INTRODUCTION
Development of bispecific antibodies (BsAbs) as therapeutic agents has recently attracted
significant attention, and investments in this modality have been steadily increasing.
This review discusses challenges, and suggestions to overcome them, associated with
the development of BsAbs, specifically those pertaining to clinical pharmacology,
pharmacometrics, and bioanalysis. These challenges and possible solutions are discussed
by presenting several case studies of BsAbs that have gained regulatory approval or
that are currently in clinical development.
BsAbs, also termed “dual‐targeting” or “dual‐specificity” antibodies, have the ability
to bind two different targets on the same or different cell(s); the targets may be
cell‐surface receptors or soluble ligands, as shown in Figure
1. These dual‐nature antibodies have key advantages that can potentially enhance therapeutic
efficacy compared with monotherapy or traditional combination therapies by: i) simultaneously
blocking two different targets or mediators that have a primary role in the disease
pathogenesis; ii) inducing cell signaling pathways (e.g., proliferation or inflammation);
iii) retargeting to mediate antibody‐dependent cell‐mediated cytotoxicity (ADCC);
iv) avoiding the development of resistance and increasing antiproliferative effects,
specifically in oncology; and v) temporarily engaging a patient's own cytotoxic T
cells to target cancer cells, thus activating cytotoxic T cells to cause tumor lysis
(e.g., bispecific T‐cell engagers (BiTE)).
Figure 1
Various designs for BsAb molecules (a) Dimers inhibition: BsAbs can bind to two receptors/targets
(HER2/HER3, HER2/HER4) on the same cell (e.g., MM‐111); (b) Dual inhibition: BsAbs
can inhibit two different cytokines simultaneously, for example, COVA322 that inhibits
TNF‐α and IL17A; (c) Triomabs: The antigen binding site binds to target cell receptors
(EpCAM, HER2, or CD20) and the T‐cell receptors (CD3). The heavy chain site binds
to NK cells or dendritic cells or macrophages/phagosome (e.g., catumaxomab, ertumaxomab,
FBTA05); (d) Two‐ligand inactivation: two arms bind to different ligands on different
cells belonging to the same population, such as DLL4 x VEGF, TNF‐α x IL17A, IL4 x
IL13 (e.g., OMP‐305B83, COVA322, SAR156597); (e) Transmembrane/transcytosis: The BsAbs
are designed specifically to cross the barriers/membrane via receptor transport (transferrin
receptor) and bind to enzymes/receptors (BACE1) on the other side; (f) BiTE antibody
construct: These are designed to bridge T cells and target cells by binding to CD3/CD28
or CD19/CD20/CD22/CEA/EpCAM, respectively (e.g., blinatumomab, MEDI‐565, MT110). The
examples mentioned above can be found in Table
2 for further information. BACE1, β‐secretase 1; BiTE, bispecific T‐cell engagers;
BsAbs, bispecific antibodies; DDL4, delta‐like ligand 4; EpCAM, epithelial cell adhesion
molecule; HER, human epidermal growth factor receptor; IL, interleukin; NK, Natural
Killer; TNF‐α, tumor necrosis factor‐alpha; VEGF, vascular endothelial growth factor.
Traditional combination therapies using monoclonal antibodies (mAbs) can also modulate
multiple therapeutic targets. However, the development of mAbs presents challenges
not encountered with BsAbs. For example, regulatory agencies have established stringent
criteria for the codevelopment of new drugs that are intended for use as combination
therapies. The sponsor has to demonstrate i) the rationale for use of the combination
therapy rather than individual treatments; ii) a strong justification for why the
individual drugs cannot be studied and developed independently; iii) that the nonclinical
and clinical studies provide adequate evidence showing that the combination therapy
provides significant therapeutic gain; and iv) a reasonable toxicity profile and more
durable response than the monotherapy and existing standard of care.1 These guidelines
can potentially make the drug development process for combination therapy lengthy
and expensive. Conversely, BsAbs are able to address the biology associated with two
different targets simultaneously via a similar regulatory pathway as that required
for with a single‐target mAb. BsAbs may therefore offer the opportunity to benefit
patients more quickly, and to access less costly development routes than can be afforded
via classic combination therapies.
BsAbs also offer the opportunity to modulate unexplored biology in novel ways that
may not be possible with single‐target mAbs. Avidity is defined as the measure of
the overall strength of binding of an antigen with multiple antigenic determinants
to multivalent antibodies. According to the “avidity hypothesis,” BsAbs may surpass
combination therapy in terms of both biology and mechanistic behavior as a result
of this theoretical concept. The theory states that avidity increases when two receptors
are bound to a target cell, leading to efficacy greater than which could be expected
from the additive combination each single mAb. A specific example is the development
of JNJ‐61186372 (BsAb targeting epidermal growth factor receptor (EGFR) and c‐Met),
which showed that the BsAb was more potent than the combination of single receptor‐binding
antibodies.2 Furthermore, BsAbs are less likely than combination treatment to undergo
off‐target binding in the presence of a surplus of decoy cells.3 BsAbs therefore have
the theoretic potential to improve therapeutic window (safety and efficacy), selectivity,
and regulatory efficiency as compared with a true combination therapy approach.
As a result of the aforementioned advantages of BsAbs (Table
1), BsAbs are one of the fastest growing classes of investigational drugs. In addition
to the approved BsAbs, blinatumomab (BLINCYTO, Amgen, Thousand Oaks, CA) and catumaxomab
(Removab, Fresenius Biotech, Homburg, Germany, initially marketed by Fresenius Biotech)
for cancer immunotherapy, there are more than 50 additional BsAbs in clinical development4,
5, 6, 7, 8, 9, 10 (Table
2), with the potential for sales of up to $4.4 billion by 2023.11 Of note, almost
all BsAbs that are currently in development target indications in oncology, with the
following exceptions: COVA322 for plaque psoriasis, BsAbs targeting transferrin receptor
(TfR) and β‐secretase 1 (BACE1) for central nervous system disorders, ABT981 for osteoarthritis,
ALX‐0761 for psoriasis, AMG 570 for systemic lupus erythematosus, and JNJ‐61178104
and MDG010 for autoimmune diseases.12 In this review we aimed at increasing awareness
of the multiple facets of translational and clinical development of BsAbs. We are
presenting clinical pharmacology considerations (Table
3) and modeling simulation strategies with select examples as well as bioanalytical
challenges and strategies, opportunities, and approaches supported by a variety of
case studies. However, published information on some of the aspects discussed here
is limited, thus restricting a broader selection of case studies and underscoring
the need for a more extensive application of modeling simulation approaches to support
efficient drug development.
Table 1
Comparison of therapeutic modalities
Properties
Small molecules
Peptides
mAbs
Antibody‐Drug Conjugate (ADC)
BsAb
Molecular weight
<1 kDa
<10 kDa (or <50 amino acids)
Few kDa to 150 kDa
Few kDa to 1,000 kDa
Few kDa to 1,000 kDa
Route of administration
PO, IV, SC, IM
PO (limited), IV, SC or IM
IV, SC or IM
IV, SC or IM
IV, SC or IM
PK
Linear at low doses; nonlinear at high doses
Linear at low doses; nonlinear at high doses
Mostly nonlinear at low doses. The linear PK is from FcR mediated clearance and nonlinear
arises from TMDD. Linear at high doses
Nonlinear at low doses and linear at high doses.
May or may not be linear, dictated by the presence of Fc domain. Absence of Fc domain
can lead to linear PK.
