Biology drives the discovery of bispecific ... - Oxford Academic

45
To whom correspondence should addressed. Jijie Gu or Siwei Nie. Email: [email protected]; [email protected]. © The Author(s) 2020. Published by Oxford University Press on behalf of Antibody Therapeutics. Antibody Therapeutics, 2020, Vol. 3, No. 1 18–62 doi:10.1093/abt/tbaa003 Advance Access Publication on 17 February 2020 Review Article Biology drives the discovery of bispecic antibodies as innovative therapeutics Siwei Nie 1, *, Zhuozhi Wang 1 , Maria Moscoso-Castro 2 , Paul D’Souza 2 , Can Lei 2 , Jianqing Xu 1 and Jijie Gu 1, * 1 WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and 2 Clarivate Analytics, Friars House, 160 Blackfriars Road, London SE1 8EZ, UK Received: December 10, 2019; Revised: February 7, 2020; Accepted: Month 0, 2000 ABSTRACT A bispecic antibody (bsAb) is able to bind two different targets or two distinct epitopes on the same target. Broadly speaking, bsAbs can include any single molecule entity containing dual specicities with at least one being antigen-binding antibody domain. Besides additive effect or synergistic effect, the most fascinating applications of bsAbs are to enable novel and often therapeutically important concepts otherwise impossible by using monoclonal antibodies alone or their combination. This so-called obligate bsAbs could open up completely new avenue for developing novel therapeutics. With evolving understanding of structural architecture of various natural or engineered antigen-binding immunoglobulin domains and the connection of different domains of an immunoglobulin molecule, and with greatly improved understanding of molecular mechanisms of many biological processes, the landscape of therapeutic bsAbs has signicantly changed in recent years. As of September 2019, over 110 bsAbs are under active clinical development, and near 180 in preclinical development. In this review article, we introduce a system that classies bsAb formats into 30 categories based on their antigen-binding domains and the presence or absence of Fc domain. We further review the biology applications of approximately 290 bsAbs currently in preclinical and clinical development, with the attempt to illustrate the principle of selecting a bispecic format to meet biology needs and selecting a bispecic molecule as a clinical development candidate by 6 critical criteria. Given the novel mechanisms of many bsAbs, the potential unknown safety risk and risk/benet should be evaluated carefully during preclinical and clinical development stages. Nevertheless we are optimistic that next decade will witness clinical success of bsAbs or multispecic antibodies employing some novel mechanisms of action and deliver the promise as next wave of antibody-based therapeutics. Statement of Significance: This article comprehensively reviewed various bispecic antibody formats and the biology driving the design and selection of a right bispecic antibody to enable novel therapeutic concept and match intended therapeutic applications. The principles and the examples discussed could provide a general guidance for people interested in exploring bispecic antibody therapeutics. KEYWORDS: bispecic antibody; bsAb; multispecic antibody; msAb A BRIEF HISTORICAL VIEW OF BISPECIFIC ANTIBODIES The invention of hybridoma technology in 1975 marked the arrival of new era of monoclonal antibody (mAb)- based therapy [1]. However, the first wave of clinical attempts with mouse antihuman mAb therapeutics during 1975–86 largely failed, due to immunogenicity of mouse sequences, with only one mAb (anti-CD3 muromonab) being approved. It took another decade for the field to solve the immunogenicity issues, and the lessons learned from the first wave of clinical trials of antibody therapeutics is the key driver leading to invention of innovative anti- body humanization technologies represented by antibody chimerization, CDR graft, in vitro display of human Downloaded from https://academic.oup.com/abt/article/3/1/18/5739255 by guest on 30 May 2022

Transcript of Biology drives the discovery of bispecific ... - Oxford Academic

∗To whom correspondence should addressed. Jijie Gu or Siwei Nie. Email: [email protected]; [email protected].

© The Author(s) 2020. Published by Oxford University Press on behalf of Antibody Therapeutics.

Antibody Therapeutics, 2020, Vol. 3, No. 1 18–62doi:10.1093/abt/tbaa003

Advance Access Publication on 17 February 2020

Review Article

Biology drives the discovery of bispecificantibodies as innovative therapeuticsSiwei Nie1,*, Zhuozhi Wang1, Maria Moscoso-Castro2, Paul D’Souza2,Can Lei2, Jianqing Xu1 and Jijie Gu1,*1WuXi Biologics, 299 Fute Zhong Road, Waigaoqiao Free Trade Zone, Shanghai 200131, China and 2ClarivateAnalytics, Friars House, 160 Blackfriars Road, London SE1 8EZ, UK

Received: December 10, 2019; Revised: February 7, 2020; Accepted: Month 0, 2000

ABSTRACT

A bispecific antibody (bsAb) is able to bind two different targets or two distinct epitopes on the sametarget. Broadly speaking, bsAbs can include any single molecule entity containing dual specificities withat least one being antigen-binding antibody domain. Besides additive effect or synergistic effect, the mostfascinating applications of bsAbs are to enable novel and often therapeutically important concepts otherwiseimpossible by using monoclonal antibodies alone or their combination. This so-called obligate bsAbs couldopen up completely new avenue for developing novel therapeutics. With evolving understanding of structuralarchitecture of various natural or engineered antigen-binding immunoglobulin domains and the connectionof different domains of an immunoglobulin molecule, and with greatly improved understanding of molecularmechanisms of many biological processes, the landscape of therapeutic bsAbs has significantly changedin recent years. As of September 2019, over 110 bsAbs are under active clinical development, and near180 in preclinical development. In this review article, we introduce a system that classifies bsAb formatsinto 30 categories based on their antigen-binding domains and the presence or absence of Fc domain.We further review the biology applications of approximately 290 bsAbs currently in preclinical and clinicaldevelopment, with the attempt to illustrate the principle of selecting a bispecific format to meet biologyneeds and selecting a bispecific molecule as a clinical development candidate by 6 critical criteria. Given thenovel mechanisms of many bsAbs, the potential unknown safety risk and risk/benefit should be evaluatedcarefully during preclinical and clinical development stages. Nevertheless we are optimistic that next decadewill witness clinical success of bsAbs or multispecific antibodies employing some novel mechanisms ofaction and deliver the promise as next wave of antibody-based therapeutics.

Statement of Significance: This article comprehensively reviewed various bispecific antibody formats andthe biology driving the design and selection of a right bispecific antibody to enable novel therapeuticconcept and match intended therapeutic applications. The principles and the examples discussed couldprovide a general guidance for people interested in exploring bispecific antibody therapeutics.

KEYWORDS: bispecific antibody; bsAb; multispecific antibody; msAb

A BRIEF HISTORICAL VIEW OF BISPECIFICANTIBODIES

The invention of hybridoma technology in 1975 markedthe arrival of new era of monoclonal antibody (mAb)-based therapy [1]. However, the first wave of clinicalattempts with mouse antihuman mAb therapeutics during1975–86 largely failed, due to immunogenicity of mouse

sequences, with only one mAb (anti-CD3 muromonab)being approved. It took another decade for the field tosolve the immunogenicity issues, and the lessons learnedfrom the first wave of clinical trials of antibody therapeuticsis the key driver leading to invention of innovative anti-body humanization technologies represented by antibodychimerization, CDR graft, in vitro display of human

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 19

antibody repertoire, and human immunoglobulin trans-genic rodents. The approval of rituximab by the USFDA in 1997 marked the field entering into the boomingstage. About 100 antibody-based therapeutics have beenapproved by the regulatory agencies worldwide, since then,antibody therapeutics have now become one of the main-stays for developing new medicines. The history of devel-opment of bispecific antibodies (bsAbs) almost followedthe footprint of the development of mAb therapeutics.As illustrated in a recent review article [2], starting in the1960s, scientists explored generation of antigen-bindingfragments (Fabs) from two different polyclonal sera andreassociated them into bispecific F(ab’)2 molecules. Afterhybridoma technology was established in 1975, chemicalconjugation of two rodent mAbs or fusion of two antibody-producing hybridomas (so-called quodroma) was exploredimmediately to make bsAbs with defined specificities.The first therapeutic bsAb catumaxomab (Removab

®)

approved by the EMA in 2009 was made by this earlytechnology. The bsAbs made by these two methods prior toestablishing antibody humanization technologies, however,suffered from the same issue of immunogenicity in additionto stability, solubility, and manufacturability challenges.The development of methods to produce recombinantantibodies in the 1980s enabled the rapid generation ofvarious bsAbs with defined structure, composition, andbiochemical, functional, and pharmacological properties,but it still took scientists more than 2 decades to reallyunderstand the unique structural features of variousantigen-binding building blocks such as Fab, Fv, scFv,SDA, etc., to develop various innovative engineeringsolutions to generate homo- and heterodimerizationbuilding blocks necessary for making various bispecificformats and most importantly understand the structuralbiology of how to connect them together to enablevarious biology concepts while maintaining favorabledevelopability. In the later paragraphs, we will review theevolution of some of those landmark solutions for bsAbconstruction. But before we get into detailed discussion ofhow to make various recombinant bsAbs, we will discussthe principles governing how to define and identify a goodbsAb therapeutics first.

THE PRINCIPLES GOVERNING A GOODTHERAPEUTIC BISPECIFIC ANTIBODY

Though mAbs have demonstrated definitive therapeuticbenefits in multiple disease areas, it is believed that bsAbscan further advance the success of therapeutic antibodiesby enabling the molecules with new mechanisms of action(MOAs) and by providing new functional advantages thatcannot be achieved by mAbs. We believe that identificationof a good bsAb should be based on three principles (Fig. 1):(1) the molecule should be able to provide unique biologicalfunction to achieve desired efficacy with appropriate safetyprofile, driven by unique biology; (2) the format chosenshould enable the molecule to fulfill its proposed function,match biology with an optimal format; and (3) the moleculeselected as a clinical development candidate should satisfythe six criteria critical for clinical development and

commercial manufacturing, i.e., desired clinical efficacy,appropriate safety profile, favorable pharmacokinetic/pharmacodynamic (PK/PD) properties, appropriate physic-ochemical properties, scalable manufacturability, and min-imal or no immunogenicity risk—select a right molecule.Unfortunately, these six criteria, particularly those crit-ical for biological function (efficacy, safety, PK/PD,immunogenicity) and those critical for developability(expression, homogeneity, solubility, stability, viscosity,formulation ability, etc.), often are not correlated witheach other, sometimes even counterbalance each other thatrequires balancing when selecting a therapeutic molecule.Identification of a good therapeutic bispecific moleculetherefore usually requires starting with good therapeuticmolecular design defined by molecular product profile(MPP) that is developed based on target product profile(TPP), followed by rigorous molecular and functionalscreen, selection and characterization using pharmacologi-cal assays, mechanistic and/or disease models, and otherpreclinical translational systems relevant to the humandisease one intends to treat.

THE MAKING OF RECOMBINANT BISPECIFICANTIBODIES

In a recent review article, Brinkmann and Kontermannthoroughly reviewed many experimentally verified formatsthat had been described in the literature as of September2016 [3]. We concur with their opinion that besides thefreedom-to-operate (FTO) and the desire to generate pro-prietary intellectual properties (IP) for competitive reason,one of the critical drivers for explosive diversity of so manybsAb formats is the plethora of desired functionalities andapplications of bsAbs. Format variability is essential toserve diverse bsAb applications defined by different TPP.These formats may vary in size, domain composition andarrangement, binding kinetics and valencies, flexibility andgeometry of their binding modules, as well as in their bio-distribution and pharmacokinetic properties to fulfill a par-ticular clinical application. Small variations, such as minorchanges in linker length or composition of domains, canbe crucial determinants for functionality. Some designedparameters may be deduced from structural modeling ofdrug-target interaction. In many cases, however, a suitablemolecule must be identified by generating and compar-ing the functionalities of different formats and differentmolecules in the systems relevant to clinical settings.

Here we review various bsAb formats and classify theminto 30 categories: (1) what are the building blocks ofantigen-binding and their combination, and (2) whetherthey contain fragment of crystallizable region (Fc) domain.From published reports and our practice, most bispecificformats contain the antigen-binding sites derived fromimmunoglobulin domain of native antibodies. We identifysingle-domain antibody (SDA or VHH), variable fragment(Fv), single-chain variable fragment (scFv), Fab, and single-chain antigen-binding fragment (scFab) as the five keybuilding blocks of bispecific formats. As shown in Fig. 2,most of bsAb formats can be classified into 30 groups basedon the above classification. As there are more than 200

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

20 Antibody Therapeutics, 2020

Figure 1. The principles, criteria and screening funnel in discovering a good therapeutic bsAb. (A) Three principles of governing the discovery of a goodbsAb, (B) Six criteria of defining a bsAb as a clinical development candidate. (C) Detailed function and developability screenings to identify a goodtherapeutic bsAb molecule.

bispecific formats from published data and our practice,we do not intend to list all these formats in Fig. 2. Instead,we have just listed an example of each category to illustratethe concept.

In each category, the bispecific formats can be furtherclassified by their geometry (such as homodimer vs. het-erodimer) and valency (number of antigen-binding sites). AbsAb with one binding site to target A and one binding siteto target B is called 1 + 1 format. Similarly there are 1 + 2,1 + 3, and 2 + 2 formats. The formats with more than fourantigen-binding sites are uncommon but growing, so theyare just mentioned as examples in this review.

In addition to the building blocks, absence or presenceof Fc, and different valency, multiple fusion sites of Fc-containing formats increase the complexity of bispecificformats. As shown in Fig. 3A, an antigen-binding build-ing block can be fused to N-terminus or C-terminus ofan Fc fragment or inserted between CH2 domain andCH3 domain. On a heterodimeric Fc-containing bispe-cific format, there are at least six fusion sites. If an Fc-containing format also comprises of CL, the fusion sitesincrease to 12 (Fig. 3A). Moreover, theoretically all theloops of each immunoglobulin domain (CL, CH1, CH2,and CH3) can be used as fusion sites to integrate an antigen-

binding building block. It becomes obvious to employ thesefusion sites to make a bispecific format with desired bindingactivity.

