractory acute lymphoblastic leukaemia, but trastuzumab conjugated to the maytansine DM1. three other calicheamicin ADCs in development Padcev™ was granted accelerated approval by the have been discontinued due to having a very nar- FDA in December 2019 for treatment of locally- row Therapeutic Index (TI). advanced or metastatic urothelial cancer and is con- Around 40% of clinically-active ADCs employ jugated to the auristatin MMAE. tubulin inhibitors from the auristatin and maytan- Tubulysins have been developed as next genera- sine classes5. Auristatins are derived from dolas- tion tubulin inhibitors. Tubulysins bind to the tatin 10, isolated from the marine sea hare vinca binding site of tubulin and exhibit potent Dolabella auricularia. They block tubulin assembly cytotoxic activity in the picomolar (pM) range. and cause G2/M phase cell cycle arrest. One of the Unlike the auristatins and maytansines, tubulysins first ADCs to be approved, Adectris®, for treatment have been shown to bypass drug efflux pumps and of Hodgkin lymphoma and systemic anaplastic retain potency in MDR1 expressing cell lines6. large cell lymphoma, is conjugated to Monomethyl Three out of four of the tubulysin-based conjugates auristatin E (MMAE), as is Polivy™ for the treat- (two Small Molecule Drug Conjugates and one ment of relapsed refractory diffuse large B cell lym- ADC) to have entered the clinical development phoma. Maytansine was isolated from the bark of phase have been discontinued due to either safety the African shrub Maytenus ovatus and is a potent or efficacy reasons. inhibitor of microtubulin assembly. Maytansines Although first-generation tubulin inhibitors Drug Discovery World Spring 2020 25 Therapeutics A structural model of an Antibody Drug Conjugate (photograph courtesy of Iksuda Therapeutics) have been approved in several indications, they are to the nucleophilic C2-amino group of a guanine not as effective in indications with lower expres- base and are 600 times more potent than PBD sion of target antigen or cells that are less sensitive monomers in vitro. As with tubulysins, PBDs avoid to tubulin inhibition. It has become clear that there MDR1 mediated drug resistance7. The first PBD is a need for payloads with increased potency and dimer-based ADCs entered the clinic in 2013. alternative mechanism of action to the tubulin Vadastuximab talirine targets CD33 which is over- inhibitor class. There has therefore been a push expressed in AML. Pre-clinically vadastuximab towards the design of payloads to address targets talirine was shown to be more potent than where tubulin ADCs are proving to be insufficient- Mylotarg® against a panel of AML cell lines and ly active, such as colon cancer. primary AML cells in vitro and in vivo. Unlike Third-generation ADCs have looked to payloads Mylotarg®, anti-leukaemic activity was observed with novel modes of action and activity against in AML models with the multidrug-resistant phe- non-proliferating cells so targets can be widened to notype8. However, in June of 2017 Seattle Genetics include tumour initiating cells (TICs). Higher announced the discontinuation of the Phase III potency allows for targeting of antigens with rela- CASCADE clinical trial of vadastuximab talirine tively low tumour expression levels. (SGN-CD33A) in frontline older AML patients fol- Pyrrolobenzodiazepines (PBD) are based on nat- lowing consultation with the Independent Data urally-occurring, anti-tumour antibiotics that bind Monitoring Committee (IDMC) and after review- to the DNA minor groove in a sequence-specific ing unblinded data. The data indicated a higher manner. PBD dimers can form interstrand and rate of deaths, including fatal infections in the intrastrand DNA cross-links by covalently binding vadastuximab talirine-containing arm versus the 26 Drug Discovery World Spring 2020 Therapeutics control arm of the trial. In March 2018 Seattle tivity reaction (7%) and pleural effusion (5%)9. Genetics announced it was no longer developing The development of Rova-T was discontinued in vadastuximab tailrine as a result of recent portfolio August 2019 