Distribution
Passive diffusion
Passive diffusion and convective transport
Convective transport
Convective transport
Convective transport
Metabolism
CYP's
Proteolytic degradation
Proteolytic degradation
Proteolytic degradation and CYPs
Proteolytic degradation
Serum T1/2
Varies based on physicochemical properties
Short (<10 min) but can be increased with modifications
Usually long and depends on target mediated clearance and FcRn mediated antibody recycling
Usually long
Varies from h to days
Renal clearance
May or may not be a major route
Possible (if peptides are resistant to proteolysis)
Major route if Mol wt is <69 kDa
Possible route for both mAb and cytotoxic agent
Possible, very low to negligible
Hepatic clearance
May or may not be a major route
Not a major route with few exceptions
Target or site of action (liver or pancreas) dependent.
Route for cytotoxic agent
Unlikely
Target mediated clearance
No
Undergoes TMDD
May undergo TMDD depending on the target
Undergoes TMDD
Can possibly undergo TMDD for individual target
Intestinal clearance
Possible route driven by transporters and enzymes (transferases)
Not applicable
Not applicable
Not applicable
Not applicable
Typical dosing regimen
QD, BID or TID
Daily to weekly
Varying dosage and dosing regimens throughout the length of treatment. Regimen typically
ranges from weekly to monthly to 6 months.
Weekly to monthly cycles
Weekly to monthly cycles
Toxicity
Mediated by Structure, physicochemical properties, metabolites, dose, and off‐target.
Limited
Immune‐mediated adverse events and immunotoxicity such as immunosuppression, immunostimulation,
hypersensitivity and auto‐immunity
Immune‐mediated adverse events and immunotoxicity from antibody. Small molecules related
toxicities form conjugated drug.
Immune‐mediated adverse events and immunotoxicity such as immunosuppression, immunostimulation,
hypersensitivity and autoimmunity
Immunogenicity
Very rare
Low
May have non, low, or high ADA, neutralizing ADA can affect CL of mAbs
Very high, neutralizing ADA can affect CL of mAbs
Very likely, unless one of the targets is B‐cell.
DDI
High likelihood
Low
Implicit DDI resulting from the changes in immune system influencing the CYPs activities
Very low
Implicit short term DDI resulting from the changes in immune system influencing the
CYPs activities
John Wiley & Sons, Ltd.
Table 2
List of BsAbs currently undergoing clinical trials. (Source: clinicaltrials.gov)
BsAb
Sponsor
Format
Target
Biological function
Clinical trial
Identifier
Conditions
Allogeneic cytomegalovirus‐specific cytotoxic T lymphocytes
Fred Hutchinson Cancer Research Center
Allogenic Hematopoietic stem cells (HSCT)
CD8 X CD19
CAR‐T Cells
Completed phase I/II
NCT01475058
Precursor Acute Lymphoblastic Leukemia, Lymphoma
AMG 330
Amgen
BiTE antibody construct
CD3 X CD33
T cell recruitment
Ongoing phase I (On hold)
NCT02520427
Myeloid Leukemia, Acute non lymphoblastic leukemia
Anti CD3 X anti‐CD20 BsAb‐armed activated T cells
Barbara Ann Karmanos Cancer Institute
Activated T‐cells armed with BsAb
CD3 X CD20
Activated T cells
Completed phase I
NCT00938626
Multiple Myeloma and Plasma Cell Neoplasm
Anti‐CEA x anti‐DTPA and di‐DTPA‐131I peptide
Nantes University Hospital
scFv‐IgG
CEA X di‐DTPA‐131I
Radioimmunotherapy
Completed phase II
NCT00467506
Thyroid Neoplasms
anti‐EpCAM x anti‐CD3 (removab)
AGO Study Group
Triomab
EpCAM X CD3
T cell recruitment
Completed phase II
NCT00189345
Ovarian Cancer
BAY2010112
Bayer
BiTE antibody construct
CD3 X PSMA
T cell recruitment
Enrolling phase I
NCT01723475
Prostatic Neoplasms
BI 836909 (AMG 420)
Boehringer Ingelheim
BiTE antibody construct
B‐cell maturation antigen (BCMA)
T cell recruitment
Ongoing phase I (recruiting participants)
NCT02514239
Multiple myeloma
Blinatumomab
Amgen
BiTE antibody construct
CD3 X CD19
T cell recruitment
Ongoing phase III
NCT02393859
Leukemia, Acute Lymphoblastic
National Cancer Institute (NCI)
Not yet open (phase I)
NCT02568553
B‐Cell Lymphoma (Unclassifiable with intermediate features)
Ongoing (phase II)
NCT02143414
B Acute Lymphoblastic Leukemia, Untreated Adult Acute Lymphoblastic Leukemia
Ongoing (phase III)
NCT02101853
B Acute Lymphoblastic Leukemia, Recurrent Adult/childhood Acute Lymphoblastic Leukemia
Ongoing (phase III)
NCT02003222
Adult B Acute Lymphoblastic Leukemia, Untreated Adult Acute Lymphoblastic Leukemia
Amgen Research (Munich)
Ongoing phase I/II
NCT01471782
Acute Lymphoblastic Leukemia
Ongoing
NCT01741792
Diffuse Large B‐cell Lymphoma
Ongoing phase II
NCT01466179
Acute Lymphoblastic Leukemia
Ongoing phase II
NCT01209286
B‐ALL
Ongoing phase II
NCT01207388
B‐cell Acute Lymphoblastic Leukemia
Completed
NCT00560794
Acute Lymphoblastic Leukemia
Completed phase I
NCT00274742
Non‐Hodgkin's Lymphoma, Relapsed
Catumaxomab
Neovii Biotech
Triomab
EpCAM X CD3
T cell recruitment
Completed phase II
NCT00464893
Gastric Cancer, Gastric Adenocarcinoma
JSehouli
Completed phase II
NCT01815528
Recurrent Epithelial Ovarian Cancer
AIO‐Studien‐gGmbH
Ongoing phase II
NCT01504256
Gastric Adenocarcinoma With Peritoneal Carcinomatosis, Siewert Type II/III Adenocarcinoma
of Esophagogastric Junction With Peritoneal Carcinomatosis
Grupo Español de Investigación en Cáncer de Ovario
phase II
NCT01246440
Ovarian Cancer
Neovii Biotech
Completed phase II
NCT01065246
Malignant Ascites Due to Epithelial Carcinoma
Completed phase III
NCT00822809
Cancer, Neoplasms, Carcinoma, Malignant Ascites
Completed phase II
NCT00377429
Ovarian Cancer
Completed phase II
NCT00326885
Malignant Ascites
Completed phase II/III
NCT00836654
EpCam Positive Tumor (e.g.Ovarian, Gastric, Colon, Breast), Malignant Ascites
Completed phase II
NCT00352833
Gastric Cancer, Gastric Adenocarcinoma
CD20Bi‐activated T cells (ATC)
Barbara Ann Karmanos Cancer Institute
Activated T‐cells armed with BsAb
CD3 X CD20
Activated T cells
Completed phase I
NCT00244946
Lymphoma
DT2219ARL
Masonic Cancer Center, University of Minnesota
2 scFv linked to diphtheria toxin
CD19 X CD22
Targeting of protein toxin to tumor
Ongoing phase I
NCT02370160
Refractory and relapsed B‐Lineage Leukemia/Lymphoma
Ongoing phase I
NCT00889408
Leukemia/Lymphoma
EGFRBi‐Armed Autologous T Cells
Barbara Ann Karmanos Cancer Institute
T cells armed with BsAb
CD3 X EGFR
Autologous activated T cells to EGFR‐positive tumor
Withdrawn (phase I/II)
NCT02521090
Adult Brain Glioblastoma, Adult Gliosarcoma, Recurrent Brain Neoplasm
GD2Bi‐aATC
Barbara Ann Karmanos Cancer Institute
Activated T‐cells armed with BsAb
CD3 X GD2
Activated T cells
phase I/II
NCT02173093
Desmoplastic Small Round Cell Tumor, Disseminated Neuroblastoma, Metastatic Childhood
Soft Tissue Sarcoma, etc
HER2Bi‐aATC
Barbara Ann Karmanos Cancer Institute
Activated T‐cells armed with BsAb
CD3 X HER2
Activated T cells
