Targeting Multiple EGFR Expressing Tumors with a Highly Potent Tumor-Selective Antibody

Total Page:16

File Type:pdf, Size:1020Kb

Targeting Multiple EGFR Expressing Tumors with a Highly Potent Tumor-Selective Antibody Author Manuscript Published OnlineFirst on August 26, 2020; DOI: 10.1158/1535-7163.MCT-20-0149 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Targeting Multiple EGFR Expressing Tumors with a Highly Potent Tumor-Selective Antibody Drug Conjugate Mark G. Anderson1, Hugh D. Falls1, Michael J. Mitten1, Anatol Oleksijew1, Kedar S. Vaidya2, Erwin R. Boghaert1, Wenqing Gao1, Joann P. Palma1, Diana Cao3, Puey-Ling Chia3, Thomas John3, Hui K. Gan3, Andrew M. Scott3 and Edward B. Reilly1 Authors Affiliation 1 AbbVie Inc., 1 North Waukegan Rd., North Chicago, IL 60064 2 Former AbbVie employee 3Olivia Newton-John Cancer Research Institute, and School of Cancer Medicine, La Trobe University Austin Hospital, Heidelberg, Victoria, Australia Running Title Anti-EGFR PBD dimer ADC Correspondence Edward B. Reilly, Ph.D. AbbVie Oncology Discovery, R460 1 North Waukegan Road North Chicago, IL 60064-6099 Phone: (847) 937-0815 Email: [email protected] Authors Disclosure The design, study conduct, and financial support for the study were provided by AbbVie. AbbVie participated in the interpretation of the data, review, and approval of the publication. MGA, HDF, MJM, 1 Downloaded from mct.aacrjournals.org on September 28, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 26, 2020; DOI: 10.1158/1535-7163.MCT-20-0149 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. AO, ERB, WG, JPP and EBR are employees of AbbVie. KSV is a former employee of AbbVie. PLC, DC, TJ, have nothing to disclose, and HKG has been a consultant for AbbVie 2 Downloaded from mct.aacrjournals.org on September 28, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 26, 2020; DOI: 10.1158/1535-7163.MCT-20-0149 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Abstract ABBV-321 (serclutamab talirine), a next-generation epidermal growth factor receptor (EGFR)- targeted antibody-drug conjugate (ADC) incorporates a potent pyrrolobenzodiazepine (PBD) dimer toxin conjugated to the EGFR-targeting ABT-806 affinity matured AM1 antibody. ABBV- 321 follows the development of related EGFR targeted ADCs including depatuxizumab mafodotin (depatux-m, ABT-414), ABT-806 conjugated to monomethyl auristatin F (MMAF), and ABBV-221 (losatuxizumab vedotin), AM1 antibody conjugated to monomethyl auristatin E (MMAE). The distinct tumor selectivity of ABBV-321 differentiates it from many previous highly active antibody PBD conjugates that lack a therapeutic window. Potency of the PBD dimer, combined with increased binding of AM1 to EGFR-positive tumor cells, opens the possibility to target a wide array of tumors beyond those with high levels of EGFR overexpression or amplification, including those insensitive to auristatin-based ADCs. ABBV-321 exhibits potent anti-tumor activity in cellular and in vivo studies including xenograft cell line and patient- derived xenograft (PDX) glioblastoma (GBM), colorectal, lung, head & neck and malignant mesothelioma tumor models which are less sensitive to depatux-m or ABBV-221. Combination studies with ABBV-321 and depatux-m suggest a promising treatment option permitting suboptimal, and potentially better tolerated, doses of both ADCs while providing improved potency. Collectively, these data suggest that ABBV-321 may offer an extended breadth of efficacy relative to other EGFR ADCs while extending utility to multiple EGFR-expressing tumor indications. Despite its highly potent PBD dimer payload, the tumor selectivity of ABBV-321 - coupled with its pharmacology, toxicology and pharmacokinetic profiles - support continuation of ongoing Phase 1 clinical trials in patients with advanced EGFR-expressing malignancies. 