The Antibody Society

the official website of the antibody society

An international non-profit supporting antibody-related research and development.

  • LOG IN
  • BECOME A MEMBER
  • About
    • Mission & Activities
    • Directors and Officers
    • Marketing & Promotions
    • The Antibody Society’s Committees
      • Meetings Committee
      • AIRR Community Working Groups & Sub-Committees
    • Sponsors & Partners
  • Society meetings
    • Computational Antibody Discovery: State of the Art
      • Computational Antibody Discovery Symposium Participants
    • Harnessing Cytokines for Cancer Immunotherapy Symposium
    • Biopharmaceutical Informatics Symposium
    • Emerging Cancer Therapies Leveraging Gamma-Delta Effector T cells Symposium
    • Emerging Immunotherapeutics for Ovarian Cancer Symposium
    • AIRR Community Meetings
    • Antibody Engineering & Therapeutics (US) 2024
      • 2022 Antibody Engineering & Therapeutics
      • 2020 Antibody Engineering & Therapeutics
      • 2019 Antibody Engineering & Therapeutics
      • 2018 Antibody Engineering & Therapeutics
      • What is INN a Name?
        • INN issue updates
    • Antibody Engineering & Therapeutics Europe 10 – 12 June, 2025 | Congress Center, Basel Switzerland.
      • Scientific Advisors, Antibody Engineering & Therapeutics Europe
    • FOCIS Symposia
  • AIRR Community
    • AIRR Community News
    • AIRR Community Newsletter
    • AIRR Community Seminar Series
    • AIRR Community Meetings
      • Zooming into the Community III
      • AIRR Community Meeting VII – Learnings and Perspectives
      • AIRR Community Special Event 2023  – Zooming in to the Community II
      • AIRR Community Meeting VI: “Exploring New Frontiers”
      • AIRR Community Meeting V: “Zooming in to the AIRR Community”
      • AIRR Community Meeting V Pre-Meetings
        • AIRR-seq in the Pandemic
        • AIRR-seq Biological Standards and Workflows
      • AIRR Community Special Event: “Response to COVID-19”
      • AIRR Community Meeting IV: “Bridging the Gaps”
      • AIRR Community Meeting III
        • Day 1
        • Day 2
        • Day 3
        • Day 4
      • AIRR Community Meeting II
      • AIRR Community Meeting I
    • On AIRR – An AIRR Community Podcast
    • AIRR Data Commons
    • AIRR-C Germline Database Resources
    • AIRR Community Publications
    • AIRR Community Working Groups
      • Biological Resources Working Group
      • Common Repository Working Group
      • Diagnostics Working Group
      • Germline Database Working Group
      • Legal and Ethics Working Group
      • Software Working Group
      • Standards Working Group
    • AIRR Community Sub-Committees
      • Communications Sub-Committee
      • Executive Sub-Committee
      • Inferred Allele Review Committee
      • Meetings Sub-Committee
      • Strategic Planning Sub-Committee
    • AIRR Community Webinar Series
    • AIRR Community Calendar
    • AIRR Community Resources
  • Members only
    • Login
    • Note to members
    • Member discount codes
    • 2025 Calendar of Events
    • James S. Huston Antibody Science Talent Award
      • 2024 James S. Huston Antibody Science Talent Award Recipient
      • 2023 James S. Huston Antibody Science Talent Award Recipient
      • 2022 James S. Huston Antibody Science Talent Award Recipient
      • 2021 James S. Huston Antibody Science Talent Award Recipient
      • 2020 James S. Huston Antibody Science Talent Award Recipient
      • Huston Award submission guidelines
    • Research Competitions
      • Research Competition Winners
    • Science Writing Competition
      • Science Writing Competition Winners
    • Imaging Competition
      • Imaging Calendar Competition winners
        • The Antibody Society 2025 Calendar
        • The Antibody Society 2024 Calendar
    • Antibodies in early-stage studies
    • Presentations
  • Upcoming meetings in 2025
  • Web Resources
    • Society Publications
    • Antibody News
    • Antibody News Podcast
    • Antibody therapeutics approved or in regulatory review in the EU or US
      • Antibody therapeutics product data
    • Antibodies in late-stage clinical studies
    • Research Resources
    • Education Resources
  • Career Center
    • Career Shorts
  • Learning Center
    • Upcoming Webinars in 2025
    • The Antibody Series Lectures
    • Antibody Discovery & Development
    • Adaptive Immune Receptor Repertoires
    • Antibodies to Watch
    • Commercializing Antibody Therapeutics
    • Antibody Validation
      • 4th International Antibody Validation Meeting, Sep 2023
    • Snakebite antivenoms: Global challenges and progress toward recombinant antibody therapeutics
You are here: Home / Archives for Phase 3 pipeline