Bispecific molecules containing non-antibody-bindingdomains such as peptides, ligands, receptors, or alternativescaffolds may not fall into this classification system.However, depending on how many polypeptide chainsof the antigen-binding sites are used, the non-antibodybispecific molecules can be constructed using similarapproaches as the above bsAbs.

Bispecific antibody fragments without Fc

In this category, all antigen-binding sites are from the afore-mentioned building blocks (SDA, Fv, scFv, Fab, and scFab)and the bsAbs do not contain Fc. Many different bispecificformats, including 1 + 1, 1 + 2, 1 + 3, and 2 + 2 formats,and trispecific formats have been used for preclinical andclinical development (Tables 1–6). BsAb fragments usuallyare smaller than IgG and lack of Fc-related functions suchas Fcγ R-, FcRn-, and complement-binding and relatedactivities. Due to large number of the bsAb fragment for-mats, only some examples of bsAbs fragments are brieflydescripted below.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 21

Tab

le1.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

tobl

ock

the

angi

ogen

esis

and/

ortu

mor

igen

esis

for

canc

ertr

eatm

ent

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

Dilp

acim

ab,

AB

T-1

65A

bbV

ieV

EG

DL

L4

Pha

seII

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctF

ab+

Fv

wit

hF

c,2

+2

NC

T01

9460

74,

NC

T01

9460

74,

NC

T03

3688

59,

NC

T03

3688

59M

P02

50M

olec

ular

Par

tner

sA

GV

EG

HG

albu

min

Pha

seII

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctSc

affo

ld1

+1

+1

NC

T02

1944

26,

NC

T03

1366

53,

NC

T03

4185

32A

BL

-001

,N

OV

-150

1,T

R-0

09A

BL

Bio

,TR

IGR

The

rape

utic

sV

EG

DL

L4

Pha

seI

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2N

CT

0329

2783

Van

uciz

umab

,R

G-7

221

Roc

he,H

arva

rdM

edic

alSc

hool

,N

atio

nalC

ance

rC

entr

eof

Sing

apor

e

AN

GP

T2

×V

EG

FP

hase

IA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

CT

0168

8206

,N

CT

0214

1295

,N

CT

0266

5416

BI-

8368

80B

oehr

inge

rIn

gelh

eim

,Sa

nofi

AN

GP

T2

×V

EG

F,al

bum

inP

hase

IA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

VH

+V

H,1

+1

+1

NC

T02

6741

52,

NC

T02

6895

05,

NC

T03

4684

26,

NC

T03

8612

34,

NC

T03

9721

50N

avic

ixiz

umab

,O

MP

-305

B83

Onc

oMed

Pha

rmac

euti

cals

VE

GF

×D

LL

4P

hase

IA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

CT

0229

8387

,N

CT

0303

0287

,N

CT

0303

5253

KN

-026

Jian

gsu

Alp

ham

abB

ioph

arm

aceu

tica

lsH

ER

HE

R2

Pha

seII

Ant

i-tu

mor

igen

esis

Bip

arat

opic

Fab

+F

abw

ith

Fc,

1+

1N

CT

0361

9681

,N

CT

0384

7168

,N

CT

0392

5974

,N

CT

0404

0699

ZW

-25

Zym

ewor

ks,B

eiG

ene

HE

R2

×H

ER

2P

hase

IIA

nti-

tum

orig

enes

isB

ipar

atop

icF

ab+

scF

vw

ith

Fc,

1+

1N

CT

0289

2123

,N

CT

0392

9666

MC

LA

-128

Mer

usH

ER

HE

R2

Pha

seII

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

CT

0291

2949

,N

CT

0332

1981

EM

B-0

1,F

IT-0

13a

Epi

mA

bB

ioth

erap

euti

csE

GF

cME

TP

hase

I/II

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

2+

2N

CT

0379

7391

JNJ-

6118

6372

,JN

J-63

72Ja

nsse

nE

GF

cME

TP

hase

IA

nti-

tum

orig

enes

isC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NC

T02

6097

76,

NC

T04

0774

63B

CD

-147

Bio

cad

HE

R2

×H

ER

2P

hase

IA

nti-

tum

orig

enes

isB

ipar

atop

icF

ab+

scF

vw

ith

Fc,

1+

2N

CT

0391

2441

MB

S-30

1B

eijin

gM

abw

orks

Bio

tech

HE

R2

×H

ER

2P

hase

IA

nti-

tum

orig

enes

isB

ipar

atop

icF

ab+

Fab

wit

hF

c,1

+1

NC

T03

8420

85

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

22 Antibody Therapeutics, 2020

Tab

le1.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

BI-

9056

77B

oehr

inge

rIn

gelh

eim

LR

P5/

6P

hase

IA

nti-

tum

orig

enes

isB

ipar

atop

icSD

A+

SDA

,1+

1N

CT

0360

4445

MP

0274

Mol

ecul

arP

artn

ers

AG

Her

Her

2P

hase

IA

nti-

tum

orig

enes

isB

ipar

atop

icSC

AF

FO

LD

,1+

1N

CT

0308

4926

VE

GF

R2/

Ang

2E

liL

illy

&C

oV

EG

FR

AN

GP

T2

Pre

clin

ical

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2N

A

FS-

101

F-s

tar

The

rape

utic

sL

tdE

GF

HG

FP

recl

inic

alA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Fab

+SD

Aw

ith

Fc,

2+

2N

A

MP

-E-8

-3/1

959

Med

iaP

harm

aE

ndos

ialin

×L

GA

LS3

BP

Pre

clin

ical

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctN

otdi

sclo

sed

NA

PM

C-0

01P

harm

Abc

ine

Tie

-2×

VE

GF

R2

Pre

clin

ical

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

PM

C-2

01P

harm

Abc

ine

DL

L4

×V

EG

FR

2P

recl

inic

alA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dN

AP

MC

-404

Pha

rmA

bcin

eA

NG

PT

VE

GF

-cP

recl

inic

alA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dN

AM

CL

A-1

29B

etta

Pha

rmac

euti

cals

;M

erus

VE

GF

×cM

ET

Pre

clin

ical

Ant

i-an

giog

enes

is,

anti

-tum

orig

enes

isC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NA

MP

-EV

20/1

959

Med

iaP

harm

aH

ER

LG

AL

S3B

PP

recl

inic

alA

nti-

angi

ogen

esis

,an

ti-t

umor

igen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dN

A

CK

D-7

02C

hong

Kun

Dan

gP

harm

aceu

tica

lE

GF

cME

TP

recl

inic

alA

nti-

tum

orig

enes

isC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2N

A

CB

A-0

702

Sorr

ento

The

rape

utic

sH

er3

×cM

ET

Pre

clin

ical

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

scF

v+

scF

vw

ith

Fc,

1+

1N

A

SRB

-19

SunR

ock

Bio

phar

ma

EG

FR

×H

er3

Pre

clin

ical

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dN

AA

nti-

HE

R2

and

HE

R3

mA

bB

ioca

dL

tdH

er2

×H

er3

Pre

clin

ical

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dN

A

BT

A-1

06Z

enya

kuK

ogyo

Co

Ltd

IgM

×H

LA

-DR

Pre

clin

ical

Ant

i-tu

mor

igen

esis

Com

bina

tori

alef

fect

?F

ab+

Fab

wit

hF

c,1

+1

NA

TX

B4-

BC

2O

ssia

nix

Inc

TfR

×E

GF

RvI

IIP

recl

inic

alA

nti-

tum

orig

enes

isT

roja

nho

rse

Fab

+SD

Aw

ith

Fc,

2+

2N

A

TX

B4-

BC

1O

ssia

nix

Inc

TfR

×C

D20

Pre

clin

ical

Ant

i-tu

mor

igen

esis

Tro

jan

hors

eF

ab+

SDA

wit

hF

c,2

+2

NA

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 23

Tab

le2.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

toen

hanc

etu

mor

imm

unit

yfo

rca

ncer

trea

tmen

t

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

KN

-046

Jian

gsu

Alp

ham

abB

ioph

arm

aceu

tica

lsP

D-L

CT

LA

-4P

hase

IIE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

SDA

+SD

Aw

ith

Fc,

2+

2N

CT

0352

9526

,N

CT

0373

3951

,N

CT

0383

8848

,N

CT

0387

2791

,N

CT

0392

5870

,N

CT

0392

7495

,N

CT

0404

0699

AK

-104

Ake

soB

ioph

arm

aP

D-1

×C

TL

A-4

Pha

seI/

IIE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2N

CT

0326

1011

,N

CT

0385

2251

Duo

Bod

y-P

D-L

1x4-

1BB

,G

EN

-104

6

Bio

NT

ech,

Gen

mab

PD

-L1

×4-

1BB

Pha

seI/

IIE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+F

abw

ith

Fc,

1+

1N

CT

0391

7381

RE

GN

-567

8R

egen

eron

PSM

CD

28P

hase

I/II

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

Fab

wit

hF

c,1

+1

NC

T03

9726

57

FS1

18m

Ab2

,F

S-11

8,L

AG

-3/P

D-L

1m

Ab2

F-s

tar

PD

-L1

×L

AG

-3P

hase

IE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+SD

A,2

+2

NC

T03

4404

37

IBI-

318

Inno

vent

Bio

logi

cs,L

illy

PD

-1×

PD

-L1

Pha

seI

Enh

ance

tum

orim

mun

ity

Pro

mot

edo

wnr

egul

atio

nN

otdi

sclo

sed

NC

T03

8751

57

LY-3

4341

72E

liL

illy

PD

-1×

PD

-L1

Pha

seI

Enh

ance

tum

orim

mun

ity

Pro

mot

edo

wnr

egul

atio

nF

ab+

Fab

wit

hF

c,1

+1

NC

T03

9369

59

MG

D-0

13M

acro

Gen

ics,

ZA

IL

abP

D-1

×L

AG

-3P

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

v+

Fv

wit

hF

c,2

+2

NC

T03

2192

68,

NC

T04

0823

64X

mA

b-23

104

Xen

cor

PD

-1×

ICO

SP

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

1+

1N

CT

0375

2398

AB

BV

-428

Abb

Vie

MSL

CD

40P

hase

IE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

scF

v+

scF

vw

ith

Fc,

2+

2N

CT

0295

5251

AD

C-1

015,

AT

OR

-101

5A

lliga

tor

Bio

scie

nce

OX

40×

CT

LA

-4P

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

CT

0378

2467

INB

RX

-105

-1,

INB

RX

-105

,ES-

101

Inhi

brx,

Elp

isci

ence

Bio

Pha

rma

PD

-L1

×4-

1BB

Pha

seI

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nSD

A+

SDA

wit

hF

c,2

+2

NC

T03

8096

24

MC

LA

-145

Mer

us,I

ncyt

eP

D-L

4-1B

BP

hase

IE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+F

abw

ith

Fc,

1+

1N

CT

0392

2204

ME

DI-

5752

Med

Imm

une

PD

-1×

CT

LA

-4P

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NC

T03

5303

97

MG

D-0

19M

acro

Gen

ics

PD

-1×

CT

LA

-4P

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

v+

Fv

wit

hF

c,2

+2

NC

T03

7610

17

PR

S-34

3P

ieri

sH

ER

4-1B

BP

hase

IE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+SC

AF

FO

LD

wit

hF

c,2

+2

NC

T03

3305

61,

NC

T03

6503

48R

G-7

769,

RO

-712

1661

Roc

heP

D-1

×T

IM-3

Pha

seI

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

CT

0370

8328

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

24 Antibody Therapeutics, 2020

Tab

le2.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

Xm

Ab-

2071

7X

enco

rP

D-1

×C

TL

A-4

Pha

seI

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T03

5174

88

Xm

Ab-

2284

1X

enco

rC

TL

A-4

×L

AG

-3P

hase

IE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

1+

1N

CT

0384

9469

RG

-782

7R

oche

FAP

×4-

1BB

Pha

seI

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

LIG

AN

Dw

ith

Fc,

1+

3C

ompa

nyde

velo

pmen

tpi

pelin

eM

P03

10M

olec

ular

Par

tner

sA

G,

Am

gen

FAP

×C

D40

Pha

seI

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nSC

AF

FO

LD

,1+

1N

CT

0404

9903

HX

-009

Han

XB

ioph

arm

aceu

tica

lsP

D-1

×C

D47

IND

File

dE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

CT

0409

7769

AK

-112

Ake

soB

ioph

arm

aV

EG

PD

-1IN

DF

iled

Enh

ance

dtu

mor

imm

unit

y,an

ti-a

ngio

gene

sis

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,2

+2

NC

T04

0472

90

INV

-531

Inve

nra

Inc

OX

40bi

para

topi

cP

recl

inic

alE

nhan

cetu

mor

imm

unit

yB

ipar

atop

icF

ab+

Fab

wit

hF

c,1

+2

NA

AT

OR

-114

4A

lliga

tor

Bio

scie

nce

GIT

CT

LA

-4P

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

BH

-299

6h

Bei

jing

Han

mi

Pha

rmac

euti

cal

PD

-1×

PD

-L1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Pro

mot

edo

wnr

egul

atio

nF

ab+

Fab

wit

hF

c,1

+1

NA

GE

N-1

042

Bio

NT

ech;

Gen

mab

CD

40×

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NA

CB

-213

Cre

scen

doB

iolo

gics

PD

-1×

LA

G-

albu

min

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

SDA

+SD

A+

SDA

,1

+1

+2

NA

FS-

120

F-s

tar

The

rape

utic

sO

X40

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+SD

A,2

+2

NA

ME

DI-

3387

Med

Imm

une

LL

CG

ITR

×P

D-1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+L

IGA

ND

wit

hF

c,2

+2

NA

ME

DI-

5771

Med

Imm

une

LL

CG

ITR

×P

D-1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+L

IGA

ND

wit

hF

c,2

+2

NA

PT

-302

Pha

nes

The

rape

utic

sL

AG

-3×

TIM

-3P

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctN

otdi

sclo

sed

NA

TSR

-075

TE

SAR

OIn

cP

D-1

×L

AG

-3P

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctN

otdi

sclo

sed

NA

PD

-1/L

AG

-3bi

spec

ific

mA

bsX

enco

rIn

cP

D-1

×L

AG

-3P

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

1+

1N

A

AM

-105

AbC

lon

Inc

EG

FR

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nN

otdi

sclo

sed

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 25

Tab

le2.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

AL

G-A

PV

-527

Alli

gato

r;A

ptev

oT

hera

peut

ics

Inc

5T4

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nsc

Fv

+sc

Fv

wit

hF

c,2

+2

NA

BY

-24.