after the IDMC recommended termi- and resource prioritisation decisions. nating the Phase III MERU study due to lack of Rovalpituzumab tesirine (Rova-T) targets cancer survival benefit. stem cell-associated target delta-like protein 3 Since 2013, 20 PBD dimer-based ADCs have (DLL3) which is over-expressed in small cell lung entered clinical development with eight being dis- cancer (SCLC), a solid tumour with few treatment continued. The apparent narrow TI of PBD dimer- options. In Phase I/II studies of relapsed SCLC based drugs has led to a shift away from cross-link- patients who had previously failed one or more ers towards DNA monoalkylators such as the standard therapies, Rova-T demonstrated overall indolinobenzodiazepine pseudodimers (IGNs) and response rates of 44% in the patients identified duocarmycins. with high expression of DLL3. Based on these Within the IGN class, it was discovered that the results, in 2016 Abbvie announced that it would cross-linking analogues had an unfavourable toxi- acquire Stemcentrx and its lead late-stage asset city profile in mice with the animals displaying rovalpituzumab tesirine in a deal worth upwards prolonged body weight loss, as well as delayed of $9 billion. By 2018, things were looking less lethality. Development of a DNA alkylating IGN promising when disappointing results were payload that does not have cross linking capability announced for the Phase II TRINITY trial. An provided a distinct mode of action compared with independent review committee assessed efficacy as that of the PBDs. In vivo testing revealed that the an overall response rate of 16% (95% CI, 11, 22) alkylating IGN ADC did not display any signs of with the most common (≥5%) grade ≥3 severe tox- prolonged or delayed toxicity10. icities being thrombocytopenia (11%), photosensi- To date, there are four pre-clinical IGN-based Empower Your Research With IncuCyte® Live-Cell Analysis The IncuCyte® Live-Cell Analysis System speeds scientific discovery by combining lab-tested protocols and reagents with powerful, automated image acquisition and analysis. Gain dynamic insights into the health, morphology, movement and function of cell models, all from the stable environment of a tissue culture incubator.
Recommended publications
  • Prior Approaches and Future Directions Marna Williams1, Anna Spreafico2, Kapil Vashisht3, Mary Jane Hinrichs4
    Author Manuscript Published OnlineFirst on June 16, 2020; DOI: 10.1158/1535-7163.MCT-19-0993 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Patient Selection Strategies to Maximize Therapeutic Index of Antibody Drug Conjugates: Prior Approaches and Future Directions Marna Williams1, Anna Spreafico2, Kapil Vashisht3, Mary Jane Hinrichs4 1Translational Medicine, Oncology, AstraZeneca, Gaithersburg, USA 2Drug Development Program, Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada 3AstraZeneca, Gaithersburg, USA. 4Early Development Oncology, Ipsen LLC, USA Corresponding author: Marna Williams Email: [email protected] Postal address: One MedImmune Way, Gaithersburg, MD, 20878 Phone: 301-398-1241 Fax: 240-306-1545 Running title Patient Selection Strategies for Antibody Drug Conjugates Keywords Antibody drug conjugates; therapeutic index; patient selection; biomarkers; cancer Conflicts of interest This study was funded by AstraZeneca. MW, KV and MJH are all current or former employees of AstraZeneca with stock/stock options in AstraZeneca. AS has been Advisory Board Consultant to Merck, Bristol-Myers Squibb, Novartis, Oncorus, Janssen, and has received Grant/Research support from Novartis, Bristol-Myers Squibb, Symphogen AstraZeneca/Medimmune, Merck, Bayer, Surface Oncology, Northern Biologics, Janssen Oncology/Johnson & Johnson, Roche, Regeneron, and Alkermes. Word count: Abstract: 197 Manuscript: 5,268 (excluding abstract) + 683 figure/table legends and footnotes Figures/tables: 5 (as agreed) 1 Downloaded from mct.aacrjournals.org on September 24, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on June 16, 2020; DOI: 10.1158/1535-7163.MCT-19-0993 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.
