2021-01-11
At The end of each year, starting in 2010,
The Antibody Society publishes a report that summarizes The progress of new
drugs in The field of antibodies. This is the agency's 12th report. According
to the report, 16 antibody drugs, including antibody conjugates, are under
regulatory review in the United States or the European Union, with a chance of
receiving the world's first regulatory approval in 2021. However, two drugs in
the report were approved by the US FDA in late December 2020, respectively:
Margenza (Margetuximab-cmkb) :
Margetuximab-cmkb, developed by Zai Pharmaceutical Partners MacroGenics, is a
HER2-targeted therapy approved by the FDA in combination with chemotherapy for
adult patients with metastatic HER2-positive breast cancer who have received at
least two anti-HER2 regimens, of which at least one is used for the treatment
of metastatic disease. This drug is a novel Fc domain optimized immunoenhanced
mAb developed using MacroGenics proprietary Fc optimization technology
platform. It has HER2 binding and anti-proliferation effects similar to
trastuzumab, and its engineered Fc domain can enhance the participation of the
immune system. Notably, Margenza is the first HER2-targeted therapy to
significantly improve progression-free survival (PFS) compared to trastuzumab
in a head-to-head phase 3 clinical trial.
-- Ebanga (Ansuvimab-Zykl, MAB114) : Ebanga
(Ansuvimab-Zykl, MAB114), a monoclonal antibody developed by Ridgeback
Biotherapeutics, has been approved by the FDA for use in adult and pediatric
patients, including newborn babies born to mothers who test positive for the
Zaire Ebola virus (Zaire ebolavirus), for the treatment of infections caused by
the Zaire Ebola virus. Ebanga is the second Ebola drug to be approved by the
FDA, following Inmazeb, a cocktail of regenerative antibodies. Ebanga blocks
the binding of the Ebola virus to a cell receptor, preventing the virus from
entering cells.
The following is a review of the remaining
14 antibody drugs in the report, updated and supplemented according to the
information on each company's website. Among them, the first 10 antibody drugs
(tanezumab, narsoplimab, evinacumab, aducanumab, tralokinumab, teplizumab,
inolimomab, bimekizumab, anifrolumab, sutimlimab) are used for non-cancer indications.
The latter four antibody drugs (oportuzumab monatox, dostarlimab, balstilimab,
loncastuximab tesirine) were used for cancer indications.
1, tanezumab
Tanezumab is a humanized IgG2K monoclonal
antibody that works by selectively targeting the blocking of nerve growth
factor (TNF). Tanezumab is being evaluated for the treatment of osteoarthritis
(OA) pain and chronic low back pain (CLBP). Levels of NGF are elevated when the
body is in a state of injury, inflammation, or chronic pain. By selectively blocking
NGF, tanezumab may help prevent pain signals produced by muscles, skin or
organs from reaching the spinal cord and brain. Tanezumab has a novel mechanism
of action that is different from opioids and other pain relievers, including
non-steroidal anti-inflammatory drugs [NSAIDs]. In studies to date, tanezumab
has not been shown to be at risk for addiction, misuse, or dependence.
Tanezumab is made by Pfizer Inc., with
which Lilly signed a $1.8 billion deal in 2013 to co-develop and commercialize
the drug globally. In June 2017, the US FDA granted Tanezumab fast track status
for the treatment of OA pain and CLBP. Notably, Tanezumab is the first NGF
inhibitor to be fast-tracked and has the potential to be the first
first-in-class drug of its kind for the treatment of OA pain and CLBP.
Currently, Tanezumab's biologic license
application (BLA) for the treatment of OA pain is under review by the FDA, with
a December 2020 target date under the Prescription Drug User Fee Act (PDUFA).
But tanezumab has not yet been approved, according to the FDA's new drug
database and the Pfizer/Eli Lilly website. It is also under review by the
European Medicines Agency (EMA).
2, narsoplimab
Narsoplimab is a full-body IgG4λ antibody
targeting mannose-binding lectin associated serine proteinase 2 (MASP-2) for
the treatment of thrombotic microangiopathy (HSCT-TMA) associated with
hematopoietic stem cell transplantation. Narsoplimab is a full-body IgG4λ
antibody targeting mannose-binding lectin associated serine proteinase 2 (MASP-2)
for the treatment of thrombotic microangiopathy (HSCT-TMA) associated with
hematopoietic stem cell transplantation.