3B

eijin

gB

eyon

d;H

angz

hou

Sum

gen

VE

GF

×P

D-1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Not

disc

lose

dN

A

BH

-292

2B

eijin

gH

anm

iE

GF

PD

-1P

recl

inic

alE

nhan

cetu

mor

imm

unit

yC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NA

BH

-295

0B

eijin

gH

anm

i;In

nove

ntH

er2

×P

D-1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

Fab

wit

hF

c,1

+1

NA

Duo

Bod

y-P

D-L

1x4-

1BB

Bio

NT

ech;

Gen

mab

PD

-L1

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

Fab

wit

hF

c,1

+1

NA

CD

X-5

27C

elld

exT

hera

peut

ics

PD

-L1

×C

D27

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

scF

vw

ith

Fc,

2+

2N

A

CB

-307

Cre

scen

doB

iolo

gics

PSM

4-1B

albu

min

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nSD

A+

SDA

+SD

A,

1+

1+

1N

A

ND

-021

CSt

one;

Num

abP

D-L

4-1B

albu

min

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nsc

Fv

+SD

A+

SDA

,1

+1

+1

NA

FS-

222

F-s

tar

PD

-L1

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

SDA

,2+

2N

A

EG

FR

/CT

LA

-4bi

spec

ific

mA

b2F

-sta

rE

GF

CT

LA

-4P

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+SD

A,2

+2

NA

IBI-

323

Inno

vent

Bio

logi

csP

D-L

LA

G-3

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nN

otdi

sclo

sed

NA

KY

-105

5K

ymab

PD

-L1

×IC

OS

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

SDA

wit

hF

c,2

+2

NA

1D8N

/CE

Ga1

Lea

dArt

isE

GF

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

scF

v+

SDA

,3+

3N

A

4-1B

Bx5

T4

Mac

roG

enic

s5T

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+F

vw

ith

Fc,

1+

2N

A

4-1B

BxH

ER

2M

acro

Gen

ics

Her

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+F

vw

ith

Fc,

1+

2N

A

PD

-L1x

4-1B

BM

acro

Gen

ics

Inc

PD

-L1

×4-

1BB

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

Fv

wit

hF

c,2

+2

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

26 Antibody Therapeutics, 2020

Tab

le2.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

ME

DI-

1109

Med

Imm

une

PD

-L1

×O

X40

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

PR

S-30

0se

ries

AP

ieri

sH

er2

×C

TL

A-4

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Tum

oror

tiss

uelo

caliz

atio

nN

otdi

sclo

sed

NA

PR

S-34

2P

ieri

sG

PC

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

SCA

FF

OL

D+

SCA

F-

FO

LD

wit

hF

c,2

+2

NA

PR

S-34

4P

ieri

s;Se

rvie

rP

D-L

4-1B

BP

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

umor

orti

ssue

loca

lizat

ion

Fab

+SC

AF

FO

LD

wit

hF

c,2

+2

NA

PD

-1×

BT

LA

Xen

cor

BT

LA

×P

D-1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,1

+1

NA

TX

B4-

BC

3O

ssia

nix

Inc

TfR

×P

D-L

1P

recl

inic

alE

nhan

cetu

mor

imm

unit

yT

roja

nho

rse

Fab

+SD

Aw

ith

Fc,

2+

2N

A

CB

A-0

710

Sorr

ento

cME

PD

-L1

Pre

clin

ical

Enh

ance

tum

orim

mun

ity,

anti

-tum

orig

enes

is

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

A

Tab

le3.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

tom

odul

ate

TM

Efo

rca

ncer

trea

tmen

t

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

Bin

traf

usp

alfa

Gla

xoSm

ithK

line,

Mer

ckK

GaA

PD

-L1

×T

GF

beta

Pha

seII

IM

odul

ate

TM

ET

umor

orti

ssue

loca

lizat

ion

Fab

+R

EC

EP

TO

Rw

ith

Fc,

2+

2N

CT

0406

6491

,N

CT

0384

0902

,N

CT

0383

3661

,N

CT

0363

1706

,N

CT

0384

0915

,N

CT

0269

9515

,N

CT

0251

7398

AG

EN

-142

3,G

S-14

23A

genu

s,G

ilead

CD

73×

TG

Fbe

taP

hase

IM

odul

ate

TM

EC

ombi

nato

rial

effe

ctN

otdi

sclo

sed

NC

T03

9547

04

SHR

-170

1Ji

angs

uH

engr

uiP

D-L

TG

Fbe

taP

hase

IM

odul

ate

TM

ET

umor

orti

ssue

loca

lizat

ion

Fab

+R

EC

EP

TO

Rw

ith

Fc,

2+

2N

CT

0371

0265

,N

CT

0377

4979

AK

-123

Ake

soB

ioph

arm

aP

D-1

×C

D73

Pre

clin

ical

Enh

ance

tum

orim

mun

ity,

mod

ulat

eT

ME

Tum

oror

tiss

uelo

caliz

atio

nN

otdi

sclo

sed

NA

Uni

TI-

101

Els

tar

The

rape

utic

sC

CR

CSF

1RP

recl

inic

alM

odul

ate

TM

EC

ombi

nato

rial

effe

ctF

ab+

Fab

wit

hF

c,1

+1

NA

Fm

Ab-

2B

ioco

n;IA

TR

ICa

EG

FR

×T

GF

beta

Pre

clin

ical

Mod

ulat

eT

ME

Tum

oror

tiss

uelo

caliz

atio

nF

ab+

RE

CE

PT

OR

wit

hF

c,2

+2

NA

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 27

Tab

le4.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

topr

omot

eta

rget

cell

depl

etio

nfo

rca

ncer

trea

tmen

t

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

Teb

enta

fusp

Imm

unoc

ore

gp10

0/H

LA

-A

∗ 020

CD

3P

hase

III

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

TC

R+

scF

v,1

+1

NC

T03

0703

92,

NC

T02

8898

61,

NC

T02

5703

08,

NC

T02

5350

78,

NC

T01

2112

62,

NC

T01

2096

76O

XS-

1550

,DT

-221

9G

TB

ioph

arm

aC

D19

×C

D22

Pha

seII

Tar

get

cell

depl

etio

nA

DC

scF

v+

scF

v,1

+1

NC

T00

8894

08,

NC

T02

3701

60A

FM

-13

Aff

imed

CD

16×

CD

30P

hase

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

2+

2N

CT

0122

1571

,N

CT

0232

1592

,N

CT

0266

5650

,N

CT

0319

2202

,N

CT

0407

4746

Odr

onex

tam

ab,

RE

GN

-197

9R

egen

eron

CD

CD

20P

hase

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T02

6516

62,

NC

T03

8881

05IM

C-C

103C

Gen

ente

ch;

Imm

unoc

ore

MA

GE

-A

4/H

LA

∗ A02

01×

CD

3P

hase

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tT

CR

+sc

Fv,

1+

1N

CT

0397

3333

IMC

nyes

oG

laxo

Smit

hKlin

e;Im

mun

ocor

eN

Y-E

SO-

1/H

LA

∗ A02

01×

CD

3P

hase

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tT

CR

+sc

Fv,

1+

1N

CT

0351

5551

Mos

unet

uzum

ab,

RG

-782

8G

enen

tech

,Roc

he,

Chu

gai

CD

CD

20P

hase

I/II

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0250

0407

,N

CT

0367

1018

,N

CT

0367

7141

,N

CT

0367

7154

OX

S-35

50,

CD

1615

33T

riK

EG

TB

ioph

arm

a,A

ltor

Bio

Scie

nce,

U.

Min

neso

ta

CD

16×

CD

33,I

L-1

5P

hase

I/II

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v+

LIG

AN

D,

1+

1+

1N

CT

0321

4666

GE

N-3

013

Gen

mab

CD

CD

20P

hase

I/II

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0362

5037

MC

LA

-117

Mer

usC

D3

×C

LE

C12

Pha

seI/

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T03

0382

30

Flo

tetu

zum

ab,

MG

D-0

06M

acro

Gen

ics,

Serv

ier

CD

CD

123

Pha

seI/

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

1+

1N

CT

0215

2956

,N

CT

0373

9606

MG

D-0

07M

acro

Gen

ics

CD

GPA

33P

hase

I/II

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

1+

1N

CT

0224

8805

,N

CT

0353

1632

RE

GN

-401

8R

egen

eron

,San

ofi

CD

MU

C16

Pha

seI/

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T03

5643

40

Cib

isat

amab

,R

O-6

9586

88,

RG

-780

2

Gen

ente

ch,R

oche

,C

huga

iC

D3

×C

EA

Pha

seI/

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T02

3242

57,

NC

T02

6507

13,

NC

T03

3376

98,

NC

T03

8662

39

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

28 Antibody Therapeutics, 2020

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

huG

D2-

BsA

bY

-mA

bsC

D3

×G

D2

Pha

seI/

IIT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

2+

2N

A

AM

G-7

01A

mge

nC

D3

×B

CM

AP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0328

7908

A−3

37G

ener

on(S

hang

hai)

CD

EpC

AM

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv,

1+

2C

ompa

nyde

velo

pmen

tpi

pelin

eA

MG

-160

Am

gen

CD

PSM

AP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0379

2841

AM

G-3

30,

MT

-114

Am

gen

CD

CD

33P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0252

0427

AM

G-4

24A

mge

nC

D3

×C

D38

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T03

4456

63

AM

G-4

27A

mge

nC

D3

×F

LT3

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T03

5413

69

AM

G-5

62A

mge

nC

D3

×C

D19

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T03

5718

28

AM

G-5

96A

mge

nC

D3

×E

GF

RvI

IIP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0329

6696

AM

G-6

73A

mge

nC

D3

×C

D33

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T03

2248

19

AM

G-7

57A

mge

nC

D3

×D

LL

3P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0331

9940

AM

V-5

64,

Tan

dAb

T56

4A

ffim

ed,F

red

Hut

ch,A

mph

iven

aC

D3

×C

D33

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

v,2

+2

NC

T03

1442

45,

NC

T03

5165

91A

PV

O-4

36A

ptev

oC

D3

×C

D12

3P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv

wit

hF

c,2

+2

NC

T03

6478

00

BI-

8369

09,

AM

G-4

20A

mge

n,B

oehr

inge

rIn

gelh

eim

CD

BC

MA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T02

5142

39,

NC

T03

8360

53R

G-6

026,

RO

-708

2859

Roc

heC

D3

×C

D20

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

2C

ompa

nyde

velo

pmen

tpi

pelin

eE

M-9

01,

CC

-932

69C

elge

neC

D3

×B

CM

AP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+2

NC

T03

4860

67

ER

Y-9

74C

huga

iC

D3

×G

PC

3P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T02

7488

37

GB

R-1

302

Gle

nmar

k,H

arbo

urB

ioM

edC

D3

×H

ER

2P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

CT

0282

9372

,N

CT

0398

3395

GB

R-1

342

Gle

nmar

kC

D3

×C

D38

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T03

3091

11

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 29

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

GE

M-3

33G

EM

oaB

,Cel

gene

CD

CD

33P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

CT

0351

6760

GE

M-3

PSC

A,

GE

M3P

SCA

GE

Moa

B,C

elge

neC

D3

×P

SCA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T03

9275

73

IGM

-232

3IG

MB

iosc

ienc

esC

D3

×C

D20

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+10

NC

T04

0829

36

JNJ-

6757

1244

,JN

J-12

44Ja

nsse

nR

esea

rch

&D

evel

opm

ent

CD

CD

33P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T03

9153

79

JNJ-

6370

9178

,JN

J-91

78Ja

nsse

nR

esea

rch

&D

evel

opm

ent

CD

CD

123

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0271

5011

JNJ-

6400

7957

,JN

J-79

57Ja

nsse

nR

esea

rch

&D

evel

opm

ent

CD

BC

MA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0314

5181

JNJ-

6389

8081

,JN

J-80

81Ja

nsse

nR

esea

rch

&D

evel

opm

ent

CD

PSM

AP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T03

9260

13

Orl

otam

ab,

MG

D-0

09M

acro

Gen

ics

CD

B7-

H3

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

1+

1N

CT

0262

8535

,N

CT

0340

6949

Pas

otux

izum

ab,

AM

G-2

12,

Am

gen,

Bay

erC

D3

×P

SMA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NC

T01

7234

75,

NC

T01

7234

75P

F-0

6671

008

Pfi

zer

CD

CD

H3

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

1+

1N

CT

0265

9631

PF

-068

6313

5,P

F-3

135

Pfi

zer

CD

BC

MA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0326

9136

RE

GN

-545

8R

egen

eron

,San

ofi

CD

BC

MA

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

CT

0376

1108

RG

-619

4,B

TR

C-4

017A

Gen

ente

chC

D3

×H

ER

2P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tN

otdi

sclo

sed

NC

T03

4480

42

TN

B-3

83B

Ten

eoB

io,A

bbV

ieC

D3

×B

CM

AP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

SDA

wit

hF

c,1

+2

NC

T03

9337

35

Xm

Ab-

1367

6,T

HG

-338

Xen

cor

CD

CD

20P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

CT

0292

4402

Xm

Ab-

1404

5,SQ

Z-6

22X

enco

r,N

ovar

tis

CD

CD

123

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T02

7303

12

Xm

Ab-

1808

7,X

EN

P-1

8087

Xen

cor

CD

SST

R2

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T03

4119

15

HP

N-4

24H

arpo

onC

D3

×P

SMA

×al

bu-

min

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

SDA

-SD

A-s

cFv,

1+

1+

1N

CT

0357

7028

M-8

02W

uhan

YZ

YB

ioph

arm

aC

D3

×H

ER

2P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

A

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

30 Antibody Therapeutics, 2020

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

JNJ-

6440

7564

Jans

sen

CD

GP

RC

5DP

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T04

1081

95,

NC

T03

3997

99R

G-6

160

Gen

ente

chC

D3

×F

cRH

5P

hase

IT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NC

T03

2751

03

NI-

1701

,TG

-180

1N

ovIm

mun

e,T

GT

hera

peut

ics

CD

19×

CD

47P

hase

IT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Fab

+F

abw

ith

Fc,

1+

1N

CT

0380

4996

MC

LA

-158

Mer

usE

GF

LG

R5

Pha

seI

Tar

get

cell

depl

etio

nF

cef

fect

orF

ab+

Fab

wit

hF

c,1

+1

NC

T03

5268

35

ZW

-49

Zym

ewor

ksH

ER

HE

R2

Pha

seI

Tar

get

cell

depl

etio

nA

DC

Fab

+sc

Fv

wit

hF

c,1

+1

NC

T03

8212

33

A-3

19G

ener

on(S

hang

hai)