3 Downloaded from mct.aacrjournals.org on September 28, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 26, 2020; DOI: 10.1158/1535-7163.MCT-20-0149 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Introduction Targeted therapies are commonly prescribed anti-neoplastic agents in the United States and worldwide. EGFR plays a causal role in the development and maintenance of many human carcinomas. EGFR is a well-validated oncology target, as EGFR-directed therapies [monoclonal antibodies (mAbs) and small molecules] have gained widespread use for a variety of cancer types, including lung, head & neck, colon, and pancreatic cancer. These EGFR-directed therapies have shown improvements in both progression-free survival and overall survival while preserving quality of life (1-8). Despite the enhancements, these approaches are limited by toxic side effects and development of resistance. The most common toxicity of these agents is a well characterized skin rash, similar in appearance to acne, usually limited to the face, upper chest and back. Other reported toxicities include diarrhea, constipation, stomatitis, fatigue, and electrolyte disturbances (9,10). These mechanism based therapies are further limited by narrow efficacy profiles because the mutational status of tumorigenic genes including EGFR, K- Ras, B-Raf, phosphoinositide 3-kinase and the phosphatase and tensin homolog gene each play a role in EGFR resistance mechanisms (11-13). Oncology ADCs are a class of therapeutics that combine mAb specificity with cytotoxicity of potent anti-cancer small molecules. Significant advancements in linker stability and toxin potency are primarily responsible for the resurgence in ADC development. Recent examples of clinically relevant, approved ADCs include brentuximab vedotin/Adcetris®, an anti-CD30 ADC for Hodgkin's lymphoma and anaplastic large cell lymphoma, polatuzumab vedotin/Polivy®, an anti- CD79b ADC targeting the B-cell receptor for diffuse large B-cell lymphoma; trastuzumab emtansine/Kadcyla® , an anti-Her2 ADC for metastatic breast cancer; inotuzumab ozogamicin/Besponsa®, an anti-CD22 ADC for relapsed or refractory B-cell precursor acute lymphoblastic leukemia; gemtuzumab ozogamicin/Mylotarg® an anti-CD33 for acute myeloid leukemia; Trodelvy, an anti-Trop-2 ADC for metastatic TNB, and Enhertu, an anti-Her2 ADC for relapsed/recurrent metastatic Her2+ breast cancer (14-18). A distinct clinical advantage of ADCs is their ability to selectively deliver toxic loads to a tumor, bypassing downstream resistance mechanisms related to intracellular signaling (15,16). 4 Downloaded from mct.aacrjournals.org on September 28, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on August 26, 2020; DOI: 10.1158/1535-7163.MCT-20-0149 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. The ABT-806 antibody is a humanized derivative of the murine anti-EGFR mAb (mAb806) that binds preferentially to tumors expressing EGFR or EGFR with a deletion of exons 2-7 (EGFRvIII); the latter predominantly exposes a unique epitope that is also exposed when EGFR is expressed above physiological levels (19). Depatux-m, an ADC comprised of ABT-806 and MMAF cytotoxin, has been in multiple clinical studies in glioblastoma patients with amplified EGFR although dosing has been limited by corneal side effects common to MMAF conjugates (20-23). The ABT-806 component of depatux-m was affinity matured using recombinant extracellular domain EGFRC271A,C283A (a derivative exposing the ABT-806 epitope) and wild-type EGFR extracellular domain proteins as selection antigens (24). The resulting affinity matured antibody AM1 contains only three amino acid differences in the complementarity-determining