    [Show full text]
  • Predictive QSAR Tools to Aid in Early Process Development of Monoclonal Antibodies
    Predictive QSAR tools to aid in early process development of monoclonal antibodies John Micael Andreas Karlberg Published work submitted to Newcastle University for the degree of Doctor of Philosophy in the School of Engineering November 2019 Abstract Monoclonal antibodies (mAbs) have become one of the fastest growing markets for diagnostic and therapeutic treatments over the last 30 years with a global sales revenue around $89 billion reported in 2017. A popular framework widely used in pharmaceutical industries for designing manufacturing processes for mAbs is Quality by Design (QbD) due to providing a structured and systematic approach in investigation and screening process parameters that might influence the product quality. However, due to the large number of product quality attributes (CQAs) and process parameters that exist in an mAb process platform, extensive investigation is needed to characterise their impact on the product quality which makes the process development costly and time consuming. There is thus an urgent need for methods and tools that can be used for early risk-based selection of critical product properties and process factors to reduce the number of potential factors that have to be investigated, thereby aiding in speeding up the process development and reduce costs. In this study, a framework for predictive model development based on Quantitative Structure- Activity Relationship (QSAR) modelling was developed to link structural features and properties of mAbs to Hydrophobic Interaction Chromatography (HIC) retention times and expressed mAb yield from HEK cells. Model development was based on a structured approach for incremental model refinement and evaluation that aided in increasing model performance until becoming acceptable in accordance to the OECD guidelines for QSAR models.
    [Show full text]
  • Advances and Limitations of Antibody Drug Conjugates for Cancer
    biomedicines Review Advances and Limitations of Antibody Drug Conjugates for Cancer Candice Maria Mckertish and Veysel Kayser * Sydney School of Pharmacy, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia; [email protected] * Correspondence: [email protected]; Tel.: +61-2-9351-3391 Abstract: The popularity of antibody drug conjugates (ADCs) has increased in recent years, mainly due to their unrivalled efficacy and specificity over chemotherapy agents. The success of the ADC is partly based on the stability and successful cleavage of selective linkers for the delivery of the payload. The current research focuses on overcoming intrinsic shortcomings that impact the successful devel- opment of ADCs. This review summarizes marketed and recently approved ADCs, compares the features of various linker designs and payloads commonly used for ADC conjugation, and outlines cancer specific ADCs that are currently in late-stage clinical trials for the treatment of cancer. In addition, it addresses the issues surrounding drug resistance and strategies to overcome resistance, the impact of a narrow therapeutic index on treatment outcomes, the impact of drug–antibody ratio (DAR) and hydrophobicity on ADC clearance and protein aggregation. Keywords: antibody drug conjugates; drug resistance; linkers; payloads; therapeutic index; target specific; ADC clearance; protein aggregation Citation: Mckertish, C.M.; Kayser, V. Advances and Limitations of Antibody Drug Conjugates for 1. Introduction Cancer. Biomedicines 2021, 9, 872. Conventional cancer therapy often entails a low therapeutic window and non-specificity https://doi.org/10.3390/ of chemotherapeutic agents that consequently affects normal cells with high mitotic rates biomedicines9080872 and provokes an array of adverse effects, and in some cases leads to drug resistance [1].
    [Show full text]
  • Patient Resource Free
    PATIENT RESOURCE FREE Third Edition CancerUnderstanding Immunotherapy Published in partnership with CONTENT REVIEWED BY A DISTINGUISHED PRP MEDICAL PATIENT ADVISORY RESOURCE BOARD PUBLISHING® Understanding TABLE OF CONTENTS Cancer Immunotherapy Third Edition IN THIS GUIDE 1 Immunotherapy Today 2 The Immune System 4 Immunotherapy Strategies 6 Melanoma Survivor Story: Jane McNee Chief Executive Officer Mark A. Uhlig I didn’t look sick, so I didn’t want to act sick. Publisher Linette Atwood Having and treating cancer is only one part of your life. Co-Editor-in-Chief Charles M. Balch, MD, FACS Jane McNee, melanoma survivor Co-Editor-in-Chief Howard L. Kaufman, MD, FACS Senior Vice President Debby Easum 7 The Road to Immunotherapy Vice President, Operations Leann Sandifar 8 Cancer Types Managing Editor Lori Alexander, MTPW, ELS, MWC™ 14 Side Effects Senior Editors Dana Campbell Colleen Scherer 15 Glossary Graphic Designer Michael St. George 16 About Clinical Trials Medical Illustrator Todd Smith 16 Cancer Immunotherapy Clinical Trials by Disease Production Manager Jennifer Hiltunen 35 Support & Financial Resources Vice Presidents, Amy Galey Business Development Kathy Hungerford 37 Notes Stephanie Myers Kenney Account Executive Melissa Amaya Office Address 8455 Lenexa Drive CO-EDITORS-IN-CHIEF Overland Park, KS 66214 For Additional Information [email protected] Charles M. Balch, MD, FACS Advisory Board Visit our website at Professor of Surgery, The University of Texas PatientResource.com to read bios of MD Anderson Cancer Center our Medical and Patient Advisory Board. Editor-in-Chief, Patient Resource LLC Editor-in-Chief, Annals of Surgical Oncology Past President, Society of Surgical Oncology For Additional Copies: To order additional copies of Patient Resource Cancer Guide: Understanding Cancer Immunotherapy, Howard L.