CD

CD

19IN

DF

iled

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv,

1+

2N

CT

0405

6975

SAR

-440

234

Sano

fiC

D3

×C

D12

3Su

spen

ded

(1/2

)T

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fv

wit

hF

c,1

+1

NC

T03

5949

55

AF

M-1

1A

ffim

edC

D3

×C

D19

Susp

ende

d(1

)T

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

2+

2N

CT

0210

6091

,N

CT

0284

8911

cMet

×E

GF

RA

DC

Sorr

ento

EG

FR

×cM

ET

Pre

clin

ical

Tar

get

cell

depl

etio

nA

DC

Not

disc

lose

dN

A

AP

LP

HE

R2

AD

CR

egen

eron

AP

LP

HE

R2

Pre

clin

ical

Tar

get

cell

depl

etio

nA

DC

Fab

+F

abw

ith

Fc,

1+

1N

A

AB

P-1

50A

bpro

Cla

udin

18.2

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AB

P-1

10A

bpro

GP

C3

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AB

P-1

40A

bpro

;Luy

eC

EA

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AB

P-1

30A

bpro

;Luy

eC

D38

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AB

P-1

00A

bpro

;MSK

Can

cer

Cen

ter

Her

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

2+

2N

A

CD

16×

BC

MA

×C

D20

0A

ffim

edB

CM

CD

16×

CD

200

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

v+

Fv,

1+

2+

1N

A

AF

M-2

6A

ffim

edB

CM

CD

16P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

2+

2N

A

AF

M-2

4A

ffim

edE

GF

CD

16P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

2+

2N

A

AF

M-2

1A

ffim

edE

GF

RvI

II×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv,

2+

2N

A

B05

/CD

3A

ffim

ed;I

mm

atic

sM

MP

1-00

3/H

LA

-A

∗ 02

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

v,2

+2

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 31

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

CD

FLT

3A

lloge

ne;M

aver

ick;

Pfi

zer

FLT

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NA

Fol

-aC

D3

Am

brx

Fol

Ra

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+L

IGA

ND

wit

hF

cN

A

CD

MSL

NA

mge

nM

SLN

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NA

CD

H19

×C

D3

HL

EB

iTE

Am

gen

Cad

heri

n19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

vw

ith

Fc,

1+

1N

A

CD

EG

FR

Pb-

TC

BA

mge

n;C

ytom

XT

hera

peut

ics

EG

FR

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AM

X-1

68A

mun

ixE

pCA

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

A

AP

VO

-425

Apt

evo

The

rape

utic

sIn

cR

OR

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv

wit

hF

c,2

+2

NA

AR

B-2

01A

rbel

eC

adhe

rin-

17×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv

wit

hF

c,1

+1

NA

AVA

-012

Ava

cta

CD

22×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tN

otdi

sclo

sed

NA

CD

CD

19A

vact

aC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Not

disc

lose

dN

A

CD

CD

123

Bay

lor

Scot

t&

Whi

teR

esea

rch

Inst

itut

e

CD

123

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

vw

ith

Fc,

2+

2N

A

CD

HE

R2

Bei

jing

Han

mi

Her

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tN

otdi

sclo

sed

NA

CD

DL

L3

Boe

hrin

ger

Inge

lhei

mD

LL

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv

wit

hF

c,1

+1

NA

CC

W-7

02C

IBR

∗ ;Sc

ripp

sP

SMA

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+SM

OL

∗N

A

CB

A-1

535

Chi

ome

Bio

scie

nce

5T4

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv,

1+

2N

A

CT

X-4

419

Com

pass

The

rape

utic

sB

CM

NK

p30

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

2+

2N

A

CO

VA-4

231

Cov

agen

;Fre

dH

utch

CD

33×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

SCA

FF

OL

Dw

ith

Fc,

2+

2N

A

CD

EG

FR

vIII

Duk

eU

nive

rsit

yE

GF

RvI

II×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

A

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

32 Antibody Therapeutics, 2020

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

ESK

1E

urek

a;M

SKC

ance

rC

ente

r;N

ovar

tis

WT

1p/H

LA

-A

0201

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NA

FPA

-151

Fiv

eP

rim

eB

CM

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tN

otdi

sclo

sed

NA

CD

CD

79b

Gen

ente

chIn

cC

D79

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NA

CD

HE

R2

bipa

rato

pic

Gen

ente

chH

er2

bipa

rato

pic

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

ab+

Fab

wit

hF

c,1

+1

+1

NA

GB

R-1

372

Gle

nmar

kE

GF

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

A

PM

-CD

3-G

EX

Gly

coto

peT

A-M

UC

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

2+

2N

A

HP

N-2

17H

arpo

onB

CM

CD

albu

-m

inP

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tSD

A-S

DA

-scF

v,1

+1

+1

NA

HL

X-3

1H

enlix

;Hen

lixH

er2

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Not

disc

lose

dN

A

p95H

ER

2-T

CB

Hos

pita

lVal

lD

’Heb

ron;

MSK

Can

cer

Cen

ter;

Roc

he;U

.A

uton

oma

deB

arce

lona

Her

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+2

NA

E1-

3sIB

CP

harm

aceu

tica

ls;

Imm

unom

edic

s

Tro

p2×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+F

ab,1

+2

NA

CD

123/

CD

3bs

Ab

IGM

Bio

scie

nces

CD

123

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+10

NA

CD

38/C

D3

bsA

bIG

MB

iosc

ienc

esC

D38

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+10

NA

IPH

-61

Inna

teP

harm

a;Sa

nofi

TA

NK

p46

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+F

abw

ith

Fc,

1+

1N

A

CD

123-

CO

DV

-T

CE

INSE

RM

;San

ofi

CD

123

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

2+

2N

A

GN

R-0

47IB

CG

ener

ium

CD

19×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,2

+2

NA

JNJ-

0819

Jans

sen

Hem

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 33

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

9/V

δ2

TC

EG

FR

Lav

aE

GF

g9/d

2T

CR

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

SDA

+SD

A,1

+1

NA

CD

5T4

Mac

roG

enic

s5T

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,1

+1

NA

Nex

t-ge

nera

tion

CD

19×

CD

3D

AR

T

Mac

roG

enic

sC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

1+

1N

A

CD

123

×C

D3

DA

RT

Mac

roG

enic

sC

D12

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,1

+1

NA

Eph

A2x

CD

3D

AR

TM

acro

Gen

ics

Eph

a2×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,1

+1

NA

CD

IL13

Ra2

Mac

roG

enic

sIL

-13R

a2×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,1

+1

NA

CD

RO

R1

Mac

roG

enic

sR

OR

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

v+

Fv

wit

hF

c,1

+1

NA

CD

CD

133

McM

aste

rU

nive

rsit

y;U

nive

rsit

yof

Toro

nto

CD

133

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv,

1+

1N

A

h8F

4-B

iTE

MD

And

erso

nC

ance

rC

ente

rP

R1/

HL

A-A

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tsc

Fv

+sc

Fv,

1+

1N

A

ZW

-38

Mer

ck;Z

ymew

orks

CD

19×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

A

CD

HE

R2

Mol

ecul

arP

artn

ers

Her

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tSC

AL

FF

OL

D,1

+1

NA

CD

PSM

AR

egen

eron

PSM

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NA

CD

CD

20R

inat

-Pfi

zer

CD

20×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fab

wit

hF

c,1

+1

NA

CD

RO

R1

Scri

pps

Res

earc

hIn

stit

ute

RO

R1

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

vw

ith

Fc,

1+

1N

A

B-1

93Sh

ando

ngD

anho

ng;