region relative to parental ABT-806 but displays increased binding to wild-type EGFR. Despite increased affinity for EGFR, AM1 competes with ABT-806 to bind the same epitope, suggesting that the unique tumor-selective properties of ABT-806 are retained in AM1. ABBV-221, a second-generation ADC, is comprised of AM1 conjugated to a AM1 different toxin, MMAE (25). ABBV-221 demonstrated clinical activity in select patients with advanced solid tumor types (25,26). Results from this study, however, have shown dose-related infusion reactions to be a frequently occurring adverse event, potentially limiting the maximum tolerated dose. PBD dimers, DNA-crosslinking agents, represent an emerging class of warheads with enhanced potent anti-cancer activity compared to the clinically validated auristatins or maytansinoids (27). PBD dimers exhibit significant cell permeability, potentially enabling bystander killing of neighboring tumor cells (28). Multiple ADCs with PBD-dimer payloads and a drug antibody ratio (DAR) of 2 have advanced to the clinic although, at least partially owing to increased toxicities, there have been no approvals for clinical use (29). The highly tumor-selective nature of ABT-806 AM1 suggested that an AM1 PBD dimer conjugate may impart an expanded breadth of efficacy relative to depatux-m or ABBV-221, extending utility beyond tumors with amplified or highly expressed EGFR, while providing for an acceptable therapeutic window. To test this premise, an AM1 PBD dimer conjugate (ABBV-321; serclutamab talirine) was characterized and evaluated against EGFR positive tumor types in cellular and in vivo studies including PDX 5 Downloaded from mct.aacrjournals.org on September 28, 2021. © 2020 American Association for Cancer Research. Author Manuscript
Recommended publications
  • Depatuxizumab Mafodotin (ABT-414)
    Published OnlineFirst May 5, 2020; DOI: 10.1158/1535-7163.MCT-19-0609 MOLECULAR CANCER THERAPEUTICS | CANCER BIOLOGY AND TRANSLATIONAL STUDIES Depatuxizumab Mafodotin (ABT-414)-induced Glioblastoma Cell Death Requires EGFR Overexpression, but not EGFRY1068 Phosphorylation Caroline von Achenbach1, Manuela Silginer1, Vincent Blot2, William A. Weiss3, and Michael Weller1 ABSTRACT ◥ Glioblastomas commonly (40%) exhibit epidermal growth factor Exposure to ABT-414 in vivo eliminated EGFRvIII-expressing receptor EGFR amplification; half of these tumors carry the EGFR- tumor cells, and recurrent tumors were devoid of EGFRvIII vIII deletion variant characterized by an in-frame deletion of exons expression. There is no bystander killing of cells devoid of EGFR 2–7, resulting in constitutive EGFR activation. Although EGFR expression. Surprisingly, either exposure to EGF or to EGFR tyrosine kinase inhibitors had only modest effects in glioblastoma, tyrosin kinase inhibitors reduce EGFR protein levels and are thus novel therapeutic agents targeting amplified EGFR or EGFRvIII not strategies to promote ABT-414–induced cell killing. Further- continue to be developed. more, glioma cells overexpressing kinase-dead EGFR or EGFR- Depatuxizumab mafodotin (ABT-414) is an EGFR-targeting vIII retain binding of mAb 806 and sensitivity to ABT-414, antibody–drug conjugate consisting of the mAb 806 and a toxic allowing to dissociate EGFR phosphorylation from the emer- payload, monomethyl auristatin F. Because glioma cell lines and gence of the “active” EGFR conformation required for ABT-414 patient-derived glioma-initiating cell models expressed too little binding. EGFR in vitro to be ABT-414–sensitive, we generated glioma The combination of EGFR-targeting antibody–drug conju- sublines overexpressing EGFR or EGFRvIII to explore determinants gates with EGFR tyrosine kinase inhibitors carries a high risk of ABT-414–induced cell death.