phase I
NCT02470559
Malignant Ovarian Clear Cell Tumor, Malignant Ovarian Serous Tumor, Recurrent Fallopian
Tube Carcinoma, Recurrent Ovarian Carcinoma, Recurrent Primary Peritoneal Carcinoma
Enrolling phase I
NCT02662348
Esophageal, Gastric, Pancreatic, Liver, Gallbladder, Bowel Cancer
IMCgp100
Immunocore Ltd
ImmTAC
CD3 X gp100
T cell recruitment
Ongoing phase I
NCT01211262
Malignant Melanoma
phase I (Recruitment has not begun)
NCT02570308
Uveal Melanoma
Indium labeled IMP‐205xm734
Radboud University
Radioimmunotherapy
Unknown (phase I)
NCT00185081
Colonic Neoplasms
JNJ‐61186372
Janssen Research & Development, LLC
Bispecific human IgG1 mAbs
EGFR X cMET
Inhibits receptor phosphorylation
Ongoing phase I (Recruitment has not started yet)
NCT02609776
non‐small cell lung cancer (NSCLC)
LY3164530
Eli Lilly and Company
OrthoFab‐IgG
MET X EGFR
Blockade of 2 receptors
Ongoing phase I
NCT02221882
Neoplasms, Neoplasm Metastasis
MDX447
Dartmouth‐Hitchcock Medical Center
2 (Fab') crosslinked
CD64 x EGFR
Active monocytes to kill tumor
Completed phase I,
NCT00005813
Brain and Central Nervous System Tumors
MEDI‐565
MedImmune LLC
BiTE antibody construct
CEA X CD3
T cell recruitment
Completed phase I
NCT01284231
Gastrointestinal Adenocarcinomas
MGD006
MacroGenics
Dual Affinity Re‐Targeting (DART)
CD123 x CD3
Re‐targeting T cells to tumors
Ongoing phase I
NCT02152956
AML
MGD007
MacroGenics
DART
gpA33 X CD3
Re‐targeting T cells to tumors
Ongoing phase I
NCT02248805
Colorectal Carcinoma
MGD010
MacroGenics
Dual Affinity Re‐Targeting (DART)
CD32B x CD79B
Safety assessment
Ongoing phase I
NCT02376036
Healthy Subjects
Mitoxantrone packaged EDV (EnGeneIC Delivery Vehicle)
Dr David Ziegler
Delivery of nanoparticles
phase I (Recruitment has not begun)
NCT02687386
Solid Tumor and CNS Tumor
MM‐111
Merrimack Pharmaceuticals
HSA body
HER2 X HER4
Blockade of 2 receptors
Completed phase I
NCT00911898
Her2 Amplified Solid Tumors, Metastatic Breast Cancer
MM‐111 + Herceptin
Merrimack Pharmaceuticals
HSA body
HER2 X HER3
Blockade of 2 receptors
Completed phase I
NCT01097460
Breast Neoplasms
MOR209/ES414
Emergent Product Development Seattle LLC
scFv‐IgG
PSMA X CD3
T cell recruitment
Ongoing phase I
NCT02262910
Prostate Cancer
MT 110
Amgen Research (Munich) GmbH
BiTE antibody construct
CD3 X EpCAM
T‐cell recruitment
Completed phase I
NCT00635596
Solid Tumors
OMP‐305B83
OncoMed Pharmaceuticals, Inc.
DVD‐Ig
DLL4 X VEGF
2‐ligand inactivation
Ongoing phase I
NCT02298387
Advanced Solid Tumor Malignancies
REGN1979
Regeneron Pharmaceuticals
CD20 X CD3
T cell recruitment
Ongoing phase I
NCT02290951
Non‐Hodgkin's Lymphoma, Chronic Lymphocytic Leukemia
rM28
University Hospital Tuebingen
Tandem scFv
CD28 X HMV‐MAA
Retargeting autologous lymphocytes to tumor
Completed phase I/II
NCT00204594
Malignant Melanoma
RO6958688
Hoffmann‐La Roche
Crossmab
CEA X CD3
T cell recruitment
Ongoing phase I
NCT02324257
Solid Cancers
TargomiRs
University of Sydney
EGFR X EDV
Delivery of nanoparticles
Ongoing phase I
NCT02369198
Malignant Pleural Mesothelioma, Non‐Small Cell Lung Cancer
TF2
Radboud University
Dock and lock
CEA x HSG
Radioimmunotherapy
Completed phase I
NCT00860860
Colorectal Neoplasms
Garden State Cancer Center at the Center for Molecular Medicine and Immunology
enzyme‐linked immunosorbent assay, pharmacological study
phase I
NCT00895323
Colorectal Cancer
Nantes University Hospital
Immuno‐PET
Ongoing phase I/II
NCT01730638
Medullary Thyroid Carcinoma
Centre René Gauducheau
Radioimmunotherapy
Ongoing (phase I/II)
NCT01221675
Small Cell Lung Cancer, CEA‐expressing Non‐Small Cell Lung Carcinoma (NSCLC)
TF2 ‐ 68 Ga‐IMP‐288
Nantes University Hospital
Dock and lock
CEA X HSG
Immuno‐PET
Ongoing phase I/II
NCT01730612
HER2 Negative Breast Carcinoma Expressing CEA
TF2 antibody/68Ga‐IMP‐288
Nantes University Hospital
Dock and lock
CEA X HSG
Radioimmunotherapy
Not yet open (phase II)
NCT02587247
Metastatic Colorectal Cancer
John Wiley & Sons, Ltd.
Table 3
Opportunities for translational and clinical pharmacology in drug development for
BsAbs
Functional areas
Question
Possible approaches
Bioanalytical
What are the key fundamental points in selecting a bioanalytical strategy and optimizing
the assay for BsAb?
The key rationale is around what to measure for interrogating the exposure response
relationship (or safety) of the BsAb. If bifunctional form assay is not available,
subsequent assay risk assessment should be considered. There is a timing aspect as
well. The approaches are then dictated often by the nature of the BsAb which then
can steer towards an LBA as appropriate or LC‐MS for example. The method optimization
in terms of what are measured (free, partially bound or total) may be required along
with program progression during development.
Preclinical and translational
What are the considerations for the receptor occupancy calculation applied in dose
determination for BsAbs?
It is dictated by the affinity and avidity of the BsAbs with the target and any prior
information from the mAbs agent.
What is the basis of selecting the dose and dosing regimen for the FIH study?
The doses are selected from the dose range finding studies in cynomolgus monkeys using
MABEL approach. PKPD modeling is performed with preclinical studies and in vitro cytotoxicity
data to project the FIH doses.
What are the considerations for the PD end points that can influence the optimal dose
and dosing regimen decision?
The time points when samples are collected to measure PD.
The source of the samples (e.g. surrogate tissue or diseased tissue) in which the
PD is measured.
Measuring PD that is specific to each side of a bispecific molecule and understanding
how each one relates to the overall purpose of the PD measure and intended use.
How does modeling and simulation inform the selection and design of BsAbs?
M&S approaches can elucidate the conditions under which the BsAb modality is superior
to a traditional combination therapy and inform the design of a BsAb molecule with
optimal characteristics for efficacy.
Clinical
What is the rationale for determining doses in combination treatment involving BsAbs
as one of the therapeutics?
The rationale is based on the prior knowledge of clinical data with individual targets,
toxicity and efficacy studies, and may be different than the equivalent combination
of individual molecules due to factors such as avidity.
How are DDI studies mitigated?
Based on the known existing potential and interactions with the individual targets,
in vitro data and PBPK modeling.
How are safety and efficacy end points selected?
The efficacy and safety end points may be specific for a BsAb and may not be applied
horizontally across all the existing BsAbs. This can often vary from molecule to molecule.