Shan

ghai

Yan

yiC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Not

disc

lose

dN

A

CD

Sial

yl-T

nSi

amab

The

rape

utic

sSi

alyl

-Tn

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

19-3

-19

Spec

traM

abC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+2

NA

SV-2

02SY

SVA

XC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

SDA

+SD

A,1

+1

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

34 Antibody Therapeutics, 2020

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

SV-2

01SY

SVA

XH

er2

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

SDA

+SD

A,1

+1

NA

TN

B-5

85T

eneo

Bio

PSM

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

SDA

wit

hF

c,1

+1

or1

+1

+1

NA

TN

B-4

86T

eneo

Bio

CD

19×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

SDA

wit

hF

c,1

+1

NA

TN

B-3

81M

Ten

eoB

ioB

CM

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

SDA

wit

hF

c,1

+1

NA

CD

CD

19T

ianj

inC

hase

Sun

Jinb

oda

CD

19×

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tN

otdi

sclo

sed

NA

CD

MO

SPD

2V

BL

The

rape

utic

sM

OSP

D2

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

scF

v+

scF

v,1

+1

NA

M-7

01W

uhan

YZ

YE

pCA

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

scF

vw

ith

Fc,

1+

1N

A

Y-1

50W

uhan

YZ

YC

D38

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

CD

EM

P2

Wuh

anY

ZY

EM

P2

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

CD

EG

FR

Wuh

anY

ZY

EG

FR

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

CD

CD

19W

uhan

YZ

YC

D19

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

CD

CD

20W

uhan

YZ

YC

D20

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

MS-

133

Wuh

anY

ZY

CD

133

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

Xm

Ab-

1448

4X

enco

rP

SMA

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,1

+1

NA

YB

L-0

13Y

-Bio

logi

csP

D-L

CD

3P

recl

inic

alT

arge

tce

llde

plet

ion

Cyt

otox

icef

fect

oren

gage

men

tF

ab+

Fv,

1+

2N

A

huC

D33

-BsA

bY

-mA

bsC

D33

×C

D3

Pre

clin

ical

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fab

+sc

Fv

wit

hF

c,2

+2

NA

BI-

9057

11B

oehr

inge

rIn

gelh

eim

Cad

heri

n-17

×T

RA

IL-R

2P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

apop

tosi

sF

ab+

scF

vw

ith

Fc,

2+

2N

A

Nov

otar

gP

rom

ethe

raC

D20

×C

D95

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceap

opto

sis

Fab

+sc

Fv,

1+

1N

A

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 35

Tab

le4.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

cal

func

tion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

AB

P-1

60A

bpro

PD

-L1

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isN

otdi

sclo

sed

NA

BH

-29x

xB

eijin

gH

anm

iP

D-L

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Fab

+F

abw

ith

Fc,

1+

1N

A

IMM

-030

6G

atew

ayB

iolo

gics

;Im

mun

eOnc

oC

D20

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

HM

BD

-004

AH

umm

ingb

ird

CD

33×

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Fab

+sc

Fv,

1+

1N

A

HM

BD

-004

BH

umm

ingb

ird

BC

MA

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isF

ab+

Fab

wit

hF

c,1

+1

NA

IMM

-250

5Im

mun

eOnc

oP

D-L

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Fab

+L

IGA

ND

wit

hF

c,2

+2

NA

IMM

-260

11Im

mun

eOnc

oF

LT-3

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

IMM

-020

7Im

mun

eOnc

oV

EG

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

RE

CE

PT

OR

+L

IGA

ND

wit

hF

c,2

+2

NA

IMM

-290

2Im

mun

eOnc

oH

er2

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isF

ab+

LIG

AN

Dw

ith

Fc,

2+

2N

A

IBI-

322

Inno

vent

PD

-L1

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isN

otdi

sclo

sed

NA

NI-

1801

Nov

imm

une

MSL

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Fab

+F

abw

ith

Fc,

1+

1N

A

PT

-886

Pha

nes

The

rape

utic

sC

laud

in18

.2×

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Not

disc

lose

dN

A

PT

-217

Pha

nes

The

rape

utic

sD

LL

CD

47P

recl

inic

alT

arge

tce

llde

plet

ion

Enh

ance

phag

ocyt

osis

Not

disc

lose

dN

A

PM

C-1

22P

harm

Abc

ine

PD

-L1

×C

D47

Pre

clin

ical

Tar

get

cell

depl

etio

nE

nhan

ceph

agoc

ytos

isN

otdi

sclo

sed

NA

Duo

Hex

aBod

y-C

D37

Gen

mab

CD

37bi

para

topi

cP

recl

inic

alT

arge

tce

llde

plet

ion

Fc

effe

ctor

Fab

+F

abw

ith

Fc,

1+

1N

A

PM

-PD

L-G

EX

Gly

coto

peT

A-M

UC

PD

-L1

Pre

clin

ical

Tar

get

cell

depl

etio

nF

cef

fect

orF

ab+

scF

vw

ith

Fc,

2+

2N

A

CD

38×

IGF

-1R

I’ro

mG

roup

CD

38×

IGF

-1R

Pre

clin

ical

Tar

get

cell

depl

etio

nF

cef

fect

orsc

Fv

+sc

Fv

wit

hF

c,1

+1

NA

∗ CIB

R,C

alif

orni

aIn

stit

ute

for

Bio

med

ical

Res

earc

h;SM

OL

,sm

allm

olec

ule.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

36 Antibody Therapeutics, 2020

Tab

le5.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

for

infl

amm

ator

yco

ndit

ions

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

Ozo

raliz

umab

,T

S-15

2,P

F-5

2308

96,

AT

N-1

03

Sano

fi,T

aish

o,E

ddin

gpha

rmT

NF

×al

bum

inP

hase

III

Hal

f-lif

eex

tens

ion

Hal

f-lif

eex

tens

ion

SDA

+SD

A,1

+2

NC

T00

9590

36,

NC

T01

0071

75,

NC

T01

0638

03,

NC

T04

0775

67V

obar

ilizu

mab

Abb

Vie

;Abl

ynx

IL-6

albu

min

Pha

seII

Hal

f-lif

eex

tens

ion

Hal

f-lif

eex

tens

ion

SDA

+SD

A,1

+1

NC

T02

5186

20,

NC

T02

4378

90,

NC

T02

3093

59,

NC

T02

2879

22R

omilk

imab

,SA

R-1

5659

7Sa

nofi

IL-4

×IL

-13

Pha

seII

Com

bina

tori

alef

fect

Com

bina

tori

alef

fect

Fab

+F

vw

ith

Fc,

2+

2N

CT

0152

9853

,N

CT

0234

5070

,N

CT

0292

1971

M-1

095,

AL

X-0

761

Avi

llion

;Mer

ckSe

rono

IL-1

7A×

albu

min

×IL

-17F

Pha

seII

Com

bina

tori

alef

fect

Com

bina

tori

alef

fect

SDA

+SD

A,1

+1

+1

NC

T03

3847

45,

NC

T02

1564

66M

GD

-010

,P

RV

-327

9M

acro

Gen

ics,

Pro

vent

ion

CD

32B

×C

D79

BP

hase

I/II

Dom

inan

tne

gati

veD

omin

ant

nega

tive

Fv

+F

vw

ith

Fc,

1+

1N

CT

0237

6036

AM

G-5

70,

ME

DI-

0700

Am

gen,

Vie

laB

io,

Ast

raZ

enec

aB

AF

ICO

SLP

hase

IC

ombi

nato

rial

effe

ctC

ombi

nato

rial

effe

ctF

ab+

PE

PT

IDE

wit

hF

c,2

+2

NC

T02

6189

67,

NC

T03

1560

23,

NC

T04

0580

28T

ibul

izum

abE

liL

illy

BA

FF

×IL

-17A

Pha

seI

Com

bina

tori

alef

fect

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,2

+2

Com

pany

deve

lopm

ent

pipe

line

JNJ-

6117

8104

Jans

sen

Res

earc

h&

Dev

elop

men

tT

NF

×IL

-17A

Pha

seI

Com

bina

tori

alef

fect

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

CT

0275

8392

ON

O-4

685

Ono

CD

PD

-1P

hase

ID

omin

ant

nega

tive

Dom

inan

tne

gati

veF

ab+

Fab

wit

hF

c,1

+1

NC

T04

0790

62

ES-

210,

AP

VO

-210

Apt

evo

The

rape

utic

sC

D86

-IL

10P

hase

IT

issu

esp

ecif

icit

yT

umor

orti

ssue

loca

lizat

ion

scF

v+

scF

vw

ith

Fc,

2+

2N

CT

0376

8219

CD

19×

CD

11c

Nat

iona

lJew

ish

Hea

lth

CD

19×

CD

11c

Pre

clin

ical

Tar

get

cell

depl

etio

nF

cef

fect

orF

ab+

Fab

wit

hF

c,1

+1

NA

AM

-201

AbC

lon

IL-6

TN

FP

recl

inic

alA

nti-

infl

amm

atio

nC

ombi

nato

rial

effe

ctF

ab+

SCA

FF

OL

Dw

ith

Fc,

2+

2N

A

IL4R

alph

a/IL

-5bs

Ab

arG

EN

-XIL

-4R

IL-5

Pre

clin

ical

Ant

i-in

flam

mat

ion

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,2

+2

NA

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 37

Tab

le5.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

linic

alst

udie

s

BH

-165

7B

eijin

gH

anm

iT

NF

×IL

-17A

Pre

clin

ical

Ant

i-in

flam

mat

ion

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1N

A

IL-4

×IL

-13

Bei

jing

VD

JBio

IL-4

×IL

-13

Pre

clin

ical

Ant

i-in

flam

mat

ion

Com

bina

tori

alef

fect

Not

disc

lose

dN

AIL

-1×

TN

Bei

jing

VD

JBio

IL-1

×T

NF

Pre

clin

ical

Ant

i-in

flam

mat

ion

Com

bina

tori

alef

fect

Not

disc

lose

dN

AC

MX

-02

Com

plix

TN

IL-2

3P

recl

inic

alA

nti-

infl

amm

atio

nC

ombi

nato

rial

effe

ctF

ab+

SCA

FF

OL

Dw

ith

Fc,

2+

2N

A

ND

-016

Inta

rcia

TN

IL-

17A

×al

bum

inP

recl

inic

alA

nti-

infl

amm

atio

nC

ombi

nato

rial

effe

ctF

v+

Fv

+F

v,1

+1

+1

NA

MT

-619

4M

itsu

bish

iTan

abe

Pha

rma

IL-6

IL-1

7AP

recl

inic

alA

nti-

infl

amm

atio

nC

ombi

nato

rial

effe

ctF

ab+

SCA

FF

OL

Dw

ith

Fc,

2+

2N

A

YB

L-0

04Y

-Bio

logi

csT

NF

×IL

-17A

Pre

clin

ical

Ant

i-in

flam

mat

ion

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,2

+2

NA

PT

-001

Pan

dion

MA

dCA

PD

-1P

recl

inic

alA

nti-

infl

amm

atio

nT

umor

orti

ssue

loca

lizat

ion

Fab

+sc

Fv

wit

hF

c,2

+2

NA

AL

XN

-172

0A

lexi

onC

albu

min

Pre

clin

ical

Hal

f-lif

eex

tens

ion

Ext

ende

dha

lf-l

ife

scF

v+

scF

v,1

+1

NA

SDA-based. Two different VHHs can be fused to forma bsAb [4]. This format may be the smallest bsAb formatwith molecular weight approximately 25 KD. It has beenreported that two different VHHs can be fused to coiled-coil peptide to form Combody. The peptide facilitates theoligomerization of the antibody and renders the antibodyavidity effect [5]. Two different VHH can also be engineeredon the N-terminus of CH1 and CL to form a Fab-like 1 + 1bsAb fragment [6].

ScFv-based. Bispecific T cell engager (BiTE), one ofthe formats used to redirect T cells to tumor cells, com-prises two tandem linked scFvs: one scFv against a tumor-associated antigen and another binding to CD3 on T cells.The structure and mechanism of BiTE was well reviewed byWolf [7]. Two scFvs can also be indirectly linked, such asvia a CL, to form a bsAb in scFv-CL-scFv format [8]. Dueto aggregative tendency of scFv, various techniques wereemployed to stabilize scFv. Brolucizumab (Beovu) was engi-neered using scFv-λcap platform [9]. Similar technologywas also used to build multispecific antibody (msAb)-basedtherapeutics by cognate heterodimerization (MATCH) [10],where up to four distinct binding sites can be integrated intoa multispecific Fv- or scFv-based molecule.

Fv-based. A diabody molecule is formed by two polypep-tides: one polypeptide contains VHa and VLb; anotherpolypeptide contains VHb and VLa. Due to the shortlinker, VHa associates with VLa on another polypeptideand similarly VLb associates with VHb to form 1 + 1 bsAbfragment. A diabody-based bispecific format is called dual-affinity retargeting antibody or DART [11–13]. DARTmolecules may contain Fc domain to extend in vivo half-life and grand effect functions. TandAb is another Fv-basedbispecific fragment: two polypeptides are forced to fold ina head-to-tail fashion to form 2 + 2 bsAb fragment [14].

Combination. In a native antibody, VH and VL are onthe N-terminus of Fab region and CH1 and CL on C-terminus. It was found that CH1 and CL can also facilitatethe association of VH and VL on C-terminus of a Fab-Fv fusion protein. This Fab directed VH-VL associationcan be further improved by introducing a disulfide bondbetween the VH and VL on C-terminus [15]. A VH on theC-terminus of a Fab-Fv may associate with a C-terminalVL on another Fab-Fv to form 2 + 2 tetramer Fab-Fv[16]. Similarly, a scFv can be fused on the C-terminus of aFab to form Fab-scFv fusion proteins. The so-called bibodyhas one Fab with one scFv, and “tribody” has one Fabwith two scFvs [17]. A tribody can be either bispecific ortrispecific, depending on the specificity of the two attachedscFvs. A VHH can be fused to a light chain C-terminus ofa Fab to form 1 + 1 bispecific antibody fragment [18]. Itwas reported that three tandem linked VHHs can be fusedwith a scFv to form 1 + 3 bispecific fragment [19]. A bsAbfragment containing a VHH or scFv specific to humanserum albumin is a common strategy to extend serum half-life of such molecules.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

38 Antibody Therapeutics, 2020

Tab

le6.

Pro

gram

sin

clin

ical

and

prec

linic

alst

ages

for

othe

rco

ndit

ions

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

ondi

tion

sC

linic

alst

udie

s

Far

icim

ab,

RG

-771

6,R

O-6

8674

61

Roc

he,C

huga

iP

harm

aceu

tica

lV

EG

AN

GP

T2

Pha

seII

IA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1O

cula

r,di

abet

icre

tino

path

yN

CT

0194

1082

,N

CT

0248

4690

,N

CT

0269

9450

,N

CT

0303

8880

,N

CT

0362

2580

,N

CT

0362

2593

,N

CT

0382

3287

,N

CT

0382

3300

IBI-

302

Inno

vent

VE

GF

×co

mpl

e-m

ent

Pha

seI

Ant

i-an

giog

enes

is;

anti

-inf

lam

mat

ion

Com

bina

tori

alef

fect

Not

disc

lose

dO

cula

rN

CT

0381

4291

Gre

mub

amab

,M

ED

I390

2M

edIm

mun

eP

crV

×P

sIP

hase

IIC

ombi

nato

rial

effe

ctC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2A

ntib

acte

rial

NC

T02

2557

60,

NC

T02

6969

02M

ED

I-73

52A

stra

Zen

eca

NG

TN

FP

hase

IIC

ombi

nato

rial

effe

ctC

ombi

nato

rial

effe

ctsc

Fv

+R

EC

EP

-T

OR

wit

hF

c,2

+2

Ana

lges

icdr

ugs

NC

T02

5081

55,

NC

T03

7559

34

10E

8.4/

iMab

Tai

Med

,Aar

onD

iam

ond

AID

SR

esea

rch

Cen

ter

HIV

-1E

nv×

CD

4P

hase

IB

road

enpr

otec

tion

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1H

IV-1

NC

T03

8752

09

SAR

-441

236

Sano

fi,N

IHH

IV-1

Env

trip

arat

opic

Pha

seI

Com

bina

tori

alef

fect

Com

bina

tori

alef

fect

Fab

+F

vw

ith

Fc,

1+

1+

1H

IV-1

NC

T03

7051

69

MG

D-0

14M

acro

Gen

ics,

NIA

IDC

D3

×H

IV-1

Env

prot

ein

Pha

seI

Tar

get

cell

depl

etio

nC

ytot

oxic

effe

ctor

enga

gem

ent

Fv

+F

vw

ith

Fc,

1+

1H

IV-1

NC

T03

5709

18

BF

KB

-848

8A,

RG

-799

2G

enen

tech

FG

FR

beta

-K

loth

oP

hase

IT

issu

esp

ecif

icit

yT

umor

orti

ssue

loca

lizat

ion

scF

v+

scF

vw

ith

Fc,

1+

1D

iabe

tes

NC

T02

5933

31,

NC

T03

0605

38A

BP

-201

AbM

edV

EG

AN

GP

T2

Pre

clin

ical

Ant

i-an

giog

enes

isC

ombi

nato

rial

effe

ctF

ab+

scF

vw

ith

Fc,

2+

2O

cula

rN

A

SL-6

34U

nive

rsit

yof

Col

orad

oSy

stem

VE

GF

×A

NG

PT

2P

recl

inic

alA

nti-

angi

ogen

esis

Com

bina

tori

alef

fect

Not

disc

lose

dO

cula

rN

A

KSI

-501

Kod

iak

Scie

nces

VE

GF

×IL

-6P

recl

inic

alA

nti-

angi

ogen

esis

,an

ti-i

nfla

mm

atio

nC

ombi

nato

rial

effe

ctF

ab+

RE

CE

P-

TO

Rw

ith

Fc,

2+

2

DM

E,U

veit

isN

A

FIT

-1H

umab

sB

ioM

edZ

ika

Epr

otei

nbi

para

topi

cP

recl

inic

alB

road

enpr

otec

tion

Bip

arat

opic

Fab

+F

abw

ith

Fc,

2+

2In

fect

ion

NA

TM

B-b

ispe

cifi

cA

aron

Dia

mon

dA

IDS

Res

earc

hC

ente

r;T

aiM

ed

HIV

gp12

CD

4P

recl

inic

alB

road

enpr

otec

tion

Com

bina

tori

alef

fect

Fab

+F

abw

ith

Fc,

1+

1In

fect

ion

NA

VIS

-RSV

Vis

terr

a,V

irB

iote

chno

logy

RSV

RSV

GP

recl

inic

alB

road

enpr

otec

tion

Com

bina

tori

alef

fect

Fab

+sc

Fv

wit

hF

c,1

+1

Infe

ctio

nN

A

Con

tinu

ed

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 39

Tab

le6.