Narsoplimab, developed by Omeros, is
licensed as a breakthrough drug for the treatment of high-risk patients with
HSCT-TMA in the United States, for the prevention of complement-mediated
thrombotic microangiopathy (TMA) and for the treatment of HSCT-TMA in the
European Union, and for the treatment of hematopoietic stem cell
transplantation (HSCT) in the European Union. Omeros initiated rolling BLAs for
narsoplimab in patients at high risk for HSCT-TMA to the U.S. FDA in October
2019 and completed those submissions in November 2020. The BLA is currently
under review by the US FDA. The company also plans to submit a marketing
authorization application (MAA) for narsoplimab for the treatment of HSCT-TMA
to the EMA.
3, evinacumab
Evinacumab is a full-body IgG4K antibody
that targets to bind to and block the function of angiopoietin like protein 3
(AngPTL3). AngPTL3 regulates lipid metabolism, including low density
lipoprotein cholesterol (LDL-C), high density lipoprotein cholesterol (HDL-C),
and triglycerides.
Evinacumab was developed from regenerative
cells for the treatment of homozygous familial hypercholesterolemia (HOFH). In
2017, the FDA granted evinacumab breakthrough drug status for the treatment of
hypercholesterolemia in patients with HOFH. Notably, the drug is the first of
its kind to show efficacy in treating patients with HOFH, including those with
little or no LDL receptor function. Evinacumab's BLA for HOFH is currently
under FDA review, with a PDUFA target action date of February 11, 2021. In the
European Union, the EMA awarded evinacumab for accelerated evaluation in June
2020.
4, aducanumab
Aducanumab is an IgG1K antibody that
targets amyloid beta (Aβ) for the treatment of Alzheimer's disease (AD). The
drug was licensed by Bo Jian from Neurimmune Holding AG in 2007 and has been
developed and commercialized globally in cooperation with Eisai since 2017.
Currently, aducanumab is under FDA priority review with a PDUFA target date of
March 7, 2021. In addition, aducanumab is also under review by the European
Union's EMA. The FDA had previously granted aducanumab fast track status. The
EMA has also included the drug in its Priority Medicines (PRIME) programme.
Aducanumab is the first biologic agent to
be submitted to regulatory authorities to target the decline and pathologic
mechanisms of AD-related clinical symptoms. In clinical trials, aducanumab has
been shown to remove Aβ from the brain and slow the clinical decline of AD and
mild cognitive impairment (MCI) due to mild AD dementia. The drug is a highly
anticipated and controversial one. If approved, the drug would be the first
treatment to meaningfully alter the progression of AD and slow its clinical
decline, as well as the first to demonstrate that removal of amyloid beta can
lead to better clinical outcomes.
5, tralokinumab
Tralokinumab is a global IgG4λ antibody
that targets interleukin 13 (IL-13) and is being developed for the treatment of
a variety of inflammatory diseases, including atopic dermatitis. IL-13 is an
important cytokine that plays a key role in driving the underlying inflammation
of atopic dermatitis. By specifically binding IL-13 with a high affinity,
tralokinumab blocks the interaction of IL-13 with its receptor and subsequent
downstream IL-13 signaling.
Tralokinumab is being developed by Leo
Pharma, which acquired it from AstraZeneca in July 2016. Tralokinumab's BLA for
moderate to severe atopic dermatitis is currently under FDA review, with a
PDUFA target date of 2Q2021. In addition, the European EMA accepted the MAA for
tralokinumab in June 2020. If approved, tralokinumab will be the first biologic
agent to challenge Sanofi/Dupixent, which is currently the leading product and
only biologic agent in the treatment of atopic dermatitis (AD). Tralokinumab
was approved in China in June 2020.
6, teplizumab
Teplizumab is a humanized IgG1K antibody
that targets the CD3ε epitome expressed on mature T lymphocytes to modulate the
pathological immune response inherent in type 1 diabetes mellitus (T1D) and
other autoimmune diseases. Teplizumab has been awarded Breakthrough Drug
designation by the U.S. Food and Drug Administration (FDA) and Priority Drug
designation by the European Union's European Medicines Agency (EMA) for the
prevention or delay of clinical T1D in people at high risk of developing T1D,
defined as the presence of two or more T1D-related autoantibodies.
Teplizumab was developed by ProVention Bio,
which was acquired from MacroGenics in 2018. In the United States, ProVention
Bio started rolling BLA submission to the FDA in April 2020 and completed it in
November 2020, and the company has asked the FDA for priority review. If
approved, teplizumab would be the first treatment to prevent/delay progression
to clinical T1D in high-risk individuals.