    [Show full text]
  • Deciphering Molecular Mechanisms and Prioritizing Therapeutic Targets in Cardio-Oncology
    Figure 1. This is a pilot view to explore the potential of EpiGraphDB to inform us about proteins that are linked to the pathophysiology of cancer and cardiovascular disease (CVD). For each cancer type (pink diamonds), we searched for cancer related proteins (light blue circles) that interact with other proteins identified as protein quantitative trait loci (pQTLs) for CVD (red diamonds for pathologies, orange triangles for risk factors). These pQTLs can be acting in cis (solid lines) or trans-acting (dotted lines). Proteins can interact either directly, a protein-protein interaction (dotted blue edges), or through the participation in the same pathway (red parallel lines). Shared pathways are represented with blue hexagons. We also queried which of these proteins are targeted by existing drugs. We found that the cancer drug cetuximab (yellow circle) inhibits EGFR. Other potential drugs are depicted in light brown hexagonal meta-nodes that are detailed below. Deciphering molecular mechanisms and prioritizing therapeutic targets in cardio-oncology Pau Erola1,2, Benjamin Elsworth1,2, Yi Liu2, Valeriia Haberland2 and Tom R Gaunt1,2,3 1 CRUK Integrative Cancer Epidemiology Programme; 2 MRC Integrative Epidemiology Unit, University of Bristol; 3 The Alan Turing Institute Cancer and cardiovascular disease (CVD) make by far the immense What is EpiGraphDB? contribution to the totality of human disease burden, and although mortality EpiGraphDB is an analytical platform and graph database that aims to is declining the number of those living with the disease shows little address the necessity of innovative and scalable approaches to harness evidence of change (Bhatnagar et al., Heart, 2016).
    [Show full text]
  • Seattle Genetics and Genmab Enter Into New Antibody-Drug Conjugate Collaboration
    Seattle Genetics and Genmab Enter Into New Antibody-Drug Conjugate Collaboration Company Announcement • Additional collaboration combines Genmab’s proprietary antibodies and Seattle Genetics’ ADC technology • New ADC program will target AXL expressed on multiple tumor types Bothell, WA and Copenhagen, Denmark; September 10, 2014 – Seattle Genetics, Inc. (Nasdaq: SGEN) and Genmab A/S (OMX: GEN) today announced that the companies have entered into an additional antibody-drug conjugate (ADC) collaboration. Under the new agreement, Genmab will pay an upfront fee of $11 million for exclusive rights to utilize Seattle Genetics’ auristatin-based ADC technology with Genmab’s HuMax®-AXL, an antibody targeting AXL which is expressed on multiple types of solid cancers. Seattle Genetics is also entitled to receive more than $200 million in potential milestone payments and mid-to-high single digit royalties on worldwide net sales of any resulting products. In addition, prior to Genmab’s initiation of a Phase III study for any resulting products, Seattle Genetics has the right to exercise an option to increase the royalties to double digits in exchange for a reduction of the milestone payments owed by Genmab. Irrespective of any exercise of option, Genmab remains in full control of development and commercialization. “This collaboration with Genmab further extends the reach of our industry-leading ADC technology for use with novel oncology targets, while providing us with a compelling financial value proposition as the program advances,” said Natasha
    [Show full text]
  • Advances in Epidermal Growth Factor Receptor Specific Immunotherapy: Lessons to Be Learned from Armed Antibodies
    www.oncotarget.com Oncotarget, 2020, Vol. 11, (No. 38), pp: 3531-3557 Review Advances in epidermal growth factor receptor specific immunotherapy: lessons to be learned from armed antibodies Fleury Augustin Nsole Biteghe1,*, Neelakshi Mungra2,*, Nyangone Ekome Toung Chalomie4, Jean De La Croix Ndong5, Jean Engohang-Ndong6, Guillaume Vignaux7, Eden Padayachee8, Krupa Naran2,* and Stefan Barth2,3,* 1Department of Radiation Oncology and Biomedical Sciences, Cedars-Sinai Medical, Los Angeles, CA, USA 2Medical Biotechnology & Immunotherapy Research Unit, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa 3South African Research Chair in Cancer Biotechnology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa 4Sun Yat-Sen University, Zhongshan Medical