Is R&D of antibody therapeutics for non-cancer diseases in decline?

April 23, 2018 by Janice Reichert

Although cancer is often the focus of attention, antibody-based drugs are developed and approved for many other indications, such as immune-mediated, neurological, ophthalmic and skeletal disorders, as well as cardiovascular/hemostasis, respiratory and infectious diseases. Antibody therapeutics for diseases other than cancer comprise slightly over half (58%) of all  antibody products granted their first approval in either the US or European Union (EU), and they comprise approximately half (48%) of the late-stage commercial pipeline. [1]

The number of first approvals of antibodies for non-cancer diseases is expected to be especially high in 2018, with 3 already approved in either the US or EU (burosomab, ibalizumab, tildrakizumab) and another 7 that may be approved by the end of the year. Burosumab (burosumab-twza; Crysvita), which targets fibroblast growth factor 23, was approved in the EU and US in February and April 2018, respectively, for X-linked hypophosphatemia. The anti-CD4 product ibalizumab-uiyk (Trogarzo) was first approved in the US in March 2018 for treatment of patients with multi-drug resistant HIV infection. Tildrakizumab-asmn (Ilumya), which targets interleukin-23p19, was approved in the US in March for treatment of moderate-to-severe plaque psoriasis. Antibodies for non-cancer indications that may be approved by the end of the year include three for the prevention of migraine (erenumab, fremanezumab, galcanezumab), two for cardiovascular/hemostasis indications (caplacizumab for the treatment of acquired thrombotic thrombocytopenic purpura; lanadelumab for prevention of hereditary angioedema attacks) and one (emapalumab) for treatment of  primary hemophagocytic lymphohistiocytosis, which is a clinical syndrome of hyperinflammation that is lethal if untreated. In addition, romosozumab, which targets sclerostin, is in review in the EU and US as a treatment for osteoporosis, but the US Food and Drug Administration has requested additional clinical data from Phase 3 studies.

Despite the success of antibodies for non-cancer diseases, the percentage of these molecules entering first-in-human studies has recently declined [Figure 1].

Whereas during 2010-2014 antibodies for non-cancer diseases comprised 46-60% of all antibodies entering clinical study each year, they have comprised a declining percentage in all subsequent years (44%, 37% and 22% in 2015, 2016 and 2017, respectively). It must be noted that there was a substantial increase in the total number of antibody therapeutics entering clinical studies during the 2015-17 (ave. 106/year) compared to 2010-2014 (ave. 64/year). Nevertheless, the number of antibodies for non-cancer diseases that entered studies in 2017 was the lowest (so far) in this decade. One reason for this decline may be the current focus of research on antibodies that modulate immune checkpoints or redirect T cells and on immunoconjugates such as antibody-drug conjugates, which are almost exclusively developed as treatments for cancer. While the number of antibodies for non-cancer diseases in Phase 2 studies (~130) is likely sufficient to replenish the number in Phase 3 studies and regulatory review in the short term,  early-stage studies of more will be needed to sustain the flow of these therapeutics onto the market well into the future.

[1] Kaplon H, Reichert JM. Antibodies to watch in 2018. MAbs. 2018 Feb/Mar;10(2):183-203.

Like this post but not a member? Please join! Membership is free for students, post-docs and employees of the Society’s corporate sponsors.