7, inolimomab
Inolimomab(leukoTAC) is a mouse IgG1K monoclonal
antibody that targets interleukin-2 (IL-2) receptors. IL-2 is a cytokine that
contributes to the development and proliferation of certain white blood cells,
including T cells associated with acute graft-versus-host disease (AGVHD). By
specifically binding to the alpha chain of this receptor (CD25), inolimomab
blocks the surface of IL-2 and the proliferation of donor overactive T cells.
Inolimomab, developed by Elsaly Biotech, a
division of Mediolanum Farmaceutici SpA, is currently under review under the
U.S. Food and Drug Administration's Real-Time Oncology Review Pilot Program
(0RTOR) for the treatment of adult patients with grade II-IV steroid-refractory
acute graft-versus-host disease (SR-AGVHD). The role of inolimomab in SR-AGVHD
is mainly due to its specific and preferred affinity for CD25 receptors on the
surface of T lymphocytes.
8 bimekizumab.
Bimekizumab is a humanized IgG1K antibody
that has a dual mechanism of action. It effectively and selectively neutralizes
IL-17A and IL-17F, two key cytokines that drive the inflammatory process.
IL-17A and IL-17F have similar pro-inflammatory functions and work
independently with other inflammatory mediators to drive chronic inflammation
and damage in multiple tissues. Bimekizumab's unique IL-17A/IL-17F dual
neutralization has the potential to provide a novel targeted therapy for
immune-mediated inflammatory diseases.
Bimekizumab is being evaluated for the
treatment of a variety of inflammatory diseases, including plaque psoriasis,
psoriatic arthritis, ankylost spondylitis, and non-radiological axial arthritis
of the spine. Currently, the BLA and MAA of the drug for the treatment of
moderate to severe plaque psoriasis in adults are under review by the US FDA
and the EU EMA.
9 anifrolumab.
Anifrolumab (Medi-546) is a full-body IgG1K
antibody that specifically binds type I interferon receptor subunit 1 and
blocks the activity of all type I interferons, including IFNα, IFNβ and
IFN-ω-cytokines, which are involved in inflammatory pathways. Three mutations
(L234F, L235E, and P331S) were incorporated into the heavy chain of anifrolumab
to reduce effector function. The drug was developed to treat systemic lupus
erythematosus (SLE), a debilitating autoimmune disease, but only one new
treatment has been approved in the past 60 years, GlaxoSmithKline's Benlysta
(Bilientum, Belicuximab). In China, Benlysta was approved in July 2019 and is
the world's first biologic for the treatment of SLE.
Anifrolumab, developed by AstraZeneca, is
being reviewed by the US FDA and EU EMA for BLA and MAA for the treatment of
adults with moderate to severe activity and positive autoantibodies for SLE,
with approval expected in the second half of 2021. Anifrolumab was previously
granted fast track status by the FDA for the treatment of SLE. It is estimated
that 60-80% of adults with SLE have an elevated type I interferon gene
signature, which has been shown to be associated with disease activity.
Clinical data demonstrated that anifrolumab reduces disease activity in
patients with SLE by targeting type I interferon receptors. If approved,
anifrolumab could bring a new treatment option to the population of patients
with SLE.
10, sutimlimab
Sutimlimab is a humanized IgG4 antibody
developed for the treatment of primary cold agglutinin disease (CAD) hemolysis.
Sutimlimab selectively targets and inhibs the serine protease C1s in the C1
complex of the complement system, the first step in the activation of the
classical complement pathway of the immune system.
Sutimlimab, developed by Sanofi, is
receiving priority review by the U.S. Food and Drug Administration (FDA) for
the treatment of hemolysis in adults with CAD. Sutimlimab has previously been
granted Breakthrough Drug and Orphan Drug status by the FDA, as well as Orphan
Drug status by the European Union's EMA.
11, oportuzumab monatox
Oportuzumab monatox is A local drug
delivery for the next generation of antibody coupling drugs (ADC), is the tumor
cell surface antigen of epithelial cell adhesion molecule (EpCAM) for the
targets of humanized scFv immune toxin, by recombinant humanized EpCAM
antibodies scFv and resistance pseudomonas exotoxin A coupling and become, once
combined with cancer cells express EpCAM will be internalized into the
cytoplasm, inducing cell apoptosis.