    [Show full text]
  • IMGN779, a Novel CD33-Targeting Antibody-Drug Conjugate with DNA Alkylating Activity, Exhibits Potent Antitumor Activity in Models of AML
    Author Manuscript Published OnlineFirst on March 27, 2018; DOI: 10.1158/1535-7163.MCT-17-1077 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. IMGN779, a Novel CD33-Targeting Antibody-Drug Conjugate with DNA Alkylating Activity, Exhibits Potent Antitumor Activity in Models of AML Yelena Kovtun1*, Paul Noordhuis2*, Kathleen R. Whiteman1, Krystal Watkins1, Gregory E. Jones1, Lauren Harvey1, Katharine C. Lai1, Scott Portwood3, Sharlene Adams1, Callum M. Sloss1, Gerrit Jan Schuurhuis2, Gert Ossenkoppele2, Eunice S. Wang3, and Jan Pinkas1 1ImmunoGen, Inc., Waltham, Massachusetts. 2Department of Hematology, VU University Medical Center, Amsterdam, The Netherlands. 3Department of Medicine, Roswell Park Cancer Institute, Buffalo, New York *These authors contributed equally to the work Running Title: Preclinical activity of IMGN779 in AML Corresponding author: Yelena Kovtun, ImmunoGen Inc., 830 Winter St, Waltham, MA 02451. Phone: (781) 895-0716; Fax: (781) 895-0611; Email: [email protected] Potential conflict of interests: E.S. Wang received financial support from, and has served on an advisory board for, Immunogen Inc. There are no other competing interests to declare. 1 Downloaded from mct.aacrjournals.org on September 24, 2021. © 2018 American Association for Cancer Research. Author Manuscript Published OnlineFirst on March 27, 2018; DOI: 10.1158/1535-7163.MCT-17-1077 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract The myeloid differentiation antigen CD33 has long been exploited as a target for antibody-based therapeutic approaches in acute myeloid leukemia (AML). Validation of this strategy was provided with the approval of the CD33-targeting antibody-drug conjugate (ADC) gemtuzumab ozogamicin in 2000; the clinical utility of this agent has however been hampered by safety concerns.
    [Show full text]
  • 2017 Immuno-Oncology Medicines in Development
    2017 Immuno-Oncology Medicines in Development Adoptive Cell Therapies Drug Name Organization Indication Development Phase ACTR087 + rituximab Unum Therapeutics B-cell lymphoma Phase I (antibody-coupled T-cell receptor Cambridge, MA www.unumrx.com immunotherapy + rituximab) AFP TCR Adaptimmune liver Phase I (T-cell receptor cell therapy) Philadelphia, PA www.adaptimmune.com anti-BCMA CAR-T cell therapy Juno Therapeutics multiple myeloma Phase I Seattle, WA www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 "armored" CAR-T Juno Therapeutics recurrent/relapsed chronic Phase I cell therapy Seattle, WA lymphocytic leukemia (CLL) www.junotherapeutics.com Memorial Sloan Kettering New York, NY anti-CD19 CAR-T cell therapy Intrexon B-cell malignancies Phase I Germantown, MD www.dna.com ZIOPHARM Oncology www.ziopharm.com Boston, MA anti-CD19 CAR-T cell therapy Kite Pharma hematological malignancies Phase I (second generation) Santa Monica, CA www.kitepharma.com National Cancer Institute Bethesda, MD Medicines in Development: Immuno-Oncology 1 Adoptive Cell Therapies Drug Name Organization Indication Development Phase anti-CEA CAR-T therapy Sorrento Therapeutics liver metastases Phase I San Diego, CA www.sorrentotherapeutics.com TNK Therapeutics San Diego, CA anti-PSMA CAR-T cell therapy TNK Therapeutics cancer Phase I San Diego, CA www.sorrentotherapeutics.com Sorrento Therapeutics San Diego, CA ATA520 Atara Biotherapeutics multiple myeloma, Phase I (WT1-specific T lymphocyte South San Francisco, CA plasma cell leukemia www.atarabio.com
    [Show full text]
  • A Deeper Dive Into Advanced and Future Directions in Treating Patients with Acute Myeloid Leukemia