School, Guangzhou, China 5Department of Orthopedic Surgery, New York University School of Medicine, New York, NY, USA 6Department of Biological Sciences, Kent State University at Tuscarawas, New Philadelphia, OH, USA 7Arctic Slope Regional Corporation Federal, Beltsville, MD, USA 8Department of Physiology, University of Kentucky, Lexington, KY, USA *These authors contributed equally to this work Correspondence to: Stefan Barth, email: [email protected] Keywords: epidermal growth factor receptor (EGFR); recombinant immunotoxins (ITs); targeted human cytolytic fusion proteins (hCFPs); recombinant antibody-drug conjugates (rADCs); recombinant antibody photoimmunoconjugates (rAPCs) Received: May 30, 2020 Accepted: August 11, 2020 Published: September 22, 2020 Copyright: © 2020 Biteghe et al. This is an open access article distributed under the terms of the Creative Commons Attribution License (CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
    [Show full text]
  • Homogeneous Antibody-Drug Conjugates Via Site-Selective Disulfide Bridging
    Homogeneous antibody-drug conjugates via site-selective disulfide bridging Nafsika Forte, Vijay Chudasama, and James R. Baker* Department of Chemistry, University College London, London, UK; email: [email protected] Antibody-drug conjugates (ADCs) constructed using site-selective labelling methodologies are likely to dominate the next generation of these targeted therapeutics. To this end, disulfide bridging has emerged as a leading strategy as it allows the production of highly homogeneous ADCs without the need for antibody engineering. It consists of targeting reduced interchain disulfide bonds with reagents which reconnect the resultant pairs of cysteine residues, whilst simultaneously attaching drugs. The 3 main reagent classes which have been exemplified for the construction of ADCs by disulfide bridging will be discussed in this review; bissulfones, next generation maleimides and pyridazinediones, along with others in development. Introduction In the past decades, the potential of monoclonal antibodies (mAbs) in targeted therapy against cancer has been realized, with the market of therapeutic antibodies currently being the fastest growing sector in the pharmaceutical industry. While unmodified antibodies have demonstrated unprecedented activities in the clinic, their action can also be synergistically improved by arming them with cytotoxic drugs to create a new class of targeted therapy, i.e. antibody-drug conjugates (ADCs).1,2 This strategy has great potential to dramatically reduce the side-effects observed with standard chemotherapy,
    [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]
  • 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]
  • Targeting and Efficacy of Novel Mab806-Antibody-Drug Conjugates in Malignant Mesothelioma
    pharmaceuticals Article Targeting and Efficacy of Novel mAb806-Antibody-Drug Conjugates in Malignant Mesothelioma 1,2,3, 1,3,4, 5 5 Puey-Ling Chia y, Sagun Parakh y, Ming-Sound Tsao , Nhu-An Pham , Hui K. Gan 1,2,3,4, Diana Cao 1, Ingrid J. G. Burvenich 1,2 , Angela Rigopoulos 1, Edward B. Reilly 6, Thomas John 1,2,3,4,* and Andrew M. Scott 1,2,4,7,* 1 Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Melbourne, Victoria 3084, Australia; [email protected] (P.-L.C.); [email protected] (S.P.); [email protected] (H.K.G.); [email protected] (D.C.); [email protected] (I.J.G.B.); [email protected] (A.R.) 2 Faculty of Medicine, University of Melbourne, Melbourne, Victoria 3010, Australia 3 Department of Medical Oncology, Austin Health, Melbourne, Victoria 3084, Australia 4 School of Cancer Medicine, La Trobe University, Plenty Rd &, Kingsbury Dr, Bundoora, Victoria 3086, Australia 5 Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 2C1, Canada; [email protected] (M.-S.T.); [email protected] (N.-A.P.) 6 AbbVie Inc., North Chicago, IL 60064, USA; [email protected] 7 Department of Molecular Imaging and Therapy, Austin Health, Melbourne, Victoria 3084, Australia * Correspondence: [email protected] (T.J.); [email protected] (A.M.S.); Tel.: +61-39496-5876 (A.M.S.); Fax: +61-39496-5334 (A.M.S.) These authors contributed equally to this work. y Received: 7 September 2020; Accepted: 28 September 2020; Published: 2 October 2020 Abstract: Epidermal growth factor receptor (EGFR) is highly overexpressed in malignant mesothelioma (MM).