John Wiley & Sons, Ltd.
STRUCTURAL FORMATS OF BsAbs
The functional domain architecture of mAbs have been extensively exploited to create
a number of different BsAbs formats. Spiess et al. originally classified them into
five distinct structural groups:7 i) bispecific IgG,13, 14 ii) IgG appended with an
additional antigen‐binding moiety,15, 16 iii) BsAbs fragments,17, 18 iv) bispecific
fusion proteins,19 and v) BsAb conjugates.20 The new formats are categorized based
on the Fc‐mediated effector functions and are classified as immunoglobulin G (IgG)–like
molecules and non‐IgG–like molecules, as shown in Table
4. The IgG formats are larger and undergo FcRn recycling, which results in a longer
serum half‐life, whereas non‐IgG formats have a smaller size, which enables increased
tissue penetration.
Table 4
Structural format categories for BsAbs
IgG‐like formats
Non‐IgG‐like formats
Quadroma
scFv based BsAbs
Knob‐into‐holes
Nanobodies
Dual variable domains Ig
Dock and lock methods
IgG‐single‐chain Fv (scFv)
Dual affinity retargeting molecules (DARTs)
Two‐in‐one Fab (or Dual action Fab)
Half molecule exchange
Κλ‐bodies
John Wiley & Sons, Ltd.
These unique formats vary in antigen‐binding valency properties, thereby offering
a potential opportunity to optimize valency of each component antibody based on the
biology of the mechanism perturbed by the therapeutic.7 Because these formats are
comprised of individual functional domains, the activities of the domains can be monitored
via quantification of the single domain. Elucidating the exposure of the active therapeutic
for informing model‐based drug development in the context of a BsAb is complex. The
molecular variant chosen to illicit target interrogation can affect the likelihood
of biotransformation of the molecule, a critical factor in translating active exposure
to a pharmacological response. The physiochemical BsAb features must be further coupled
with structural variants that may exist in free, partially bound, and bound forms21,
22 resulting from binding to a soluble target, for example. BsAbs are furthermore
confounded by valency properties of each antibody component binding to its respective
antigen. Partially bound forms of the molecule may alter the stoichiometry associated
with the dual‐target binding of the biotherapeutic. Innovative bioanalytical approaches
are required to fully understand the active exposure of a BsAb, which is dependent
on both its unique physical/chemical properties and the dual‐targeting strategy represented
by the molecule. These distinct structural groups serve to illustrate the structural
complexity and diversity of BsAbs, which raise unique challenges related to the bioanalytical
strategy.
CLINICAL PHARMACOLOGY CONSIDERATIONS
Several global regulatory agencies have published guidelines for the development of
both small molecules and biologics. However, development strategies may differ between
BsAbs, mAbs, and traditional combination therapies.
Immunogenicity, biomarkers, and imaging
As with any biologic molecule, BsAbs have the potential to elicit an immune reaction.
Immunogenicity is typically assessed by detection of antidrug antibodies (ADA). Formation
of ADA is not always associated with mAbs. There are cases of mAbs without ADA formation
(e.g., rilotumumab, which has an ADA incidence of 0%). Therefore, the incidence of
ADA associated with BsAbs cannot be lower than zero. Among BsAbs, catumaxomab has
an ADA incidence of 0%, while blinatumomab has a low ADA incidence of ∼1%. Our experience
in the field suggests that key factors of ADA formation related to BsAbs are similar
to those associated with other mAbs. This may include, but is not limited to, the
structure of BsAbs (i.e., whether it can be recognized by the immune system as “foreign”),
the presence of foreign sequences (e.g., asymmetric rat‐mouse hybrid BsAb), route
of administration (higher incidence of ADA with subcutaneous than with intravenous
(IV) administration), dose, and characteristics of the patient's immune system. It
is well known that the formation of ADA may alter pharmacokinetics (PK), leading to
subsequent changes in the pharmacodynamic (PD) properties of a BsAb. However, the
drug‐binding characteristics of an ADA may lead to differentiating impact on the individual
target binding.23
Biomarker development programs for BsAbs often face several unique challenges. Standard
approaches to evaluate for biological/biochemical impact via target engagement and
PD assays are routinely focused on specific analytes to ensure that an appropriate
dose and schedule are selected to advance farther along into clinical testing. BsAbs
often present the challenge of dissecting the biologic complexity and PD coverage
associated with two targets simultaneously. A BsAb targeting two antigens or ligands
may create the need to define two target coverage thresholds (unique to each marker),
all the while dealing with the reality that the BsAb provides a fixed ratio of exposure
to either target at a given exposure. In the case of a BiTE antibody construct, it
is critical in early clinical trials to demonstrate engagement of T cells (via the
CD3 binder) as well as the tumor‐specific target (CD19 in the case of blinatumomab).
Such T‐cell activation has been explored with blinatumomab by evaluating CD69 and
CD25 upregulation posttreatment in acute lymphocytic leukemia patients.24
Similarly, patient stratification hypotheses for sensitivity or resistance are generally
formulated in the preclinical development and then tested throughout development with
the ultimate goal of identifying a predictive marker for response to a single biologic
process. Patient stratification marker(s) for BsAbs may need to be tailored to each
binding arm of the BsAb, or the proposed combinatorial biology elicited via simultaneous
dual target PD.
Molecular imaging can play an important role in identifying and improving the success
rate of promising new drug candidates, including BsAbs. Goldenberg et al. described
various examples of the utility of BsAbs as tools for imaging in preclinical and clinical
settings. Imaging studies have also been demonstrated to be of value in identifying
the immunogenicity effect of BsAbs.25 These tools can be utilized in oncology for
diagnosis and detection of small tumors or lesions using differential approaches and
rationales, such as Dock and Lock, click chemistry,26 or heterodimer conjugated nanomaterials.27
Drug–drug interaction of BsAbs
Drug–drug interactions (DDIs) for BsAbs are less well understood than for monotherapies
or traditional combination of mAbs. If the investigational therapeutic protein (TP)
is a cytokine or modulates cytokine biology, studies should be conducted to determine
its effects on CYP enzymes or transporters. Lee et al. conducted a survey aimed at
systematically reviewing US Food and Drug Administration (FDA)–approved therapeutic
proteins and the implications of therapeutic protein–drug interactions. The survey
encompassed 68 new therapeutic proteins that had been approved by the FDA by the end
of 2008.28 The results showed that cytokine release followed by administration of
an mAb with cytokine‐modulating properties was a major reason for DDI after mAb administration.
Cytokines are involved in the pathophysiology of multiple human diseases, and their
levels are increased during infection and inflammation. Therefore, biologics that
modulate cytokine activities can indirectly influence the expression of specific CYP
enzymes and drug transporters by affecting cytokine concentrations. This concept is
described extensively elsewhere.29, 30 The magnitude of cytokine‐induced effects on
CYP450 depends on the level of cytokine elevation, the type of cytokine (e.g., IL‐6
being important), and the duration of cytokine elevation. The potential for DDI with
transient cytokine elevation may be very different from that with chronic cytokine
elevation.31
Kenny et al. published an article to facilitate better understanding of the current
science, investigative approaches, and knowledge gaps in this field.32 Key issues
discussed included translating in vitro to in vivo knowledge in DDI along with questions
of whether in vitro data could add value in defining the need for a clinical DDI study,
whether the acute phase response protein C‐reactive protein (CRP) could be used as
a potential biomarker for CYP modulation in inflammatory disease, whether TP‐DDI could
be quantitatively predicted from preclinical data, and how a clinical DDI study can
be designed appropriately.
Assessment of DDI generated with a BsAbs should be approached the same way as with
any large molecule. If clinical studies are restricted to patients instead of healthy
volunteers, population PK modeling provides a feasible approach for TP‐DDI assessment.