    A Deeper Dive into Advanced and Future Directions in Treating Patients with Acute Myeloid Leukemia Sandra Kurtin, PhDc, ANP-C, AOCN® The University of Arizona Cancer Center Gabrielle Zecha, PA-C The University of Washington Learning Objectives • Describe the latest WHO disease definitions for AML • Recall the mechanism of action for novel therapies for treatment of AML • Summarize the clinical relevance of molecular mutations in AML, including FLT3 and IDH2 • Identify germline mutations in patients with a predisposition for AML Financial Disclosure § Ms. Kurtin served as a consultant for Jazz, Celgene, Novartis, Genentech, and Incyte. § Ms. Zecha has no disclosures to report. This activity is supported by unrestricted educational grants from Astellas Pharma Global Development, Inc. and Jazz Pharmaceuticals. Audience Response Questions Please indicate the clinical role that best represents you: 1. Physician 2. PA 3. Nurse practitioner 4. Clinical nurse specialist 5. Nurse 6. Pharmacist 7. Other Please indicate the practice setting that best represents your practice: 1. Academic medical center, teaching hospital, or comprehensive cancer center 2. Community hospital or community cancer center 3. Private/group practice 4. Government or VA 5. Managed care, insurance, employer, or other payer 6. Pharmaceutical/biotech/device industry 7. Other Please indicate your clinical specialty: 1. Medical oncology 2. Hematology/oncology 3. Radiation oncology 4. Internal medicine 5. Gynecologic oncology 6. Genetics/genetic counseling 7. Other Please indicate your years in practice: 1. < 1 year 2. 1–5 years 3. 6–10 years 4. 11–15 years 5. 16–20 years 6. > 20 years Question #1 Your 74-year-old patient was just started on enasidenib, an oral mIDH2 inhibitor that promotes myeloid differentiation of leukemic blasts.
    [Show full text]
  • Antibody–Drug Conjugates: the Last Decade
    pharmaceuticals Review Antibody–Drug Conjugates: The Last Decade Nicolas Joubert 1,* , Alain Beck 2 , Charles Dumontet 3,4 and Caroline Denevault-Sabourin 1 1 GICC EA7501, Equipe IMT, Université de Tours, UFR des Sciences Pharmaceutiques, 31 Avenue Monge, 37200 Tours, France; [email protected] 2 Institut de Recherche Pierre Fabre, Centre d’Immunologie Pierre Fabre, 5 Avenue Napoléon III, 74160 Saint Julien en Genevois, France; [email protected] 3 Cancer Research Center of Lyon (CRCL), INSERM, 1052/CNRS 5286/UCBL, 69000 Lyon, France; [email protected] 4 Hospices Civils de Lyon, 69000 Lyon, France * Correspondence: [email protected] Received: 17 August 2020; Accepted: 10 September 2020; Published: 14 September 2020 Abstract: An armed antibody (antibody–drug conjugate or ADC) is a vectorized chemotherapy, which results from the grafting of a cytotoxic agent onto a monoclonal antibody via a judiciously constructed spacer arm. ADCs have made considerable progress in 10 years. While in 2009 only gemtuzumab ozogamicin (Mylotarg®) was used clinically, in 2020, 9 Food and Drug Administration (FDA)-approved ADCs are available, and more than 80 others are in active clinical studies. This review will focus on FDA-approved and late-stage ADCs, their limitations including their toxicity and associated resistance mechanisms, as well as new emerging strategies to address these issues and attempt to widen their therapeutic window. Finally, we will discuss their combination with conventional chemotherapy or checkpoint inhibitors, and their design for applications beyond oncology, to make ADCs the magic bullet that Paul Ehrlich dreamed of. Keywords: antibody–drug conjugate; ADC; bioconjugation; linker; payload; cancer; resistance; combination therapies 1.