Con

tinu

ed

Ant

ibod

yna

me

Org

aniz

atio

nT

arge

tsH

ighe

stph

ase

Bio

logi

calf

unct

ion

Typ

eof

mec

hani

smF

orm

atC

ondi

tion

sC

linic

alst

udie

s

Dua

lFZ

Dan

dL

RP

5/6

agon

ist

Ant

lerA

The

rape

utic

sF

ZD

×L

RP

5/6

bipa

rato

pic

Pre

clin

ical

Cof

acto

rm

imet

icC

ofac

tor

mim

etic

Fv

+F

vw

ith

Fc,

2+

1+

1T

issu

ere

pair

NA

AB

L-3

01A

BL

Bio

alph

a-sy

nucl

ein

Pre

clin

ical

Neu

tral

izin

gpa

thog

enic

targ

etT

roja

nho

rse

Fab

+sc

Fv

wit

hF

c,2

+2

Neu

rolo

gy/P

sych

iatr

icN

A

AT

V:B

AC

E1/

Tau

Den

ali

TfR

×B

AC

E1

orT

fR×

Tau

Pre

clin

ical

Neu

tral

izin

gpa

thog

enic

targ

etT

roja

nho

rse

Fab

+F

ab+

SDA

wit

hF

c,1

+1

+1

Neu

rolo

gy/P

sych

iatr

icN

A

KY

-104

9K

ymab

Ltd

FIX

×F

XP

recl

inic

alC

ofac

tor

mim

etic

Cof

acto

rm

imet

icF

ab+

Fab

wit

hF

c,1

+1

Hem

atol

ogic

NA

Bis

peci

fic

fully

hum

anIg

GSh

ire

plc

FIX

×F

XP

recl

inic

alC

ofac

tor

mim

etic

Cof

acto

rm

imet

icF

ab+

Fab

wit

hF

c,1

+1

Hem

atol

ogic

NA

Fc-containing bispecific antibodies

The Fc region, part of natural antibody, is homodimerof two polypeptide chains. Depending on the isotype ofthe antibody, each comprising two to three heavy chainconstant domains (CH2, CH3, and CH4). The Fc regionnot only imparts an antibody effector functions due toFcγ R binding and complement-binding but also extendsits in vivo half-life via FcRn binding.

When two different heavy chains and two differentlight chains of IgG antibodies are expressed in onecell, these different heavy chains and light chains mayscramble randomly, possibly to form 10 different IgGantibody molecules. Among these, statistically only 12.5%would be desired bsAb. For symmetric Fc-containingbispecific formats, a challenge is to avoid heavy chain/lightchain mispairing. For asymmetric Fc-containing bispecificformats, an additional challenge is to force heterodimericheavy chain formation.

Forming heterodimeric Fc can be achieved by engineer-ing the interface of two CH3 (for IgG) or CH4 (for IgMor IgE) domains, by changing size (knob-into-hole) [20–22] and electrostatic steering [23]. Computational methodshave been used to identify the mutations that facilitate het-erodimeric association [24, 25]. Several groups also used theinterface of different Ig proteins to design the heterodimericFc. Davis et al. developed derivatives of human IgG andIgA CH3 domains composed of alternating segments ofhuman IgA and IgG CH3 sequences [26]. Skegro et al.grafted some residues from T cell receptor (TCR) constantregion to CH3 of IgG1 or CH4 of IgM [27]. An alter-native strategy is to purify heterodimer from unwantedhomodimers. With the modification on CH3 domain, theheterodimer and homodimer have different affinity bindingon Protein A, and the bsAb with heterodimeric Fc can beisolated [28].

In order to ensure cognate heavy chain and light chainpairing, several strategies have been reported. The firststrategy is to use SDA, scFv, or scFab as antigen-bindingbuilding blocks. In addition, single-chain IgG has beenreported [29], where two heavy chains, two light chains,and three linker sequences were expressed from one gene.Protease cleavage sites were integrated into these linkers,allowing protease digestion to cleave the linkers. The secondstrategy is using common light chain or common heavychain. In the case of common light chain, an identical lightchain is used as the partner for two different heavy chains[30–33]. Common heavy chain was also reported in a κλ-body case [34]. The third strategy is to modify the interfaceof VH-CH1 and VL-CL, including developing orthogonalFab interface [35, 36], altering the location of interchaindisulfide bond [37–39], and addition of charged pairs andknob-into-hole [40, 41]. These strategies usually involvechanges on both VH-VL interface and CH1/CL interface.Yet, Bonish et al. reported that the preferential associationcan be achieved by only engineering the CH1/CL interface[42]. To associate cognate VH and VL, the CH2 domainfrom IgM or IgE have been engineered to form heterodimerto replace CH1/CL [43] [Dong, WO2017011342].

There are additional strategies to avoid heavy chain/lightchain mispairing. Schaefer et al. described a CrossMab

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

40 Antibody Therapeutics, 2020

Figure 2. The classification of bsAb formats based on assembly of antibody fragments as building blocks. The first row and column list the five basicbuilding blocks (SDA, Fv, scFv, Fab, scFab). The different color and shape of VH and VL represent their origins from different parental antibodies. Theassembly of different building blocks creates various bsAb formats classified into 30 groups. An exemplary format and its molecular weight of each groupare listed. The diagonal line divides the formats into bispecific formats without Fc (top right with number 1-15) and bispecific formats with Fc (bottomleft with number 16-30): 1, tandemly linked SDAs; 2, a SDA tandemly linked on the VH of a Fv; 3, a SDA tandemly linked on the VH of a scFv; 4,two SDAs are separately linked on the carboxyl-terminus of constant domain of a Fab; 5, a SDA tandemly linked on the VL of a scFab; 6, the VHs andVLs of two Fvs cross pair to each other to form diabody; 7, a scFv tandemly linked on the VH of a Fv; 8, the VH and VL of a Fv each linked on thecarboxyl-terminus of CH1 and CL of a Fab; 9, the VH of a Fv linked to the CH1 of a scFab; 10, two tandemly linked scFvs; 11, a scFv linked on the VHof a Fab; 12, a scFab

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 41

approach: exchange of heavy chain and light chain domainswithin the Fab of one half of the bsAb [44]. Moore et al.described a Mab-Fv approach: the first pair of variableregions was present on the regular position of an IgG. Thesecond pair variable region VL and VH were fused to theC-terminus of the heterodimeric heavy chain, respectively,to form a 1 + 2 bsAb [24]. A similar 1 + 2 bsAb formatwith protease-inducible activity was also reported [45].

Recently, a versatile bispecific platform namedWuXiBody

®has been developed [Xu, WO2019057122A1].

The CH1/CL domains of one of the two parental antibodiesare replaced by stabilized TCR α and β constant regions(Cα/Cβ) to ensure the cognate light chain-heavy chainpairing. This approach has been tested on more than100 pairs of antibodies and compatible with most of theantibody pairs (data not shown).

Below are some representative examples of Fc-containingbispecific formats with SDA, Fv, scFv, Fab, and scFab.There are more examples that use the combination of thesefive building blocks.

SDA-based. As a small, flexible, and modular antigen-binding site, it is obvious that SDA or VHH can be fusedto N-terminus or C-terminus of heavy chain or light chainof an IgG antibody. As shown in Fig. 3, SDA can alsobe inserted in to other fusion sites. Shen et al. reportedthat a SDA antibody can be fused with VL to form 2 + 2bispecific format [46, 47]. Shi et al. used two different SDAsto replace VH and VL of an IgG, respectively, to form 2 + 2biparatopic antibody [48]. In addition to variable domain,CH3 domain on Fc has been engineered as binding site[49]. Broadly speaking, the engineered CH3 is a SDA thatcan be integrated into a bispecific format, such as IgG-likeBsAb [50].

Fv-based. VH and VL domains are tandemly linked withVH and VL of another IgG antibody, to form a 2 + 2bispecific IgG format, named as “dual variable domainimmunoglobulin” or DVD-Ig [51]. Fab + Fv-based 1 + 2format called mAb-Fv [24] was mentioned above. Fv canalso replace CH2 domain to form a 1 + 2 bispecific formatcalled TriFab [52]. Seifert et al. employed diabody formatcombined with heterodimeric CH2 from IgM or IgE toconstruct 2 + 2 bispecific format [43]. AforementionedWuXiBody

®formats are also Fv-based formats, including

1 + 1, 1 + 2, and 2 + 2 formats.

ScFv-based. There are many Fc-containing bispecificformats comprising of scFv, although scFv is prone to

aggregate. A scFv can be fused with heavy chain of IgG toform symmetric 2 + 2 bsAb [53], called Morrison format.Morrison format is one of the earliest bispecific formatsthat have still been widely used. ScFv can also be fusedwith light chain [54]. To form 1 + 1 format, several groupsused scFv to replace one of the Fabs on an IgG and usedengineered Fc heterodimer [55, 56]. Two different scFvs canbe used to replace both Fabs on an IgG antibody to form1 + 1 bispecific format [57, 58]. ScFv can also be placedin the hinge region or CH3 domain to form 2 + 2 formats[59, 60]. Kim et al. fused scFv with CH1-CH2-CH3 and co-expressed LC domain, potentially masking the hydropho-bic part of scFv to make the molecule more stable [61].

Fab/scFab-based. Several Fab- or scFab-based bispecificformats have been reported. Fab-based CrossMab, a 1 + 1bispecific IgG format [44], was mentioned above. Fabs-in-tandem immunoglobulin (FIT-Ig) is a format where a Fab isfused to the N-terminus of another IgG antibody: the lightchain of the Fab is fused with the heavy chain of the IgG,and FD chain of the Fab and light chain of the IgG areseparate polypeptides. These three different polypeptidescan be co-expressed from single cells and be assembled toform the bsAb [62]. In theory, the FD of antibody A andthe light chain of antibody B can associate to each otherto form a hybrid Fab, but this hybrid Fab can be removedin Protein A purification step. Bostrom et al. described atwo-in-one bsAb, a bsAb in regular IgG form, and eacharm can bind two distinct antigens [63–65]. Hu et al. evendeveloped a four-in-one antibody [66]. Strop et al. showedthat making mutations in hinge region and CH3 domain ofhuman IgG1 and IgG2 could facilitate heterodimerizationof heavy chain. Two parental antibodies can be expressedand purified separately and mixed together under appro-priate redox conditions, resulting in formation of a stablebsAb [67]. Labrijn et al. reported a similar platform, latercalled DuoBody [68]. scFab can be used to construct 1 + 1format [69], and it can also be used as one of the buildingblocks to construct tetraspecific antibody [70].

Other binding modalities

As mentioned earlier, peptides, ligands, receptors, anddifferent protein scaffolds, either native form or engineeredform, can be used as antigen-binding sites. The non-antibody scaffolds include Adnectin, DARPin, Affilins,alpha helix scaffold, avimer, Centyrin, Duocalin, Ecallan-tide, Fynomer, microprotein, peptide, Protein A domain,trimeric, TCR, etc. There are numerous possibilities to

tandemly linked with a scFv; 13, two tandemly linked Fabs: the light chain of one Fab linked with the heavy chain of another Fab and vice versa; 14, ascFab linked on the VH of a Fab; 15, tandemly linked two scFab; 16, two tandemly linked SDA on Fc to form homodimer; 17, a SDA and the VH of a Fvlinked to the FcA to pair with the VL of a Fv linked to another FcB to form heterodimer; 18, a diabody on FcA to pair with FcB to form heterodimer;19, a SDA on FcA to pair with a scFv on FcB to form heterodimer; 20, a scFv and the VH of a Fv linked to FcA to pair with the VL of the Fv linked withFcB to form heterodimer; 21, two scFv tandemly linked to the amino- and carboxyl-terminus of a Fc to form homodimer; 22, a SDA tandemly link to thelight chain of a IgG to form homodimer; 23, a TCR constant domain anchored Fv linked to FcA to pair with a half IgG with FcB to form heterodimer(WuXiBodyTM); 24, a scFv linked FcA to pair with a half IgG with FcB to form heterodimer; 25, two tandemly linked Fabs (the light chain of a Fab linkedon the heavy chain of another Fab) on Fc to form homodimer (FIT-Ig); 26, a SDA on FcA to pair with a scFab on FcB to form heterodimer; 27, a scFaband the VH of a Fv linked on FcA to pair with the VL linked on FcB to form heterodimer; 28, a scFv linked on FcA to pair with a scFab linked on FcBto form heterodimer; 29, a half IgG with FcA paired with scFv linked on FcB to form heterodimer; 30, two scFab each linked on FcA and FcB to formheterodimer. Above mentioned FcA and FcB are engineered Fc pair to facilitate Fc heterodimerization.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

42 Antibody Therapeutics, 2020

Figure 3. Fusion sites for antigen-binding building blocks. A) A heterodimeric Fc fragment has at least six fusion sites: amino-terminus (1 and 4), carboxyl-terminus (3 and 6) of Fc and between CH2 and CH3 (2 and 5). B) The fragment made of heterodimeric Fc and two differently heterodimerized CL-CH1domains provides at least twelve fusion sites: amino-terminus of CL (1 and 7) and CH1 (3 and 9), carboxyl-terminus of CL (2 and 8) and CH3 (6 and 12),hinge region (4 and 10), between CH2 and CH3 (5 and 11).

generate bispecific formats using these binding modalities.Recent examples include peptide [71], VEGF receptor [72],TCR [73], and single-chain TRAIL [74].

In the new paradigm of bsAbs, many novel formatshave been designed and tested. The general goal is todesign a molecule to enable novel therapeutic mechanismsand to make homogeneous product in large scale to meetthe need of clinical development and commercial manu-facturing. Additionally, more multispecific formats havebeen reported in the recent years, including trispecific [75],tetraspecific [70], and pentaspecific [71].