Population PK modeling allows less intensive sampling, incorporation of TP‐DDI assessment
in larger phase II and III trials involving relevant patient populations, and integration
of data generated from multiple studies during different development phases. Population
PK modeling also supports evaluation of the effects of combined “perpetrators” on
a TP and, potentially, the effect of a TP on comedications when the analysis is prespecified
and concentrations of the comedications are evaluated. Trends identified in an exploratory
population PK analysis can be used to guide decisions for the need of additional DDI
studies.
Regulatory agencies have included recommendations in their guidance on TP‐DDI assessment
during drug development. The European Medicines Agency (EMA) guideline,33 published
in July 2007, entitled “Guideline on the Clinical Investigation of the Pharmacokinetics
of Therapeutic Proteins,” describes concerns about immunomodulators such as cytokines,
which have shown a potential for inhibition or induction of CYP enzymes, thereby altering
the metabolism of coadministered small‐molecules that are substrates of these enzymes.
The guideline suggests that the in vitro and/or in vivo studies should be considered
on a case‐by‐case basis. The 2012 FDA draft guidance on DDI similarly expands the
US agency's current recommendation on TP‐DDI assessment.34
MODELING AND SIMULATION APPROACHES
Model‐based approaches increasingly support decisions spanning the entire drug development
process, from preclinical development through postmarketing, as shown in Figure 2.
The application of such approaches to the development of BsAbs follows this standard
paradigm and is applied at various stages during the development of biologic therapeutics.
Figure 2
Various applications of modeling and simulation approaches used for BsAbs drug development.
Ang2, angiopoietin‐2; BACE1, β‐secretase 1; BsAb, bispecific antibodies; IGF‐IR, insulin‐like
growth factor‐I receptor; PBPK, physiologically based pharmacokinetic; PD‐1, programmed
death‐1; Tfr, transferrin receptor; TIM‐3, T cell immunoglobulin and mucin domain;
TMDD, target‐mediated drug disposition; TNF‐α, tumor necrosis factor‐alpha.
Translational modeling
Predicting the PK of BsAbs generally follows the same paradigm as mAbs; i.e., allometrically
scaling preclinical PK parameters to predict human PK. Although this tends to work
best for antibodies with linear PK, models incorporating nonlinear clearance mechanisms
have been developed. For example, preclinical PK of the BsAb MEHD7945 in cynomolgus
monkeys were fitted to a standard two‐compartment PK model with nonlinear and linear
clearance components, and the resulting PK parameters were translated using a common
method for scaling.35 More complicated target‐mediated drug disposition (TMDD) models
that mechanistically describe simultaneous binding to two targets have also been proposed,
although it is not straightforward to translate such models from preclinical species
to humans because of a lack of critical information, such as the relative density
of the target in preclinical species compared with humans.36 While this is true of
TMDD models in general, these challenges are exacerbated for BsAb because they bind
two targets simultaneously. Still, successful development of a TMDD model that describes
the PK of a BsAb along with an understanding of species differences impacting the
model may help guide first‐in‐human (FIH) dose selection, because such mechanistic
models predict the degree of target engagement for each BsAb arm. Projection of target
engagement is a key application of modeling and simulation approaches, particularly
for development programs that lack biomarker data. Clinical decisions regarding the
FIH starting dose are based on results from toxicology studies and on expected pharmacology,
a comprehensive approach recommended by the FDA.37 However, such approaches have had
mixed success for traditional mAb and their application has been limited. Advances
in this area represent a significant opportunity for modeling and simulation to contribute
to the transition from preclinical space to the clinic.38
Pharmacology‐based decisions
Mechanistic modeling and simulation approaches tailored to the interrogation of BsAb
pharmacology have yielded critical insights into the mechanism of action of BsAbs
and the conditions under which they offer advantages. A physiologically based pharmacokinetic
(PBPK) model developed by Friedrich et al. provided a rationale for the lack of clinical
success of molecules that retarget effector cells in solid tumor indications, and
a platform for designing molecules with a greater potential for success. Their PBPK
model suggested that effector cells dictate the distribution of the BsAb and that
modifications to the tumor microenvironment were more promising approaches to boosting
efficacy in solid tumor indications than optimizing the binding parameters of the
BsAb.39
Modality selection
For a BsAb targeting both programmed death‐1 (PD‐1) and T‐cell immunoglobulin and
mucin domain (TIM)‐3 to manipulate a patient's immune system into attacking a foreign
entity (such as a cancer), a systems pharmacology approach was used to assess the
combinations potential to provide enhanced efficacy over monotherapy or traditional
combinations. The investigators used a mechanistic model and, based on its predictions
suggesting that a BsAb offered no advantages over a fixed‐dose combination, ultimately
decided not to develop the BsAb. The model predictions also allowed for accelerated
project timelines by informing the selection of individual target candidates with
optimal pharmacological properties.40
Target selection
The development of MM‐141, a tetravalent BsAb targeting the IGF‐IR and ErbB3 pathways
(for the treatment of cancer), was also informed by mechanistic modeling.41 A mechanistic
model that integrates information across pathways can provide insight into both the
selection of the best targets for overcoming resistance mechanisms and the means by
which those targets should be modulated. The simulations for MM‐141 showed that a
BsAb should be used to modulate the targets, predicting a superior outcome with simultaneous
target binding compared with a combination of individual antibodies, for any ratio
of IGF‐IR and ErbB3 receptors.41 Indeed, this prediction was validated by preclinical
studies comparing the antitumor potency of MEHD7945A, a BsAb also targeting IGF‐IR
and ErbB3, with a combination of cetuximab and an anti‐ErbB3 antibody.42 Models of
a similar nature were also developed for rheumatoid arthritis to elucidate the effects
of an anti‐TNFα/anti‐Ang2 BsAb43 and for relapsed/refractory acute lymphoblastic leukemia
(ALL) to provide insight into the mechanisms governing response and nonresponse after
administration of blinatumomab.44
Candidate optimization
Mechanistic modeling approaches have also been used to determine the optimal characteristics
of a BsAb. For example, a mathematical model addressing PK, PD, and safety outcomes
was developed for a BsAb that targets the TfR and BACE1. This model was used to support
preclinical candidate selection. While it may seem intuitive that increasing affinity
of the TfR arm would promote delivery of the molecule to the target site (maximizing
the biologic effect), the modeling showed that there is an optimal TfR affinity range
for maximum reduction of Aβ peptides. Degradation of the molecule during transcytosis
increased with increasing affinity, resulting in lower average brain exposures over
time and reduced pharmacologic effect. Reducing affinity therefore undermined TfR‐mediated
transport through the blood–brain barrier. Similarly, the model suggested the relationship
between TfR affinity and reticulocyte depletion was biphasic in nature. This model
allowed for a rational approach to antibody engineering, preclinical study design,
and candidate selection, resulting in BsAbs with optimal safety and PK/PD profiles.45
These examples show how critical model‐based approaches allow for an improved understanding
of the mechanism of action of BsAbs (compared with corresponding combination therapies),
the conditions under which they offer advantages, and the optimal pharmacologic properties
required of such agents to maximize the therapeutic effectiveness. A combination of
traditional PK/PD modeling approaches and newly emerging quantitative systems pharmacology
provide the best path forward for maximizing the probability of success and for accelerating
the development of these molecules.
BIOANALYTICAL STRATEGY FOR BISPECIFIC ANTIBODIES
Informing meaningful PK and PD assessments of both efficacy and safety in the development
of these molecules demands an appropriate bioanalytical strategy with careful consideration
applied to method suitability for measuring functionally relevant forms of the BsAbs.
The majority of the bioanalytical methods for BsAbs are based on the principles of
ligand binding assays (LBA). The advantages and principles of LBA for measuring biotherapeutics
are well described.46 The choice of an appropriate LBA includes the selection of assay
platform, format, and critical reagents. It also requires appropriate assessment of
other contributing factors that may cause bioanalytical error and mislead the evaluation
(risk assessment). Although LBA constitutes the most commonly used approach, there
are other assay platforms, such as flow cytometry and mass spectrometry, which are
also well suited for addressing bioanalysis questions for BsAbs.