    [Show full text]
  • Tables-Of-Phase-3-Mabs.Pdf
    Table 3. Monoclonal antibodies in Phase 2/3 or 3 clinical studies for cancer indications Most advanced Primary sponsoring company INN or code name Molecular format Target(s) phase Phase 3 indications Therapeutic area Janssen Research & Development, LLC JNJ-56022473 Humanized mAb CD123 Phase 2/3 Acute myeloid leukemia Cancer Murine IgG1, Actinium Pharmaceuticals Iomab-B radiolabeled CD45 Phase 3 Acute myeloid leukemia Cancer Humanized IgG1, Seattle Genetics Vadastuximab talirine ADC CD33 Phase 3 Acute myeloid leukemia Cancer TG Therapeutics Ublituximab Chimeric IgG1 CD20 Phase 3 Chronic lymphocytic leukemia Cancer Xencor XMAB-5574, MOR208 Humanized IgG1 CD19 Phase 2/3 Diffuse large B-cell lymphoma Cancer Moxetumomab Murine IgG1 dsFv, AstraZeneca/MedImmune LLC pasudotox immunotoxin CD22 Phase 3 Hairy cell leukemia Cancer Humanized scFv, Viventia Bio Oportuzumab monatox immunotoxin EpCAM Phase 3 Bladder cancer Cancer scFv-targeted liposome containing Merrimack Pharmaceuticals MM-302 doxorubicin HER2 Phase 2/3 Breast cancer Cancer MacroGenics Margetuximab Chimeric IgG1 HER2 Phase 3 Breast cancer Cancer Gastric cancer or gastroesophageal junction Gilead Sciences GS-5745 Humanized IgG4 MMP9 Phase 3 adenocarcinoma; ulcerative colitis (Phase 2/3) Cancer; Immune-mediated disorders Depatuxizumab Humanized IgG1, AbbVie mafodotin ADC EGFR Phase 2/3 Glioblastoma Cancer AstraZeneca/MedImmune LLC Tremelimumab Human IgG2 CTLA4 Phase 3 NSCLC, head & neck cancer, bladder cancer Cancer NSCLC, head & neck cancer, bladder cancer, breast AstraZeneca/MedImmune
    [Show full text]
  • Bloomberg Intelligence: ASH Planner
    58th Annual Meeting and Exposition (December 3-6, 2016) Bloomberg Intelligence: ASH Planner Start Time End Time Day Location Room Company (s) Drug (s) Presentation Abstract # Manchester Grand Grand Hall Targeting of JAK/STAT Signaling to Reverse Stroma-Induced Cytoprotection Against BCL2 Antagonist 7:45 AM 08:00AM 12/03/16 AbbVie / Roche Venclexta 32 Hyatt San Diego: D Venetoclax in Acute Myeloid Leukemia San Diego A CD47xCD19 Bispecific Antibody That Remodels the Tumor Microenvironment for Improved Killing and 7:45 AM 08:00AM 12/03/16 Room 5AB Novimmune S.A. NA 44 Convention Center: Provokes a Memory Immune Response to Cancer B Cells San Diego 7:45 AM 08:00AM 12/03/16 Room 6AB Juno JCAR014 CD19 CAR-T Cells Are Highly Effective in Ibrutinib-Refractory Chronic Lymphocytic Leukemia 56 Convention Center: San Diego Implications of Concurrent Ibrutinib Therapy on CAR T-Cell Manufacturing and Phenotype and on Clinical 8:15 AM 08:30AM 12/03/16 Room 6AB AbbVie / J&J Imbruvica 58 Convention Center: Outcomes Following CD19-Targeted CAR T-Cell Administration in Adults with Relapsed/Refractory CLL Bristol-Myers San Diego 8:30 AM 08:45AM 12/03/16 Room 6AB Squibb / Ono / Opdivo / Imbruvica Nivolumab Combined with Ibrutinib for CLL and Richter Transformation: A Phase II Trial 59 Convention Center: AbbVie / J&J Manchester Grand Grand Hall Romiplostim in Patients Undergoing Allogeneic Stem Cell Transplantation: Results of a Phase I/II Multicenter 8:30 AM 08:45AM 12/03/16 Amgen romiplostim 65 Hyatt San Diego: B Trial San Diego AstraZeneca / Acalabrutinib
    [Show full text]
  • Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody–Drug Conjugate, in Relapsed/ Refractory B-Cell Non-Hodgkin Lymphoma Brad S