Filed Under: Antibody therapeutics pipeline, Phase 3 pipeline, Regulatory review Tagged With: antibody therapeutics, approved antibodies, clinical pipeline

Update on antibody therapeutics in late-stage clinical studies

February 27, 2017 by Janice Reichert

Five monoclonal antibody (mAb) therapeutics have recently transitioned into late-stage clinical studies. Three (utomilumab, isatuximab, SHR-1210) are being evaluated as treatments for cancer, and the effects of two (crizanlizumab, olokizumab) are being studied in patients with other disorders.  Utomilumab (PF-05082566) is a human IgG2 antibody agonist that targets the extracellular domain of 4-1BB (CD137), which is a co-stimulatory receptor expressed on activated T cells. In preclinical studies, Fisher et al (1) demonstrated that utomilumab can activate NF-κB and induce downstream cytokine production, promote leukocyte proliferation, and inhibit tumor growth in a xenograft tumor model. Utomilumab is included in a multi-center, international, randomized, open label, 2-component (Phase 1b followed by Phase 3), parallel-arm study (Javelin DLBCL; NCT02951156) of avelumab in combination with various agents for the treatment of relapsed/refractory diffuse large B-cell lymphoma. The study includes a total of 5 arms (A: avelumab/utomilumab/rituximab; B: avelumab/utomilumab/azacitidine; C: avelumab/rituximab/bendamustine; D: selected regimen from Phase 1b component, which may be the agents investigated in study arms A or B or C; E: investigator’s choice of either rituximab/bendamustine or rituximab/gemcitabine/oxaliplatin). Progression-free survival is the primary outcome measure of the Phase 3 component of the study, which has an estimated primary completion date of February 2021.

Isatuximab (SAR650984), a humanized anti-CD38 IgG1 mAb, is undergoing evaluation in a Phase 3 randomized, open-label, multicenter study (ICARIA-MM; NCT02990338) comparing the mAb in combination with pomalidomide and low-dose dexamethasone vs. pomalidomide and low-dose dexamethasone in patients with refractory or relapsed and refractory multiple myeloma. Jiang et al (2) demonstrated that isatuximab utilizes multiple mechanisms of action in mediating multiple myeloma cell cytotoxicity, and that pomalidomide augments this cytotoxicity. Progression-free survival is the primary outcome measure of the Phase 3 study, which has an estimated primary completion date of May 2018.

SHR-1210 is a humanized IgG4 antibody targeting programmed death-1. A randomized controlled multi-center Phase 2/3 study (NCT02989922) to evaluate SHR-1210 in patients with advanced hepatocellular carcinoma who failed or intolerable to prior systemic treatment is currently recruiting patients. The study will assess whether SHR-1210 treatment improves objective response rate and overall survival compared with the standard of care. The estimated primary completion date of the study is December 2018.

Crizanlizumab (SEG101, SelG1) is a humanized IgG2 P-selectin (CD62) inhibitor undergoing evaluation for prevention or reduction of the occurrence of pain crises in patients with sickle cell disease. Results from the Phase 2 SUSTAIN study (NCT01895361) showed that crizanlizumab administered at 5.0 mg/kg intravenously 14 times over a period of 52 weeks reduced the median annual rate of sickle cell-related pain crises by 45.3% compared to placebo (1.63 vs 2.98, p=0.010) (3). The median time to the first crisis was significantly longer in patients who received crizanlizumab compared with placebo (4.07 vs. 1.38 months, P=0.001), as was the median time to the second crisis (10.32 vs. 5.09 months, P=0.02). Crizanlizumab is listed as in Phase 3, with a planned filing date of 2020, in the Novartis Annual Report 2016.

Olokizumab, an anti-interleukin-6 humanized IgG4 mAb (4), is undergoing evaluation in three Phase 3 studies evaluating two doses of olokizumab (64 mg subcutaneous every 2 or 4 weeks) in patients with rheumatoid arthritis (RA). The primary outcome measures are the ACR20 response at Week 14. The Phase 3 NCT02760368 study (CREDO 1) will determine how safe and effective olokizumab is compared to placebo in RA patients who are already receiving, but not fully responding to, treatment with methotrexate. The study’s estimated primary completion date is May 2018. In study NCT02760407 (CREDO 2), the effectiveness and safety of olokizumab is being compared to placebo and adalimumab in RA patients who are taking methotrexate but have active disease. The study’s estimated primary completion date is January 2019. In study NCT02760433 (CREDO 3), the effects of olokizumab are being compared to placebo in patients with RA who are already receiving, but not fully responding to treatment with a tumor necrosis factor inhibitor (CREDO 3). The study’s estimated primary completion date is February 2019. Olokizumab, formerly known as CDP-6038, is under development by R-Pharm under a worldwide exclusive license from UCB Pharma S.A.