BsAbs can bind to various circulating partners, such as free target proteins, ADAs,
and other endogenous serum components. In addition, BsAbs may lose their binding ability
as a result of biotransformation. Consequently, BsAbs can exist in a pharmacologically
active form, characterized by dual‐target antigen binding sites, and in an inactive
form, which has partial or no target binding sites. Whether to measure the active
concentration or the total concentration (active plus inactive/partially active forms)
for PK/PD assessment continues to be debated as part of the bioanalytical strategy.14,
47 The recent white paper by the AAPS Ligand‐Binding Assay Bioanalytical Focus Group
discussed the challenges and issues of measuring free (active), total drugs and target
proteins and how these data should be used to support drug discovery and development.48
Typically, when considering which form of the therapeutic protein should be measured
to achieve the intended purpose of the study, a “fit‐for‐purpose” approach is adopted.
A working bioanalytical strategy and the selection of appropriate assay technologies
to measure the intended forms is presented in Figure
3 as well as in two examples discussed later.
Figure 3
Bioanalytical strategy and the selection of appropriate assay technologies to measure
the intended forms for BsAb. BsAb, bispecific antibodies; LBA, ligand binding assay;
LC‐MS/MS, liquid chromatography‐tandem mass spectrometry.
COVA322 ((tumor necrosis factor) TNF x (interleukin) IL‐17A), also known as a FynomAb,
is a bispecific fusion protein consisting of an antibody and a Fynomer.19 Fynomers
are small binding proteins derived from the human Fyn SH3 domain. Fynomers can be
engineered to bind to target molecules with the same affinity and specificity as antibodies.
It is critical to maintain the activity of COVA322 and monitor that the Fynomer is
not cleaved in vivo by undergoing potential biotransformation. An enzyme‐linked immunosorbent
assay (ELISA) format utilizing the antigen specificity of the BsAbs is normally used
to directly measure the concentration of the bioactive BsAbs. Two ELISA assays were
developed: bifunctional ELISA, detecting intact and bioactive COVA322 using dual antigen,
and monofunctional ELISA, detecting the anti‐TNF binding antibody portion of COVA322.
The assays demonstrated that the plasma concentrations obtained from PK samples by
both assays were comparable, indicating that COVA322 stayed fully functional (bioactive)
and that there were no indications of in vivo biotransformation. However, this may
not be representative of the results observed for target binding in the bispecific
formats. For instance, glucagon‐like peptide‐1 (GLP‐1) is a 37‐amino acid peptide
for the treatment of type II diabetes via GLP‐1 receptor binding. A CovX‐Body was
generated by conjugating the GLP‐1 peptide to the N‐terminus of a carrier mAb. After
in vivo administration of the GLP‐1 CovX‐Body to mice,49 quantification of the therapeutic
was achieved by two different ELISA assays that each used an anti‐idiotypic antibody
capture of the mAb portion of the construct. Two different detection antibodies were
used: antihuman IgG, to measure the mAb only (total assay), and anti‐GLP‐1 antibody
(N‐terminus specific), to measure the intact GLP‐1 CovX‐Body. The intact assay produced
drug concentrations that were significantly lower than the total assay throughout
the PK time course. Unlike the FynomAb, these results indicate that, although the
concentration of the mAb portion of the construct was sustained, the N‐terminal region
of the GLP‐1 moiety was quickly degraded, demonstrating a high degree of in vivo biotransformation
(e.g., cleavage of GLP‐1 moiety from the antibody).
Because BsAbs may present as a mixture of biologically active and inactive forms,
it is important to identify the BsAb form that is most pharmacologically relevant
to PK/PD assessment and to develop a validated assay that measures the appropriate
form accordingly.
CASE STUDIES
The following case studies illustrate specific examples for each of the issues that
are pertinent to the development of BsAbs.
Case study 1: Catumaxomab
Mechanism of action
The epithelial cell adhesion molecule (EpCAM) represents a potentially attractive
antigen for antitumor therapies, because the transmembrane glycoprotein is highly
expressed in a broad range of solid tumor indications and correlates with a poor patient
prognosis in most of them.50 Catumaxomab is a BsAb targeting EpCAM and CD3 that was
approved in the EU in 2009 for the treatment of malignant ascites. Exudative ascites
is fluid accumulation in the peritoneal cavity (abdominal space) that can be caused
by the presence of solid tumors. Treatment with catumaxomab along with paracentesis
(manual drainage) can significantly delay the need for repeated manual drainage thereafter
in patients with malignant ascites caused by ovarian or nonovarian cancer.51
Catumaxomab has been generated by fusing two different hybridoma cells, a technique
called “quadroma technology.” The hybrid Fc region exerts binding capacity to human
FcγR I, FcγR IIa, and FcγR III enabling activation of NK cells, dendritic cells, monocytes,
and macrophages.52 In addition, the targeting of EpCAM on tumor cells and CD3 on T
cells allows T‐cell–mediated redirected lysis of EpCAM‐positive target cells, which
results in a more than 1,000‐fold higher potency compared with monospecific anti‐EpCAM
antibodies.
Challenges in FIH studies
Catumaxomab is hypothesized to exert its effect via a combination of T‐cell–mediated
tumor cell killing, ADCC, and phagocytosis via FcγR‐activated accessory immune cells.
Delivery of catumaxomab in the clinical setting is limited to localized intraperitoneal
(IP) administration. In an FIH study where catumaxomab was administered IV, fatal
acute liver failure and cytokine release‐associated systemic toxicity were observed
even at low doses.53 In a pilot study, the feasibility of IP administration at increasing
doses was explored; however, no information on starting dose selection was provided.54
IP administration of catumaxomab delivered sufficient compartmental concentrations
observable antitumor activity with mitigation of the systemic side effects from significant
unspecific T‐cell activation caused by intravenous administration.
Bioanalytical approaches
Plasma concentrations of catumaxomab were measured by a validated two‐site ELISA.
Catumaxomab was captured by an antirat IgG λ light chain‐specific antibody. Bound
catumaxomab was then detected via an antimouse IgG2a‐specific biotin‐labeled detection
antibody followed by colorimetric measurement. The assay format suggests measurement
of the total drug concentration by utilizing the chimeric composition of catumaxomab
rather than antigen specificity; therefore, the assay results do not allow for conclusions
about the BsAbs maintenance of in vivo binding activity. A functional bioassay was
performed using PK samples to characterize the functional in vivo binding activity
by testing for the killing activity against EpCAM‐positive tumor cells.55
PK/PD
In a PK study, patients with symptomatic malignant ascites received four IP infusions
of increasing doses (10, 20, 50, and 150 μg) over 6 h each on days 0, 3, 7, and 10.
High concentrations of catumaxomab were observed in ascites, approaching effective
concentrations. The concentration levels increased with the number of infusion doses,
and peak concentrations were detected ∼19 h after completion of the last administration.