THE RESURGENCE OF BISPECIFIC ANTIBODIES

During the last few years, the number of clinical stud-ies using bsAbs has increased exponentially (Fig. 4A). Infact, this increase in 2014 matched with the launch ofBlincyto (Amgen), the first commercialized BiTE for thetreatment of acute lymphoblastic leukemia. However, it wasnot until 2017 that another bsAb, Hemlibra (Roche), waslaunched for the treatment of hemophilia A. Currently,most bsAbs in clinical development are in early studies(67% in Phase I, 25% in Phase II) with only five productsin Phase III studies (Fig. 4B). The majority of clinical stagebsAbs (∼84%) are designed to treat cancer, especially solidtumors, breast cancer, and acute myeloid leukemia. Nev-ertheless, there are some products designed to treat otherconditions such as rheumatoid arthritis or autoimmunediseases (Fig. 4C). The company with more bsAbs underactive development is Amgen, followed by MacroGenicsand then Lilly, Janssen, Roche, Sanofi, and Xencor. Thestrategy in nearly half of the developing bispecific productsis to deplete the malignant cells by engaging cytotoxic effec-tor cells including T or natural killer (NK) cells using CD3or FcGR3A (CD16) targeting arms. Another commonlyused strategy is to identify tumor or tissue-specific markersto act only in the affected areas. For that, many companieshave designed their own technology to manufacture bsAbs,including Amgen’s BiTE, MacroGenics’ DART, or Roche’sCrossMab platforms.

Accordingly, with the increase in bsAb development, thenumber of deals (excluding mergers and acquisitions) havefocused on clinical stage bsAbs resulting in an increaseof 140% in the last 3 years, while the total disclosed dealvalue decreased by 50%, from $3.2 billion to $1.6 billion;$6.8 billion is recorded across the whole period (Fig. 5A).From these deals, nine were worth more than $100 million,and most were structured with milestones, signifying thebalancing of risk and reward between the partners. Sanofiand Regeneron’s 2015 co-development agreement focusedon various antibodies, including CD3 × MUC16 (REGN-401) and CD3 × BCMA (REGN-5458) (Fig. 5B). Fromthese deals, $3.8 billion were spent on oncology followedby $1 billion on infection diseases from a total of $6.8billion, probably due to the clinical and commercial poten-tial of treating patients in these disease areas with bsAbs(Fig. 5C).

The global market of bsAb was worth $0.46 billion in2018, which was dominated almost equally by Blincyto($230 million) and Hemlibra ($229 million). As predicted,the market for Hemlibra will boom in the next few yearsand grow up to $3.96 billion by 2024. Instead, Blincytowill only have a moderate increase. With the massive salesgrowth of Hemlibra and the potential approval of newentrants, for instance, faricimab, gremubamab, MCLA-117, and XmAb-14045, the bsAb market will surpass $5.43billion in 2024 (Fig. 5D).

BIOLOGY DRIVES DEVELOPMENT OF VARIOUSBISPECIFIC ANTIBODIES

Most human diseases are complex, often driven by multipleredundant or distinct mechanisms; thereby single-targetapproach such as mAb may not be sufficient to achieveoptimal therapeutic efficacy. Especially, many therapeuticconcepts need physical linkage of two or more targets. Inthis case, bsAbs or msAbs targeting two or more targetsmay offer novel therapeutic applications that are difficultto achieve by mAbs. In a recent comprehensive reviewarticle, Aran Labrijn, Maarten Janmaat, Janice Reichert,

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 43

Figure 4. Statistics showing the booming of bsAb programs. A). The number of clinical studies associated with bsAb in the past fourteen years (up toSeptember 2019). The bsAb programs classified based on B) different clinical stages and C) different disease areas. Data source: CortellisTM CompetitiveIntelligence (CCI) and CortellisTM Drug Discovery Intelligence (CDDI, formerly Integrity) as of Sept 23, 2019.

Figure 5. Licenses and market analysis for bsAb programs. A). Licenses for clinical stage bsAbs. Line represents number of license signed each year. Blueand yellow bars represent the largest deal and total deal values for each year, respectively. B). The largest deals signed from 2014 to 2018. C). Deal valuesin disease area. D). BsAb market size in 2018 and forecast in 2024. Data source: CortellisTM CCI as of Sept 23, 2019.

and Paul Parren thoroughly reviewed global bispecific anti-body clinical pipeline using a mechanistic lens [2]. Basedon the analysis using CortellisTM, a Clarivate Analyticssolution, by the end of September 2019, there are 176bsAbs or bifunctional proteins with target disclosed underactive preclinical development, compared to 119 in clinicaldevelopment for cancer, autoimmune, and other indica-

tions (including IND filed—Phase III—and two programson clinical hold). This highlights the increased interest ofexploring bsAbs as a venue to develop novel antibody-based therapeutics. The most frequently studied target pairsof those bispecific antibodies and the number of moleculesbeing explored are illustrated in a network graph (Fig. 6).We took a step further and analyzed the disease areas cov-

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

44 Antibody Therapeutics, 2020

Figure 6. A network graph characterizing the target pairs of most bispecific programs in both preclinical and clinical investigations. Each node in thenetwork is one target, and each edge connecting two nodes represents one bispecific program. The circular edges are biparatopic programs. The node sizeshows the degree of a particular target being paired with other different targets. The colors of the edges are marked in black if only one program is availablefor that particular pair, otherwise in red if more than one are being explored. The popularity of that bispecific program is reflected from the thickness ofthe red edges. Source data are from CortellisTM (Table 1-4). Tri-specific and albumin-relevant bispecific programs are not included. The albumin-relevanttri-specific are analyzed as bispecific projects.

ered and mechanisms employed by these clinical moleculesand preclinical drug development candidates as well, withthe attempt to illustrate the principle of selecting a bispe-cific format to meet biology needs and selecting a bispecificmolecule as a clinical development candidate by six criticalcriteria.

Bispecific antibodies for cancer treatment

According to the CortellisTM’ analysis, the bsAb pipelineis composed predominantly by programs for cancertreatment, with 99/119 programs in clinical stages and153/176 preclinical programs (Fig. 4C). As reviewed byHanahan D. and Weinberg RA., there are eight hallmarksof cancer, and targeting these biological capabilities ofcancer cells may lead to new therapeutic options for cancer[76]. Therefore, based on the biological functions, wecategorize the bsAb programs into the following groups:anti-angiogenesis, anti-tumorigenesis, enhancing tumorimmunity, modulating tumor microenvironment (TME),and depletion of target cells.

Anti-angiogenesis. As angiogenesis plays an essentialrole in promoting tumor progression and metastasis, anti-angiogenesis for cancer treatments have been extensivelyexplored. Though several therapies, such as bevacizumab(anti-VEGF) and ramucirumab (anti-VEGFR2), havebeen approved to treat several types of tumors, onlymoderate levels of antitumor activity were observed.Along with the booming of bispecific programs and betterunderstanding of the angiogenesis process, new generationof anti-angiogenesis treatments are emerging. As shown inSupplementary Table S1 available online at ABT Online,12 programs are under active development. Majority of theprograms are focusing on improving the anti-angiogenesiseffect by combinatory targeting two or even three moleculesthat are involved in angiogenesis, such as VEGF, VEGFR2,DLL4, and ANGPT2.

Dilpacimab (AbbVie) targeting DLL4 and VEGF isone of the most advanced programs in this category.DLL4-Notch signaling plays a critical role in angiogenicsprouting, and DLL4 blockade alone has shown inhibition

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 45

in tumor growth [77, 78]. Dilpacimab was designed toco-inhibit both DLL4 and VEGF signaling to achievemore prominent antitumor efficacy [79]. Dilpacimab wasgenerated using DVD-Ig platform with the variable domain(VD) of anti-DLL4 located at the outer position and anti-VEGF VD located at the inner position. Interestingly, in thepresence of VEGF, dilpacimab showed 20–50× enhancedcapability of blocking DLL4 signaling, which may be dueto the VEGF homodimerization-mediated cross-linking ofdilpacimab, then enhanced its binding avidity to DLL4on the cell surface, and promoted the downregulationof DLL4 [79]. Considering higher levels of VEGF attumor sites than in peripheral circulation, this uniquecharacteristic of dilpacimab may conditionally enhanceDLL4 neutralization activity only at tumor sites. Currently,the treatment of dilpacimab along or in combinationwith chemotherapy in patients with advanced solid tumorhave shown acceptable safety profile and demonstratedpreliminary antitumor efficacy [80, 81].

Anti-tumorigenesis. Anti-tumorigenesis by targetingoncogenic receptors is another well-validated anticancertreatment. Trastuzumab targeting Her2 was approved totreat Her2-overexpressing breast cancer in 1999. Later,pertuzumab recognizing a different epitope of Her2 wasapproved in 2012 for treatment of patients with Her2-positive metastatic breast cancer in combination withtrastuzumab and chemotherapy. To develop an ideal com-binatorial treatment with trastuzumab and pertuzumab, ahandful of biparatopic Her2 bsAb programs are under earlyclinical testing. ZW25, a biparatopic Her2 bsAb designedbased on Azymetric platform, able to bind domains 2 and 4of extracellular region of HER2 simultaneously to promoteinternalization of HER2 and to inhibit HER2/HER3heterodimer formation, demonstrated promising clinicalefficacy in a Phase I study (ESMO-Asia 2019). Addi-tionally, several other oncogenic targets are also underevaluation, such as EGFR, Her3, cMET, and lipoproteinreceptor-related proteins (LRP) 5/6. Leading players in thisarea are Merus (Her3 × Her2), Jiangsu Alphamab (Her2biparatopic), Zymeworks (Her2 biparatopic), Janssen(EGFR × cMET), and EpimAb (EGFR × cMET),followed by Beijing Mabworks, Boehringer Ingelheim (BI),Molecular Partners, etc. (Table 1).

In collaboration with Genmab, Janssen has developedJNJ-61186372 to concurrently block both EGFR andcMET pathways for treatment of patients who are resistantto EGFR tyrosine kinase inhibitors (TKIs). It has beenshown that JNJ-61186372 not only effectively blocksthe ligand binding-induced EGFR and cMET activationbut also promotes the downregulation of both EGFRand cMET. To further enhance its antitumor potency,the antibody-dependent cellular cytotoxicity (ADCC)function of JNJ-61186372 is augmented by production ina fucosylation defective CHO cell line [82]. In the first-in-human (FIH) Phase I study, JNJ-61186372 has been testedon 142 patients who were progressed after EGFR TKItherapies and has shown promising antitumor activity with∼30% partial response rate.

Though most of the companies are focusing on mod-ulating the signaling of the well-validated ErBB family

members, Boehringer Ingelheim is developing a first-in-class biparatopic antibody to block the function ofLRP5/LRP6 and Wnt/β-catenin pathway. LRP5/LRP6forms trimeric complex with the serpentine receptorFrizzled and Wnt and mediates the stabilization of β-catenin, the transcriptional activator of the Wnt targetinggenes. Aberrant Wnt/β-catenin pathway activation cancontribute to the carcinogenesis and has been observedin many types of tumors. It has been suggested thatLRP5/LRP6 can interact with different Wnt ligands atdifferent domains, and mAbs blocking different domainsshowed different profile [83]. Therefore, BI generatedthe LRP5/LRP6 biparatopic nanobody, BI 905677, withhigh affinity and complete blockage of the binding ofWnt ligands to LRP5/LRP6, thereby inhibiting the Wnt-mediated cancer cell proliferation and survival [84]. Thismolecule is currently at Phase I in patients with differenttypes of solid tumors.

To further expand the antitumor activity, strategiesin combining the blockage of both angiogenic andtumorigenic pathways have been exploited, such astargeting VEGF and cMET (Merus), as well as Her3 andLGALS3BP (MediaPharma). Both programs are still atpreclinical stage (Table 1).

Enhance tumor immunity. Though the idea of immunother-apy dates back to the 1890s, it was not until the 2010s whenit had significant breakthrough with ipilimumab launchedin 2011 and Keytruda and Opdivo in 2014. The aim ofthe immunotherapy is to boost the patients’ own immunesystem to generate antitumor T cell responses. This canbe achieved by either blocking the inhibitory signals, suchas CTLA-4 and PD-1, or enhancing the co-stimulatorysignals, such as 4-1BB and OX40. Till today, anti-CTLA-4and anti-PD(L)1 treatments have shown promising efficacyand revolutionized cancer treatment. Nevertheless, only10–30% of the patients benefit from the treatment [85, 86].

The immune system is a fine-tuned system, withredundancy in most of the regulatory pathways to avoiddamage to the host while it remains effective to clearinfection and tumor cells. To further improve the antitumorefficacy of anti-CTLA-4 and anti-PD(L)1 therapies, severalstrategies are being evaluated, including combining theanti-angiogenesis treatment with anti-PD-1 treatment(VEGF + PD-1) and combining the blockage of multipleimmune checkpoints (CTLA-4 + PD-1 [87], PD-1 + LAG-3, etc.). Though the additive effect can be achieved bycombining two mAbs, bsAbs have the advantages indevelopment as a single molecule entity, sometimes mayeven have synergistic efficacy. As of September 2019,there are 11 bsAbs targeting multiple inhibitory immunemodulatory pathways such as PD-1, CTLA-4, LAG-3,TIM-3, etc. under active development at clinical and 13 atpreclinical stage (Table 2). For example, the co-expressionof PD-1 and LAG-3 on tumor-infiltrating lymphocytesidentifies the tumor-specific T cells [88], which are mostlydysfunctional [89]. The co-treatment of anti-PD-1 andanti-LAG-3 can effectively restore the T cell function [90]and have showed antitumor activity in PD-1-resistantpatients [91]. Based on this fact, several companies areevaluating PD-1 × LAG-3 bsAbs. Some of the molecules

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

46 Antibody Therapeutics, 2020

represent preferential binding on the double-positive Tcells and are more effective in upregulating the T effectorcell function, as compared to the combination of thetwo parental antibodies [WO2017019846; WO2018134279;WO2018185043; WO2019158942].