The Antibody Society maintains comprehensive tables that list mAbs in late-stage clinical studies (i.e., pivotal Phase 2, Phase 2/3 or Phase 3) for cancer and non-cancer indications. These tables are updated versions of Tables 3 and 4 in the article ‘Antibodies to watch in 2017’.  The tables will be updated with new information about antibody therapeutics that enter late-stage studies as the information is made available during the year. Information in bold was added during 2017. Antibody therapeutics that transition from late-stage studies to regulatory review are listed with the ‘Approved antibodies’ in the Members Only section of The Antibody Society’s website.

Please login using your username and password to view the content in the Members Only section.

Not a member? Please join!

Membership is free for employees of the Society’s corporate sponsors.

1.       Fisher TS, et al. Targeting of 4-1BB by monoclonal antibody PF-05082566 enhances T-cell function and promotes anti-tumor activity. Cancer Immunol Immunother. 2012; 61(10):1721-33.

2.       Jiang H, et al. SAR650984 directly induces multiple myeloma cell death via lysosomal-associated and apoptotic pathways, which is further enhanced by pomalidomide. Leukemia. 2016; 30(2):399-408.

3.       Ataga KI, et al. Crizanlizumab for the prevention of pain crises in sickle cell disease. N Engl J Med. 2017; 376(5):429-439.

4.       Shaw S, et al. Discovery and characterization of olokizumab: a humanized antibody targeting interleukin-6 and neutralizing gp130-signaling. MAbs. 2014; 6(3):774-82.

Filed Under: Ab news, Phase 3 pipeline

Antibody-drug conjugates in the spotlight

October 14, 2016 by Janice Reichert

square logo ADCAntibody-drug conjugates (ADCs) are designed to deliver cytotoxic agents into targeted cells, and they are typically developed as treatments for cancer. Due to the need for new cancer drugs, the development of ADCs is the focus of substantial efforts by the biopharmaceutical industry. Nearly 60 ADCs are currently in clinical studies, one ADC, inotuzumab ozogamicin, is undergoing regulatory review, and three ADCs have been granted approvals, although the first to be approved, gemtuzumab ozogamicin (Mylotarg®) was withdrawn from the market in 2010.

Two ADCs, brentuximab vedotin (Adcetris®) and ado-trastuzumab emtansine (Kadcyla®), are currently marketed in the United States (US) and European Union (EU), as well as other countries. These two ADCs are disparate in their composition, and are used as treatments for different indications. Brentuximab vedotin is composed of an anti-CD30 monoclonal antibody (mAb) conjugated to the tubulin inhibitor monomethyl auristatin E via a valine-citruline dipeptide linkage designed for conditional cleavage inside cells. In contrast, ado-trastuzimab emtansine comprises an anti-human epidermal growth factor receptor-2 (HER2) mAb coupled to the tubulin-disrupting maytansinoid DM1 drug via a non-reducible thioether linkage. Brentuximab vedotin was granted its first approval in 2011 for two indications: 1) classical Hodgkin lymphoma after failure of autologous hematopoietic stem cell transplantation or after failure of at least two prior multi-agent chemotherapy regimens in patients who are not auto-HSCT candidates; and 2) systemic anaplastic large cell lymphoma after failure of at least one prior multi-agent chemotherapy regimen. The first approval for ado-trastuzumab emtansine was granted in 2013; the product is indicated for the treatment of HER2-positive metastatic breast cancer in patients who previously received trastuzumab and a taxane separately or in combination.