Interestingly, these concentrations were sufficient to induce tumor cell killing in
spiking experiments, and this antitumor activity outside the peritoneum was confirmed
by the observation of systemic responses to catumaxomab therapy described in various
case reports.56, 57 The mean terminal elimination half‐life was 2.13 days. PD indication
of antitumor activity was observed by cytokine release (TNF‐α, (interferon) IFN‐γ,
IL‐2, IL‐6, and IL‐10). In the phase II/III study, a relative lymphocyte count above
13% at baseline appeared to be a potential biomarker with prognostic significance.58
In a phase II study, immuno‐monitoring revealed i) redistribution of effector T cells
from blood into the peripheral tissue, ii) expansion and shaping of a preexisting
EpCAM‐specific T‐cell repertoire, and iii) spreading of antitumor immunity to different
tumor antigens. EpCAM‐specific T cells disappeared completely from the peripheral
blood immediately after completion of catumaxomab administration and reappeared with
even higher numerical amounts 4 weeks later. Shaping of immune responses of EpCAM‐specific
T cells was characterized by the occurrence of a T‐cell repertoire more restricted
to certain epitopes with the EpCAM sequences, and expression of Th1‐type effector
T cells.59
Immunogenicity and impact on PK/PD
As expected from the murine/rat nature of the antibody, ADA responses were observed
after last administration of catumaxomab in the PK study.14 However, none of the patients
developed significant ADA responses before the last infusion was administered. Interestingly,
the phase II/III study revealed longer paracentesis‐free survival for human antimouse
antibody (HAMA)‐positive patients vs. HAMA‐negative patients.60 The authors discussed
various possible reasons for this observation. A more intact immune response may lead
to a better responsiveness to catumaxomab, given that ADA responses require a functional
immune system. Moreover, the anti‐idiotypic network hypothesis postulates that i)
anti‐idiotypic antibodies itself can induce humoral responses; and further potentiation
of the immune response can be achieved by ii) internalization of complexes comprised
of murine therapeutic antibodies and human anti‐idiotype antibodies, with its presentation
by antigen‐presenting cells possibly triggering further T‐cell activation, which can
result in antitumor response. Besides the initial humoral response, a direct cellular
immune response could also be induced by cytotoxic T cells recognizing a neoantigen
in the presentation of the murine antibody via the major histocompatibility complex
(MHC) of the antigen‐presenting cells.
CASE STUDIES: BiTE ANTIBODY CONSTRUCTS
BiTE‐mediated killing of cancer cells is independent of common immune escape mechanisms,
such as expression of MHC class 1 molecules, antigen presentation, and activation
of costimulatory molecules. Clinical studies have been conducted or are ongoing for
several BiTE antibody constructs. Some of those studies will be presented in more
detail in the following section.
Case study 2: Blinatumomab
Mechanism of action
Blinatumomab is a BiTE antibody construct that redirects CD‐3‐positive T cells to
CD19‐expressing target cells.61, 62 The targeted CD19 antigen is constitutively expressed
on normal B cells throughout a person's lifetime63 and is highly conserved in B‐cell
malignancies.64, 65 Blinatumomab has an innovative mechanism of action that utilizes
a patient's own T cells to attack CD19‐positive B cells, including malignant cells
such as those in ALL. It transiently connects T cells and B cells, inducing T‐cell‐mediated
killing of the bound B cell. A single activated T cell can trigger a serial lysis
of multiple malignant or normal cells, a process that resembles a natural cytotoxic
T‐cell reaction.
Blinatumomab activates T cells at picomolar concentrations, mimicking a natural MHC
class I/peptide interaction with the T‐cell receptor. Expression of MHC class I and
the presence of T‐cell receptor are not required for the redirected lysis, indicating
that the effect of blinatumomab is independent of peptide antigen presentation and
the presence of a T‐cell receptor.66 An advantage of this approach is that engaged
T cells will be less susceptible to major immune escape mechanisms of tumor cells,
thereby improving the effectiveness of T‐cell‐mediated killing.
Challenges in FIH studies
Blinatumomab does not cross‐react with mice, rats, or dogs. Therefore, a surrogate
molecule (muS103new, binding to equivalent murine target antigens, CD3 and CD19) was
constructed to conduct formal nonclinical toxicology and safety pharmacology investigations.
Although the surrogate mediated redirected lysis of murine B cells and induced activation
of murine T cells along with cytokine release analogous to the mechanism of blinatumomab
in human cells, the blinatumomab FIH dose calculation did not follow conventional
methods. Instead, a “trial and error” approach was applied.
Since blinatumomab was a new class of immunotherapy agent, no true “best schedule”
information could be generated in preclinical studies prior to clinical introduction,
owing to the lack of an appropriate animal model. Hence, three pilot phase I studies
were conducted before a more extensive clinical study was initiated. Blinatumomab
was first tested under 2‐ or 4‐h IV infusion 1, 2, or 3 times weekly. Of note, blinatumomab
has a short half‐life of 1 to 2 h. All three short‐term infusion studies were terminated
because of lack of clinical benefit in the face of central nervous system and cytokine
release‐related adverse events (AEs). Some mechanism‐based biologic activities, such
as cytokine release, minimal T‐cell activation, and selective decreases in peripheral
B‐cell counts, were observed. The AEs appeared to be dose‐dependent and occurred mainly
at the beginning of treatment.67
Due to the fast elimination of blinatumomab and the requirement for higher exposures
to achieve efficacy, it was hypothesized that maintaining a prolonged steady‐state
concentration with continuous IV infusion might be advantageous; a 4‐ or 8‐week continuous
IV infusion for sustained T‐cell activation and B‐cell depletion was initiated. To
mitigate the early AEs related to immune activation, step dosing regimens were introduced
along with pretreatment cytoreduction regimens (mainly via steroids) to better manage
AEs.
In this expanded phase I study, adult patients with relapsed non‐Hodgkin's lymphoma
(NHL) received blinatumomab at doses ranging from 0.5–90 μg/m2/day by continuous IV
infusion for 4 or 8 weeks. In this trial, multiple regimens were tested, including
0.5, 1.5, 5, 15, 30, 60, and 90 μg/m2/day flat dosing; 5–15, 5–30, 5–60, 15–60 μg/m2/day
1‐step dosing; and 5–15–60 μg/m2/day 2‐step dosing. The minimal effective dose and
maximal tolerable dose were established at 5 and 60 μg/m2/day, respectively, and step
dosing regimens were more effective and tolerable than the flat dosing in most cases.68
Bioanalytical approaches
The concentration of blinatumomab in human serum was determined by fluorescence activated
cell sorting (FACS) analysis. The assay is based on the upregulation of CD69 on the
activated T‐cell surface upon dual binding of blinatumomab to CD3 and CD19. The activation
of CD69 was concentration dependent, which can be monitored by FACS after labeling
with a fluorescent anti‐CD69 antibody. This activity‐based assay likely provides advantages
over ELISA in terms of detecting the presence of the drug in its active form and at
low concentrations.67
PK/PD
Blinatumomab is a recombinant non‐glycosylated protein that does not have an Fc domain,
thus it does not undergo FcRn‐mediated recycling. Blinatumomab is rapidly catabolized
into simple amino acids and cleared from the circulation, much like small proteins.
The drug exhibits a linear and time‐independent PK and has not shown signs of target‐mediated
clearance. Blinatumomab mainly remains in the blood circulation with a volume of distribution
around 3–5 L, similar to normal blood volume. It has a short elimination half‐life
of roughly 2 h and negligible renal clearance.69
Blinatumomab acts as a short adaptor, forcing T cells and tumor cells into close proximity,
which results in a transient formation of a cytolytic synapse between a cytotoxic
T cell and the cancer target cell. T cells are only activated by blinatumomab when
a target cell is present.70 Following blinatumomab‐induced T‐cell proliferation, granzyme‐containing
granules and the pore‐forming protein perforin fuse with the T‐cell membrane to discharge
their toxic content into the target cell. Blinatumomab‐induced T‐cell activation and
proliferation locally increases T‐cell numbers in the target tissue.71 The PD effects
of blinatumomab include T‐cell redistribution, activation, and expansion; dose‐dependent
B‐cell depletion; and transient cytokine elevation.69, 72
Considerations for dose selection
Blinatumomab dose selection was primarily based on its PK, PD, safety, and efficacy
profiles. Drug administration with continuous IV infusion was supported by PK and
efficacy profiles. Efficacy appeared to be dose‐dependent, requiring steady blinatumomab
concentrations in the serum.