    [Show full text]
  • Antibodies for the Treatment of Brain Metastases, a Dream Or a Reality?
    pharmaceutics Review Antibodies for the Treatment of Brain Metastases, a Dream or a Reality? Marco Cavaco, Diana Gaspar, Miguel ARB Castanho * and Vera Neves * Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Av. Prof. Egas Moniz, 1649-028 Lisboa, Portugal * Correspondence: [email protected] (M.A.R.B.C.); [email protected] (V.N.) Received: 19 November 2019; Accepted: 28 December 2019; Published: 13 January 2020 Abstract: The incidence of brain metastases (BM) in cancer patients is increasing. After diagnosis, overall survival (OS) is poor, elicited by the lack of an effective treatment. Monoclonal antibody (mAb)-based therapy has achieved remarkable success in treating both hematologic and non-central-nervous system (CNS) tumors due to their inherent targeting specificity. However, the use of mAbs in the treatment of CNS tumors is restricted by the blood–brain barrier (BBB) that hinders the delivery of either small-molecules drugs (sMDs) or therapeutic proteins (TPs). To overcome this limitation, active research is focused on the development of strategies to deliver TPs and increase their concentration in the brain. Yet, their molecular weight and hydrophilic nature turn this task into a challenge. The use of BBB peptide shuttles is an elegant strategy. They explore either receptor-mediated transcytosis (RMT) or adsorptive-mediated transcytosis (AMT) to cross the BBB. The latter is preferable since it avoids enzymatic degradation, receptor saturation, and competition with natural receptor substrates, which reduces adverse events. Therefore, the combination of mAbs properties (e.g., selectivity and long half-life) with BBB peptide shuttles (e.g., BBB translocation and delivery into the brain) turns the therapeutic conjugate in a valid approach to safely overcome the BBB and efficiently eliminate metastatic brain cells.
    [Show full text]
  • Adcetris, INN-Brentuximab Vedotin
    19 July 2012 EMA/702390/2012 Committee for Medicinal Products for Human Use (CHMP) Assessment report Adcetris International non-proprietary name: brentuximab vedotin Procedure No. EMEA/H/C/002455 Note Assessment report as adopted by the CHMP with all information of a commercially confidential nature deleted. 7 Westferry Circus ● Canary Wharf ● London E14 4HB ● United Kingdom Telephone +44 (0)20 7418 8400 Facsimile +44 (0)20 7523 7455 E -mail [email protected] Website www.ema.europa.eu An agency of the European Union Product information Name of the medicinal product: Adcetris Applicant: Takeda Global Research and Development Centre (Europe) Ltd. 61 Aldwych London WC2B 4AE United Kingdom Active substance: brentuximab vedotin International Nonproprietary Name/Common Name: brentuximab vedotin Pharmaco-therapeutic group Monoclonal antibodies (ATC Code): (L01XC12) ADCETRIS is indicated for the treatment of adult Therapeutic indication(s): patients with relapsed or refractory CD30+ Hodgkin lymphoma (HL): 1. following autologous stem cell transplant (ASCT) or 2. following at least two prior therapies when ASCT or multi-agent chemotherpay are not a treatment option ADCETRIS is indicated for the treatment of adult patients with relapsed or refractory systemic anaplastic large cell lymphoma (sALCL). Pharmaceutical form(s): Powder for concentrate for solution for infusion Strength(s): 50 mg Route(s) of administration: Intravenous use Packaging: vial (glass) Package size(s): 1 vial Adcetris CHMP assessment report Page 2/102 Rev10.11 Table of contents 1. Background information on the procedure .............................................. 9 1.1. Submission of the dossier ...................................................................................... 9 1.2. Steps taken for the assessment of the product ....................................................... 10 2. Scientific discussion .............................................................................