Of the nearly 60 ADCs in the clinic, only two (depatuxizumab mafodotin, vadastuximab talirane) are currently in late-stage (Phase 2/3 or 3) clinical studies, but two additional ADCs (sacituzumab govitecan, mirvetuximab soravtansine) may transition to Phase 3 soon. Depatuxizumab mafodotin (ABT-414) is composed of an anti-epidermal growth factor receptor (EGFR) mAb conjugated to the tubulin inhibitor monomethyl auristatin F via a stable maleimidocaproyl linker. The Phase 2b/3 Intellance 1 study (NCT02573324) of the ADC with concurrent chemoradiation and adjuvant temozolomide in adult patients with newly diagnosed glioblastoma multiforme (GBM) with EGFR amplification was initiated in late 2015. Depatuxizumab mafodotin has orphan drug designations for GBM in the US and glioma in the EU, and it was granted a US Rare Pediatric Disease Designation for pediatric EGFR-amplified diffuse intrinsic pontine glioma, a brainstem tumor that is highly aggressive and difficult to treat. Vadastuximab talirane (SGN-33A) is an anti-CD33 mAb with 2 engineered cysteine residues through which DNA cross-linking pyrrolobenzodiazepine dimer drug moieties are conjugated via a protease-cleavable valine-alanine dipeptide linker. The Phase 3 CASCADE clinical trial (NCT02785900) of vadastuximab talirine in combination with azacitidine (Vidaza) or decitabine (Dacogen) in older patients with newly diagnosed acute myeloid leukemia (AML) was initiated in May 2016.  Results from a Phase 1 study indicated that the ADC in combination with hypomethylating agents was a well-tolerated regimen with a high remission rate in older patients with AML.

The transitions of sacituzumab govitecan (IMMU-132) and mirvetuximab soravtansine (IMGN853) to Phase 3 may occur by the end of 2016. The start of a Phase 3 study (NCT02574455) that will evaluate the safety and efficacy of sacituzumab govitecan in refractory/relapsed triple-negative breast cancer (TNBC) patients is scheduled for December 2016. This ADC has received US Breakthrough Therapy and Fast Track designations for the treatment of patients with TNBC. Sacituzumab govitecan comprises an anti-TROP-2 mAb conjugated via a pH-sensitive linker to SN-38, the active metabolite of the chemotherapeutic irinotecan, in a site-specific manner. Mirvetuximab soravtansine is being assessed as a single-agent therapy in the FORWARD I trial (NCT02631876) of the ADC versus investigator’s choice of chemotherapy in adults with folate receptor (FR)-α positive advanced epithelial ovarian cancer, primary peritoneal cancer or primary fallopian tube cancer, which is being changed from a Phase 2 to a Phase 3 trial. Mirvetuximab soravtansine is composed of an anti-FRα mAb linked to the tubulin-disrupting maytansinoid DM4 via a cleavable linker.

It should be noted that, despite the increased complexity of the molecules, ADCs are also the focus of companies developing biosimilar products. As discussed in previous Society posts, biosimilars of antibody-based drugs that have lost patent protection, including adalimumab (Humira®), rituximab (Rituxan®, Mabthera®), trastuzumab (Herceptin®) and etanercept (Enbrel®), are already approved or undergoing regulatory review in the US and EU, as well as other countries.

The Antibody Society thanks Hanson Wade for access to Beacon, the World ADC database.

Like this post? Please become a member!

Filed Under: ADC, Clinical pipeline, Phase 3 pipeline Tagged With: Antibody drug conjugates, antibody therapeutics

Antibodies to watch in 2016: Mid-year update

August 18, 2016 by Janice Reichert

mabs-coverSince 2010, the “Antibodies to watch” article series has documented annually the number and identities of commercially sponsored antibody therapeutics in Phase 3 studies, regulatory review and those recently approved in the US and EU. Taken together, the articles have captured the extraordinary doubling of the number of antibody therapeutics in Phase 3 studies from 26 to 53, as identified in the “Antibodies to watch in 2010” and “Antibodies to watch in 2016” articles, respectively. Due to the highly dynamic nature of antibody therapeutics development, numerous transitions have occurred during 2016, and the Society offers here a mid-year update to data reported in the “Antibodies to watch in 2016” article.