The drug was developed by Sesen Bio for the
treatment of high-risk, non-muscular-invasive bladder cancer (NMIBC) that does
not respond to BCG. Preclinical studies have confirmed that EpCAM is
overexpressed in NMIBC cells and almost not expressed in normal bladder cells.
In the United States and the European Union, Oportuzumab monatox was granted
orphan drug status in 2005 and fast track status by the FDA in August 2018 for
the treatment of NMIBC, which is resistant to BCG immunotherapy. In December
2019, Sesen Bio initiated rolling BLA submissions to the FDA, which will be
completed in December 2020, and the company has asked the FDA for priority
review of the BLA.It is worth noting that on November 14, 2020, the FDA issued
a full response letter (CRL) to Sutimlimab's BLA, noting that certain
deficiencies were identified during preapproval inspections (PLI) of
third-party facilities responsible for manufacturing. Before the BLA can be
approved, the deficiencies mentioned in the CRL need to be satisfactorily
resolved by the third party manufacturer. Sanofi is working to address these
issues. A timely resubmission of the BLA will allow Sutimlimab to still have a
chance for approval in 2021. If approved, sutimlimab would be the first and
only drug to treat CAD hemolysis.
12, dostarlimab
DoStarLimab (TS-042, GSK4057190A) is an
anti-PD-1 humanized IgG4K antibody that binds to the PD-1 receptor with high
affinity and effectively blocks its interaction with the ligands PD-L1 and
PD-L1. The drug, an anti-PD-1 tumor immunotherapy, was developed as a result of
AnaptysBio's partnership with Tesaro, which was acquired by GlaxoSmithKline
(GSK) in 2019. In the first quarter of 2020, DoStarlimab's BLA and MAA were
accepted by the US FDA and EU EMA, respectively, for the second-line treatment
of patients with advanced or recurrent mismatched repair defect (DMMR)
endometrial cancer (EC).
Currently, GSK is also evaluating
dostarlimab treatment in early clinical studies all kinds of tumor, at the same
time in phase III RUBY trials evaluate dostarlimab combined standard treatment
(chemotherapy) treatment of recurrent or primary late women with endometrial
cancer (predict to be finished in October 2021), in the study of phase III
FIRST evaluate platinum-based chemotherapy plus dostarlimab + niraparib
combination therapy first-line treatment of stage III or IV non mucous
epithelial ovarian cancer (complete) in February 2023, is expected.
13, balstilimab
Balstilimab is an anti-PD-1 whole human
IgG4K antibody developed by Agenus, Inc., in an anti-PD-1 tumor immunotherapy
category. Several PD-(L)1 therapies, including Keytruda, Opdivo, Tecentriq,
Libtayo, Bavencio and Imfinzi, are currently available for the treatment of
various solid tumors and lymphomas. However, Agenus' clinical studies of
balstilimab have focused on the treatment of cervical cancer as a monotherapy
or in combination with the anti-CTLA4 treatment zalifrelimab (Agen1884). The
FDA has previously granted fast-track approval for balstilimab and
balstilimab/zalifrelimab for the treatment of cervical cancer.
In September 2020, Agenus has initiated the
submission of balstilimab to the U.S. FDA as a monotherapy for BLA in
relapsed/metastatic cervical cancer. It is worth noting that Balstilimab has
exclusive rights in Greater China, Recepta Biopharma has rights in South
America, and Agenus has rights in the rest of the world.
14, loncastuximab tesirine
Loncastuximab tesirine is an antibody-conjugated
drug (ADC) targeting CD19 produced by conjugation of an anti-CD19 humanized
IgG1K antibody via a linker to a pyrrolibenzodiazepine (PBD) dimer toxin. Once
bound to CD19-expressing cells, the drug is internalized by the cell, which
then releases a cytotoxin that irreversibly binds to DNA, creating strong
cross-links that prevent DNA strands from separating. This disrupts essential
DNA metabolic processes such as replication, and ultimately leads to cell
death.
Loncastuximab tesirine, developed by ADC
Therapeutics SA, was previously granted orphan drug designation by the U.S.
Food and Drug Administration for the treatment of diffuse large B-cell lymphoma
(DLBCL) and mantel cell lymphoma (MCL). In September 2020, ADC submitted
Loncastuximab tesirine to the FDA for BLA treatment of relapsed or refractory
DLBCL, which was accepted by the FDA in November, with a PDUFA target date of
May 21, 2021.