    Published OnlineFirst November 4, 2019; DOI: 10.1158/1078-0432.CCR-19-0711 Clinical Trials: Targeted Therapy Clinical Cancer Research A Phase I Study of ADCT-402 (Loncastuximab Tesirine), a Novel Pyrrolobenzodiazepine-Based Antibody–Drug Conjugate, in Relapsed/ Refractory B-Cell Non-Hodgkin Lymphoma Brad S. Kahl1, Mehdi Hamadani2, John Radford3, Carmelo Carlo-Stella4, Paolo Caimi5, Erin Reid6, Jay M. Feingold7, Kirit M. Ardeshna8, Melhem Solh9, Leonard T. Heffner10, David Ungar7, Shui He7, Joseph Boni7, Karin Havenith11, and Owen A. O'Connor12 Abstract Purpose: ADCT-402 (loncastuximab tesirine) is an anti- 200 mg/kg. Treatment-emergent adverse events (TEAEs) were body–drug conjugate comprising a CD19-targeting antibody experienced by 87/88 (98.9%) patients. Most common TEAEs and pyrrolobenzodiazepine dimers. A first-in-human study (20% of patients) were hematologic abnormalities, fatigue, evaluated the safety and preliminary clinical activity of lon- edema, liver test abnormalities, nausea, rash, and dyspnea. castuximab tesirine in patients with B-cell non-Hodgkin lym- Grade 3 TEAEs (5% of patients) included hematologic phoma (NHL). abnormalities, liver test abnormalities, fatigue, and dyspnea. Experimental Design: A multicenter, phase I, dose- Overall response rate at doses 120 mg/kg was 59.4% (41 of 69 escalation and dose-expansion study enrolled patients ages patients; 40.6% complete response; 18.8% partial response). 18 years with relapsed/refractory (R/R) B-cell NHL. Patients Median duration of response, progression-free survival, and received loncastuximab tesirine every 3 weeks at doses overall survival (all doses) were 4.8, 5.5, and 11.6 months, assigned by a 3þ3 dose-escalation design.
    [Show full text]
  • OHC Clinical Trials List V4
    FOR MORE INFORMATION OR TO REFER A PATIENT TO ANY OF OUR CLINICAL TRIALS, PLEASE CONTACT NICOLE GIVEN, BS, OHC RESEARCH DEPARTMENT AT 1-800-710-4674 Phase 1 SOLID TUMOR BRE-261: GO29831 (BAM Only) A Phase Ib, Randomized, Two-Arm Study Evaluating the Safety and Phase Ib Pharmacokinetics of MPDL328A (Anti-PD-L1 Antibody) in Combination with Therapeutic Trastuzumab Emtansine or Trastuzumab and Pertuzumab in Patients with MPDL3280A provided HER2-Positive Breast Cancer GI-219: BBI608-246 (pending 1st Qtr) A Phase Ib Clinical Study of BBI608 in Combination with Standard Phase Ib Chemotherapies in Adult Patients with Advanced Gastrointestinal Cancer Therapeutic BBI608 provided REFMAL-381: ABI-007-ST-001 (BAM Only) Phase 1, Open-Label, Multi-Center, Safety Study of Nivolumab (BMS- Phase I 936558) in Combination with Nab-Paclitaxel Plus or Minus Gemcitabine in Therapeutic Pancreatic Cancer, Nab-Paclitaxel/Carboplatin in Stage IIIB/IV Non-Small Nivolumab, Abraxane provided Cell Lung Cancer or Nab-Paclitaxel in Recurrent Metastatic Breast Cancer REFMAL-404: TGR-1202-102 (BAM Only) A Phase I Study Evaluating the Safety and Efficacy of TGR 1202 Alone and in Phase I Combination with Either Nab-Paclitaxel + Gemcitabine or with FOLFOX in Therapeutic Patients with Select Relapsed or Refractory Solid Tumors TGR-1202 provided HEMATOLOGY MM-56: CFZ013 Phase Ib Study of Carfilzomib Administered Once Weekly in Combination Phase Ib with Lenalidomide and Dexamethasone in Subjects with Multiple Myeloma Therapeutic Carfilzomib provided Page 1 of 20 January 11,
    [Show full text]