The requirement for step dosing depended on safety profiles, baseline B‐cell levels,
and target effective dose levels for selected indications. If baseline B‐cell levels
and the target dose level were low, AEs related to immune reaction appeared less prevalent,
potentially negating the need for step dosing, as shown for the treatment of minimal
residual disease (MRD)‐positive ALL.73 If baseline B‐cell levels were high but the
target dose level was low, one‐step dosing was needed for better management of AEs,
as indicated for the treatment for relapsed/refractory ALL.74 Lastly, if both baseline
B‐cell levels and target dose level were high, two‐step dosing was needed for AE management,
as reported for the treatment of relapsed/refractory diffuse large B‐cell lymphoma.75
Immunogenicity and impact on PK
Blinatumomab showed a low incidence of immunogenicity (∼1%) across studies. As the
formation of ADA requires B cells, and depletion of B cells is the primary outcome
of blinatumomab treatment, blinatumomab would prevent B cells from differentiating
into plasma cells and producing ADA. The impact of formation of ADA on PK (i.e., reduction
of drug concentrations) was observed in some cases, whereas the impact on efficacy
and safety cannot be concluded owing to the low number of cases in which ADA developed.69
Drug–drug interaction
Blinatumomab did not affect CYP450 enzyme activities based on in vitro assays with
human hepatocytes. As part of immune reactions, blinatumomab mediates transient cytokine
elevations in patients during the first 1 to 2 days of treatment. The effects of a
cytokine cocktail on P450 isozymes were examined via in vitro experiments, and the
results showed that cytokines suppressed CYP3A4, CYP1A2, and CYP2C9 enzyme activities
in a time‐ and concentration‐dependent manner; the effect was maximized at clinically
observed cytokine peak concentrations.31
It is known that cytokines, especially IL‐6, can suppress CYP450 activities.76 A PBPK
model for IL‐6 was established to evaluate magnitude and duration of IL‐6 suppression
on hepatic CYP450 activities. The results suggested that transient IL‐6 elevation
(1 to 2 days) up to clinically observed peak concentrations could suppress CYP3A4,
CYP1A2, and CYP2C9 activities by as much as 30% for a week. This may in turn cause
a < twofold increase of drug exposure to sensitive substrates of CYP3A4, CYP1A2, and
CYP2C9. This evaluation indicated that the blinatumomab‐induced transient cytokine
elevation may have a low risk for clinical drug interaction.31 Safety monitoring,
especially for CYP3A4 substrates that have narrow therapeutic windows, is suggested
in the early part of treatment.77
Case study 3: Solitomab (AMG 110, MT110)
Mechanism of action
The EpCAM‐targeting BiTE antibody construct consists of two single‐chain Fv domains
derived from two different antibodies, one targeting EpCAM on epithelial‐derived cancer
cells and the other targeting CD3ε on T cells, to form a single polypeptide chain.
The mechanism of action is similar to other BiTE antibody constructs, as described
previously. Solitomab showed potent in vitro antitumor activity and prevented tumor
outgrowth completely, or resulted in durable eradication of established tumors in
NOD/SCID mouse models.78
Challenges in FIH studies
An FIH study at doses between 1 and 96 μg/day was conducted in patients with refractory
solid tumors known to frequently express EpCAM.79 The starting dose of 1 μg/day during
weeks 1–4 was selected based on the minimal anticipated biological effect level (MABEL)
established with a murine surrogate in mice. Dose‐limiting toxicities were mainly
changes in gastrointestinal system (liver function abnormalities and severe diarrhea).
Transient liver enzyme elevations during the first days of treatment demanded a low
starting dose of 3 μg/day and slow, stepwise, intrapatient escalation. A prophylactic
corticosteroid treatment during the first 3 days of solitomab therapy was instituted
to prevent the initial changes in liver parameters. Severe diarrhea limited longer
infusions for more than 3 or 4 weeks at higher doses. The nature and severity of dose‐limiting
toxicities required multiple adjustments to the dosing regimen and made it difficult
to identify a therapeutic window. Preclinical studies in cynomolgus monkey using a
crossreactive molecule instead of rodent studies with a mouse surrogate may help identify
possible severe adverse reactions more clearly.
PK/PD
PK analysis suggested linear PK over the tested dose range, with the steady‐state
plasma level reached within 24 h.80 The half‐life of solitomab was 4.5 h. The mean
maximum serum concentration (Cmax) was reported as ∼6 ng/mL at a dose of 96 μg/day.
Preclinical studies with a murine version of solitomab showed that a 1‐week adaptation
at a lower dose permitted prolonged treatment at a high dose thereafter by blunting
the initial cytokine release. Furthermore, repeated long‐term dosing did not cause
T‐cell anergy or compromise the effector function of T cells.80, 81
Immunogenicity and impact on PK/PD
ADA were detected in 7 of 63 (11%) tested patients; two patients had altered PK. None
of the ADA‐positive patients showed signs or symptoms that could be attributed to
anaphylactic or other hypersensitivity‐type reactions in the phase I study (Amgen
data on file).
Case study 4: Other BiTE antibody constructs
Challenges in FIH and PK Considerations
Ryan et al. reported development of a CEA/CD3ε‐specific BiTE antibody construct that
targets carcinoembryonic antigen (CEA), which is known to be found on epithelial cell
membranes and in the cytoplasm of gastrointestinal adenocarcinomas, breast, and lung
cancers.82 The molecule exerts the same BiTE antibody construct‐associated mechanism
of action. Low BiTE antibody construct concentrations were sufficient to induce killing
of CEA and tumor cells after administration of T cells from patients or healthy donors.83
An FIH study was conducted in adult patients with advanced gastrointestinal adenocarcinomas.
Since no relevant animal model could be identified to perform in vivo toxicology studies,
the selection of the FIH start dose was solely based on assessment of dose–response
correlations in vitro. The MABEL that demonstrated 20% maximal effect (EC20) of BiTE
antibody construct‐induced tumor cell lysis as the most sensitive measure of biologic
activity was used to select the starting dose for the FIH study. In addition, a nonterminal
PK study conducted in cynomolgus monkeys was used to predict human PK parameters based
on allometric scaling. The exposure ranges around the identified MABEL concentration
guided selection of the start dose and administration schedule for the phase I study.84
PK considerations may also inform phase I studies. As seen with blinatumomab and solitomab,
antitumor activity may require a long‐term, steady‐state exposure above a threshold
with trough levels maintained above the range of EC50 or EC90. In addition, by avoiding
fluctuations in Cmax exposures, a sustained target coverage with less Cmax‐driven
toxicity can be expected. The start dose selection for additional phase Ia or phase
Ib studies can also be based on a simulation of average total area‐under‐the‐curve
with cIV administration utilizing PK information from intermittent dosing of the FIH
study.
Friedrich et al. reported another example of a BiTE antibody construct targeting prostate‐specific
membrane antigen (PSMA) via binding to PSMA and CD3ε of human and macaque origin.
It is the first BiTE antibody construct that contains an amino acid sequence very
close to human germline Ig‐V segments, allowing crossreactivity to human and nonhuman
primate PSMA and CD3 antigens. In preclinical experiments, the PMSA/CD3ε BiTE antibody
construct was considerably more potent and able to mediate rapid tumor shrinkage and
complete remissions of established human 22Rv1 prostate cancer xenografts after subcutaneous
(s.c.) administration of the PMSA/CD3ε BiTE antibody construct. A serum half‐life
of approximately 8 h after single bolus or s.c. administration and 18% bioavailability
after s.c. administration in mice was found.85 These results formed the rationale
for the selected route of administration in the FIH study (ClinicalTrials.gov NCT01723475).
CONCLUSION
BsAbs are an emerging drug modality with significant therapeutic value. While the
development of BsAbs is an evolving field, the growing experience and application
of BsAbs will become increasingly important for informing development steps. We have
attempted to expand the body of existing knowledge in the space of BsAb.