As described in our previous posts, 4 antibody therapeutics (atezolizumab, reslizumab, ixekizumab, obiltoxaximab) were granted first marketing authorizations in either the US or EU during January to June 2016. As of mid-2016, marketing applications for 8 antibody therapeutics are being considered for first approvals in the US or EU. Of these, 5 applications (olaratuzumab, bezlotoxumab, sarilumab, brodalumab, ocrelizumab) have Food and Drug Administration action dates during September -December 2016. Recommendations by the European Medicines Agency on applications for Xilonix and inotuzumab ozogamicin could be made in 2016, but additional time would be needed for the European Commission’s decision regarding whether to grant the marketing authorization. It thus remains to be seen whether the number of antibody therapeutics approved in the US or EU during 2016 will match or exceed the record of 9 approvals granted in a single year set in 2015.

As of mid-2016, 53 unique antibody therapeutics were in Phase 3 studies. This is the same total number noted in the “Antibodies to watch in 2016” article, but the antibodies included in the totals are not all the same. The tables included in this mid-year update result from the addition of antibodies that started a first Phase 3 study in late 2015 to mid-2016, and deletion of antibodies that transitioned to regulatory review, reverted to an earlier clinical phase or had their development suspended or terminated. Compared to the totals included in the “Antibodies to watch in 2016” article, the number of antibodies in Phase 3 studies for cancer indications as of mid-2016 decreased slightly (from 17 to 15, respectively), while those for non-cancer indications increased slightly (from 36 to 38, respectively).

Antibodies for cancer represent only 28% of the current commercial Phase 3 pipeline, although they are ~55% of the overall clinical pipeline of therapeutic antibodies. The 15 antibody therapeutics in Phase 3 studies for cancer indications are notable for the diversity in their composition. Of the 15, 6 (40%) are non-canonical antibodies (1 radiolabeled antibody, 1 scFv-containing liposome, 2 immunotoxins, 2 antibody-drug conjugates (ADCs)), and a majority of the canonical antibodies (i.e., full-length IgG1, 2 or 4) are Fc- or glyco-engineered to enhance functionality. The 2 ADCs now in Phase 3 studies represent a vanguard, as this type of antibody therapeutic has entered clinical studies in large numbers only recently. Of the ADCs currently in clinical studies, most (44/56, 79%) are in either Phase 1 or Phase 1/2 studies, and most (55/56) are for cancer indications. ADCs now comprise ~20% of the clinical pipeline of antibodies for cancer, but ~11% of all antibodies in clinical development. There is substantial diversity of the targets, drugs, linkers, and drug-to-antibody ratios of the ADCs in the clinic. For example, of the ADCs in the clinic, targets for 51 have been disclosed, and 39 of these 51 targets are unique, i.e., only one ADC in clinical studies is known to target that particular antigen. Antigens known to be the target of more than one ADC in clinical studies include CD19, CD37, EGFR, HER2 and mesothelin. The diversity of the molecules may initially serve as a hindrance, but knowledge gained by the development of this class of molecules should increase overall as more ADCs enter clinical studies, transition through the phases and join the two ADCs currently on the market, brentuximab vedotin (Adcetris®) and ado-trastuzumab vedotin (Kadcyla®).

Antibodies for non-cancer indications dominate the current commercial Phase 3 pipeline. Unlike the antibodies for cancer, the 38 antibodies in Phase 3 studies for non-cancer indications are mostly canonical full-length IgG1, 2 or 4 molecules. Only 4 of the 38 (~11%) are non-canonical molecules: 1 bispecific antibody and 3 antibody ‘fragments’ (scFv, Fab, nanobody). Like ADCs, bispecific antibodies are expected to comprise a larger percentage of the Phase 3 pipeline in the next ~6-8 years. Bispecific antibodies now comprise ~9% of the entire commercial pipeline of antibody therapeutics, but most (32/45, 71%) of those are currently in early clinical studies (either Phase 1 or Phase 1/2). Compared to ADCs, bispecific antibodies are undergoing evaluation in a broader range of indications, although the majority of bispecifics (30/45, 67%) are for cancer and they comprise ~11% of the clinical pipeline of antibodies for cancer. The two bispecific antibodies now on the market, catumaxomab (Removab®) and blinatumomab (BLINCYTO®), are both for cancer. Nevertheless, the one bispecific antibody now in Phase 3 studies, emicizumab, is for a non-cancer indication (hemophilia A).