    [Show full text]
  • Marine-Derived Anticancer Agents: Clinical Benefits, Innovative
    marine drugs Review Marine-Derived Anticancer Agents: Clinical Benefits, Innovative Mechanisms, and New Targets Renato B. Pereira 1 , Nikolai M. Evdokimov 2, Florence Lefranc 3, Patrícia Valentão 1 , Alexander Kornienko 4, David M. Pereira 1 , Paula B. Andrade 1,* and Nelson G. M. Gomes 1,* 1 REQUIMTE/LAQV, Laboratório de Farmacognosia, Departamento de Química, Faculdade de Farmácia, Universidade do Porto, R. Jorge Viterbo Ferreira, n◦ 228, 4050-313 Porto, Portugal; [email protected] (R.B.P.); valentao@ff.up.pt (P.V.); dpereira@ff.up.pt (D.M.P.) 2 Department of Chemistry and Biochemistry, University of California, Santa Barbara, CA 93106, USA; [email protected] 3 Department of Neurosurgery, Hôpital Erasme, Université Libre de Bruxelles, 808 Route de Lennik, 1070 Brussels, Belgium; fl[email protected] 4 Department of Chemistry and Biochemistry, Texas State University, San Marcos, TX 78666, USA; [email protected] * Correspondence: pandrade@ff.up.pt (P.B.A.); ngomes@ff.up.pt (N.G.M.G.); Tel.: +351-22-042-8654 (P.B.A.); +351-122-042-8500 (N.G.M.G.) Received: 15 May 2019; Accepted: 30 May 2019; Published: 2 June 2019 Abstract: The role of the marine environment in the development of anticancer drugs has been widely reviewed, particularly in recent years. However, the innovation in terms of clinical benefits has not been duly emphasized, although there are important breakthroughs associated with the use of marine-derived anticancer agents that have altered the current paradigm in chemotherapy. In addition, the discovery and development of marine drugs has been extremely rewarding with significant scientific gains, such as the discovery of new anticancer mechanisms of action as well as novel molecular targets.
    [Show full text]
  • Peptide-Drug Conjugate for Her2-Targeted Drug Delivery Yan Wang University of the Pacific, [email protected]
    University of the Pacific Scholarly Commons University of the Pacific Theses and Dissertations Graduate School 2018 Peptide-drug conjugate for Her2-targeted drug delivery Yan Wang University of the Pacific, [email protected] Follow this and additional works at: https://scholarlycommons.pacific.edu/uop_etds Part of the Biochemistry, Biophysics, and Structural Biology Commons, and the Pharmacy and Pharmaceutical Sciences Commons Recommended Citation Wang, Yan. (2018). Peptide-drug conjugate for Her2-targeted drug delivery. University of the Pacific, Thesis. https://scholarlycommons.pacific.edu/uop_etds/3567 This Thesis is brought to you for free and open access by the Graduate School at Scholarly Commons. It has been accepted for inclusion in University of the Pacific Theses and Dissertations by an authorized administrator of Scholarly Commons. For more information, please contact [email protected]. 1 PEPTIDE-DRUG CONJUGATE FOR HER2-TARGETED DRUG DELIVERY by Yan Wang A Thesis Submitted to the Graduate School In Partial Fulfillment of the Requirements for the Degree of MASTER OF SCIENCE Thomas J. Long School of Pharmacy and Health Sciences Pharmaceutical and Chemical Sciences University of the Pacific Stockton, California 2018 2 PEPTIDE-DRUG CONJUGATE FOR HER2-TARGETED DRUG DELIVERY by Yan Wang APPROVED BY: Thesis Advisor: Xiaoling Li, Ph.D. Thesis Co-Advisor: Bhaskara R. Jasti, Ph.D. Committee Member: Liang Xue, Ph.D. Dean of Graduate School: Thomas Naehr, Ph.D 3 PEPTIDE-DRUG CONJUGATE FOR HER2-TARGETED DRUG DELIVERY Copyright 2018 by Yan Wang 4 ACKNOWLEDGEMENTS I would first like to thank my thesis advisor Dr. Xiaoling Li of the Thomas J. Long School of Pharmacy and Health Sciences at the University of Pacific.
    [Show full text]