The clinical pipeline of antibody therapeutics, including at Phase 3, is highly dynamic. The Antibody Society will continue to track antibodies in the clinic, and report progress to its members.

Acknowledgements: The Antibody Society thanks Hanson Wade for access to the Beacon ADC database.

Like this post? Please become a member!

Filed Under: Clinical pipeline, Development metrics, European Medicines Agency, Food and Drug Administration, Phase 3 pipeline Tagged With: ADC, Antibody drug conjugates, antibody therapeutics, bispecific, clinical pipeline

New antibody therapeutics for multiple sclerosis

June 3, 2016 by Janice Reichert

Antibody impressionOn May 27, 2016 the Food and Drug Administration (FDA) approved daclizumab (Zinbryta®) for the treatment of adults with relapsing forms of multiple sclerosis (MS). The product, which targets interleukin-2 receptor alpha chain (CD25), is manufactured using a high-yield process. Daclizumab (Zenapax®) was first approved in 1997 for the prevention of organ transplant rejection. While the two products have the same amino acid sequence, Zinbryta has a different glycosylation pattern and reduced antibody-dependent cytotoxicity compared to Zenapax. Zinbryta’s approval was based in part on the results of the Phase 3 DECIDE study (NCT01064401) of Zinbryta versus interferon β 1a (Avonex®) in patients with relapsing-remitting MS. In this study, patients administered Zinbryta experienced fewer clinical relapses than those who received Avonex (annualized relapse rate 0.22 vs. 0.39; 45% lower rate with Zinbryta; P<0.001). Due to serious safety risks, FDA has included a boxed warning on the product label, and it is available only through a restricted distribution program under a Risk Evaluation and Mitigation Strategy.

Another antibody therapeutic, ocrelizumab, has been evaluated in a Phase 3 study of patients with primary progressive multiple sclerosis (PPMS). The antibody targets CD20 on B cells, which are implicated in the inflammatory and neurodegenerative processes of MS. Ocrelizumab was granted FDA’s Breakthrough Therapy Designation for PPMS, which is a debilitating form of MS characterized by steadily worsening symptoms. PPMS patients typically do not experience distinct relapses or periods of remission. Currently, no treatments are approved for treatment of the disease. In the pivotal Phase 3 ORATORIO study, treatment with ocrelizumab significantly reduced disability progression and other markers of disease activity compared with placebo. Ocrelizumab is also undergoing evaluation in Phase 3 studies of patients with relapsing-remitting forms of the disease. Genentech plans to submit a marketing application for ocrelizumab as a treatment for MS in 2016. If an application is submitted to FDA by the end of June and receives a priority review, which is a benefit of the Breakthrough Therapy Designation, then ocrelizumab could be approved for marketing in the US by the end of 2016.

Filed Under: Approvals, Phase 3 pipeline, Uncategorized Tagged With: antibody therapeutics, daclizumab, Food and Drug Administration, multiple sclerosis, ocrelizumab

Next Page »

mabs

mabs

The Official Journal of The Antibody Society

Career Center

Our Career Center is a premier resource to connect highly qualified talent with matching career opportunities. Visit for details on over 800 jobs!

AIRR Community

AIRR Community

The Adaptive Immune Receptor Repertoire Community is a research-driven group organizing around the use of high-throughput sequencing technologies to study antibody/B-cell and T-cell receptor repertoires.

Recent Posts

  • Zooming into the Community III Starts Tomorrow! May 20, 2025
  • Exciting news – The AIRR Community is turning 10! 🎂 May 8, 2025
  • The Antibody Society (TAbS): Win a FREE Attendance Pass to AET Basel & Present A Poster: Call For Abstracts! March 26, 2025

Archives

Follow us online

  • Email
  • LinkedIn
  • Twitter
  • YouTube
  • Home
  • Privacy & Terms of Use
  • About
  • Directors and Officers
  • Advisors
  • Sponsors & Partners
  • Mission & Activities
  • Join the Society
  • Membership Levels
  • Members only
  • Login
  • Antibody therapeutics approved or in regulatory review in the EU or US
  • Meeting reports
  • Presentations
  • Contact

©2015 - scicomvisuals