Despite the success achieved by the immune checkpointinhibitors, the development of co-stimulatory signal ago-nists was hindered by the intriguing balance between safetyand efficacy. For instance, the co-stimulatory molecule4-1BB is a promising target for cancer immunotherapy,as it can activate T cells, NK cells, and other immune cellsand has been clinically validated in CAR-T therapies tosustain T cell activation. However, the clinical developmentof anti-4-1BB monoclonal antibodies was stagnated byeither liver toxicity or lack of efficacy [92]. To minimizethe safety issue associated with the systemic activationof 4-1BB, strategies have been employed to localize theactivation of 4-1BB at tumor site. Roche developed a4-1BBL fusion protein targeting fibroblast activationprotein (FAP) with Fc region mutations to abrogate Fcγ Rbinding but maintain favorable PK profile. The in vitrofunctional tests suggested that, only in the co-presenceof anti-CD3 signal and FAP-expressing cells, FAP-4-1BBL can increase T cell activation and proliferation.Furthermore, preclinical studies showed that FAP-4-1BBLcannot inhibit tumor growth by itself. The combined treat-ment with relevant T cell-redirecting bispecific antibodies(TRBAs) or immune checkpoint inhibitor, such as anti-PD-L1, can efficiently inhibit tumor growth without prominentliver toxicity [93]. Recently, at the 34th SITC annualmeeting, Pieris Pharmaceuticals reported the preliminaryresults of the Phase I study of PRS-343 (Her2 × 4-1BB) inpatients with Her2+ malignancies. Among the 18 patientswho received active doses of PRS-343, 2 patients reachedpartial response, and 8 patients had stable diseases. This isthe first 4-1BB agonistic treatment reported with promisingefficacy as well as good safety profile.

The tumor site localization strategy has also beenexplored to selectively activate other co-stimulatoryreceptors, such as OX40, CD27, CD28, CD40, and ICOS,as well as to selectively inhibit co-inhibitory receptors,including CTLA-4 and PD-1. There are 10 bsAbs utilizingthis strategy under active development at clinical and 19at preclinical stages, which reflects the growing interests inthis area (Table 2).

Modulate TME. To evade the immune surveillance,tumor cells can commonly influence the microenviron-ment around them by expressing immunosuppressivemolecules, such as TGFβ and CD73, and by recruitingor promoting the differentiation of immunosuppressivecells, such as myeloid-derived suppressor cells (MDSCs),tumor-associated macrophages (TAMs), and regulatory Tcells (Tregs). A few bsAbs and bifunctional proteins aredeveloped to overcome immunosuppressive TME, suchas bintrafusp alfa, an anti-PD-L1, and TGFβRII fusionprotein. TGFβ is a pleiotropic cytokine and plays dualfunctions in cancer progression. Though TGFβ suppressestumor progression at tumor initiation stage, at later stages,TGFβ facilitates tumor progression and metastasis andhas been suggested to contribute to resistance to anti-PD-1

and chemotherapy. Bintrafusp alfa (aka M7824) has theTGFβRII extracellular domain fused to the C-terminalof avelumab [94]. In preclinical studies, M7824 exhibitedstrong antitumor activity and significantly decreasedthe incidence of metastasis in mouse tumor models. Inclinical tests, M7824 displayed acceptable safety profileand encouraging clinical efficacy in patients with heavilypretreated advanced solid tumors [95]. Other strategies tar-geting TME, including CD73 × TGFβ, EGFR × TGFβ,and CCR2 × CSF1R, are also under development at earlyclinical stage or preclinical stage (Table 3).

Target cell depletion. The last group represents themajority of the bsAb programs (clinical, 60/99; preclinical,99/153) to promote the target cell depletion by differentmechanisms (Table 4). According to the MOAs, this groupcan be further divided into subgroups, including cytotoxiceffector engagement, Fc effector function (ADCC, ADCP,complement-dependent cytotoxicity [CDC]), enhancedphagocytosis, enhanced apoptosis, and drug conjugation.

Cytotoxic effector engagement is the largest subgroupin this category. Two out of the three launched bsAbs,catumaxomab, and blinatumomab are in this subgroup.Catumaxomab contains the antigen-binding sites for CD3on the T cells and EpCAM on the cancer cells [96]. It wasfirst authorized for market by the EMA in 2009 for thetreatment of malignant ascites [97], but was withdrawn in2017 due to commercial reasons. On the other hand, blina-tumomab targeting CD3 and CD19 has shown impressiveclinical results since launched in 2014 [98, 99].

T cells identify the target cells by recognizing the peptidespresented by the major histocompatibility complex (MHC)through TCR. Based on the dynamic segregation model,the interaction of TCR with cognate peptide/MHC com-plex (pMHCs) brings the T cells and target cells in closeproximity (∼14 nm) and results in TCR clustering at thecenter of immune synapse (IS) and exclusion of the largeinhibitory tyrosine phosphatases from this region [100].Following TCR clustering, cytolytic granules move towardthe center supramolecular activation cluster (cSMAC) andrelease perforin and granzymes into the target cells. Oncethe target cells undergo cell death, the T cells quicklydetach from the dying cells and move to the next targetcell [100].

TRBAs are a group of bsAbs that can simultaneouslytarget a component of the TCR complex (most commonlyCD3ε) on a T cell and a target on the tumor cell surface[101–103]. By this approach, TRBAs promote IS formationbetween T cells and cancer cells independent of ligationof TCR with pMHCs, leading to T cell activation andkilling of the tumor cells [104–107]. Due to the clinicalsuccess of blinatumomab, the development of TRBAshas gained substantial attention with 51 programs are atclinical and 66 at preclinical stages. Molecules in differentformats are under evaluation to prove whether they candeliver the proposed biological function or have therapeuticwindow. For examples, BiTE and half-life extended BiTEare used by Amgen; DART and DART-Fc format areevaluated by MacroGenics; common light chain formatis under investigation by Regeneron; and DuoBody format

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 47

is under development for multiple projects by Genmaband Janssen. Glenmark’s BEAT platform, Xencor’s XmAbplatform, Aptevo’s ADAPTIR platform, and Teneo-Bio’s unique anti-CD3 platform are also under activeexploration. Recently, IGM Biosciences announced theinitiation of FIH Phase I clinical trial of IGM-2323, anIgM-based CD20 × CD3 TRBA. Unlike other formats,containing only 1 or 2 binding units for the tumor-associated antigen (TAA), the IGM-2323 contains 10binding units for CD20. It is hypothesized that the higheravidity to CD20 of IGM-2323 may provide an advantage totreat CD20low tumor cells over other formats. Moreover, theIgM-based TRBAs can more efficiently mediate CDC thanIgG antibody. However, whether the IgM-based TRBAscan deliver superior efficacy to other formats still needs tobe confirmed in the clinical studies.

Approaches to engage effector cell populations otherthan conventional αβ T cells, such as CD8 T cells, γ δ Tcells, NK cells, and iNKT cells, have also been exploredand were reviewed by Ellerman recently [108]. The γ δ Tcells represent 10% of the total T lymphocyte populationin circulating blood. Unlike conventional T cells, γ δ Tcells recognize stressed and malignant cells independent ofMHC molecules, and their activation does not require co-stimulatory signals [109]. Besides strong cytotoxic activity,one unique property of activated γ δ T cells is that theycan cross-present tumor antigens to enhance CD8 T cellresponse [110]. A few strategies to improve the antitumoractivity of the γ δ T cells have been explored at preclini-cal and early clinical stages [111]. A selective Vγ 9Vδ2 Tcell engager (Her2 × Vγ 9) showed superior activity ininducing Vγ 9Vδ2 T cell-mediated tumor cell lysis thanHer2 × CD3 TRBAs in vitro and exhibited antitumor activ-ity in combination with IL-2 and activated γ δ T cells adop-tive transfer treatments in the PancTu-1 xenograft mousemodel [112].

It has been argued that NK cell engagement may havebetter safety profile over T cell engagement therapies,while providing similar levels of clinical efficacy. CD16is the most commonly used target for engaging NKcells. Results published by Affimed have suggested thatNK engagers may induce efficient target cell killing withlower cytokine release risk, when compared to CD3 Tcell engagers [113]. Early clinical results reported at 60thASH meeting and 15th ICML meeting had shown thatAFM-13 (CD16 × CD30) was well tolerated and effica-cious when administrated alone or in combination withpembrolizumab [114, 115]. The definitive clinical benefitof NK cell engagement still remains to be demonstratedin ongoing clinical studies. Other NK cell-activatingreceptors that are considered to have distinct advantagesto overcome certain deficiencies in TME, such as NKG2D,NKp30, and NKp46, are under preclinical evaluation [116].Additionally, strategies that redirecting iNKT cells by usingCD1d extracellular domain fusion protein is also at earlyresearch stage [117].

Along with the growing depth of knowledge in Fc effec-tor function, several approaches have been adopted forbsAbs, including mutations in the Fc region to enhancethe Fcγ R binding, and afucosylation. Genmab has estab-lished a HexaBody platform, which contains mutations

(E430G) in the IgG1 Fc region to enhance hexameriationupon antigen engagement and thereby enhance the CDCeffect. By combining the HexaBody and DuoBody plat-forms, Genmab has developed a DuoHexaBody anti-CD37biparatopic antibody. In ex vivo CDC assays using samplesisolated from lymphoma patients, the DuoHexaBody anti-CD37 biparatopic antibody showed more potent tumor celllysis, as compared to the control anti-CD20 antibodies,rituximab, ofatumumab, and obinutuzumab [118].

CD47-SIRPα signaling plays an inhibitory effect onthe phagocytosis by phagocytes, such as macrophages.Tumor cells overexpress CD47 on the cell surface toescape the elimination by phagocytes. Antibodies againstCD47 and SIRPα have been developed to interruptCD47-SIRPα signaling. The combined treatment ofa CD47 antagonist, Hu5F9-G4, with rituximab showedpromising therapeutic efficacy in patients with non-Hodgkin lymphoma (NHL) [119]. By taking the advantageof the CD47 antagonist, a series of bsAbs using anti-CD47as one moiety to enhance the phagocytosis to the cancercells have been developed. Though there is only 1 programat clinical Phase I (NI-1701, CD19 × CD47), 14 programsare undergoing active development at preclinical phase withCD47 coupled with different tumor-associated antigens(Table 4), suggesting there is substantial growing interestsin this area. Recently, the results published by Hattereret al. have suggested that in addition to the enhancementof phagocytosis, the co-engagement of CD47 and CD19by NI-1701 can prevent the colocalization of CD19 toBCR cluster during B cell activation, therefore inhibitingactivated B cell proliferation [120].

Unlike previously mentioned bispecific programs, whichall rely on the cytotoxic function of the effector cells or thecomplement system, two preclinical programs are focusingon actuating the apoptotic process of the cancer cells byactivating the apoptotic receptors. BI and Promethera gen-erated bsAbs targeting CDH17 × TRAILR2 (BI-905711)and CD20 × CD95 (Novotarg), respectively. According tothe report published by BI on 2019 AACR Annual Meeting,BI-905711 induced TRAILR2 clustering on a CDH17-dependent manner and selectively triggered the apoptosisof CDH17 expressing tumor cells. BI-905711 also demon-strated significant antitumor activity in multiple colorectalcancer xenograft models [121].

Lastly, there are a couple of bsAbs that are being usedto deliver toxin into cells that are positive for either or bothtargets based on the particular design of each molecule. Forexample, Regeneron is working on APLP2 × Her2 bispe-cific antibody-drug conjugate (ADC). Amyloid precursor-like protein 2 (APLP2) has been suggested to be involvedin increased tumor cell proliferation and migration, andaberrant APLP2 expression was observed in multiple typesof cancers, such as breast cancer [122]. Though APLP2 is aninternalizing receptor, due to its ubiquitous expression andthe presence of secreted form, APLP2 is not an ideal targetfor ADC. Trastuzumab emtansine (T-DM1) has shownpotent efficacy in cancer cells with high level of Her2expression, but has little effect on cells with mid to low lev-els of Her2 expression. To improve the therapeutic efficacyof Her2 ADC, Regeneron developed the Her2 × APLP2-DM1. The bsAb binds to Her2-positive cells with the high-

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

48 Antibody Therapeutics, 2020

affinity Her2 binding arm and then bridges to the cellsurface APLP2 with the low-affinity APLP2 binding arm,which promotes rapid antibody internalization, lysosomaltrafficking, and tumor cell killing [123].

Bispecific antibodies for inflammatory conditions

Autoimmune disease is the second largest area for bsAbs’applications, with 10 clinical programs and 12 preclinicalprograms ongoing. Most of these programs are aiming toblock the function of multiple pro-inflammatory cytokinesby combining the neutralizing antibodies into one moleculeentity, such as IL-1α × IL-1β, IL-17 × IL-13, IL-4 × IL-13,and BAFF × IL-17 (Table 5).

In immune cells, when it is in-cis coupled with an activat-ing receptor, the inhibitory receptor can play a dominantnegative role by diminishing the transduction of the activesignal [124]. It has been shown that MGD-010 targetingCD79B, one component of the BCR complex and theinhibitory receptor CD32B, can decrease B cell responsewithout depleting the B cells in healthy donors [125]. Onois developing ONO-4685 (CD3 × PD-1) to turn down theT cell responses in autoimmune diseases. However, it is stillnot clear whether the effect of ONO-4685 is dependent onthe in-cis engagement of CD3 and PD-1 to block the Tcell activation or by in-trans interaction to deplete PD-1expressing activated T cells.

Bispecific antibodies for other conditions

Hemophilia A. Hemophilia A is another successfulexample in the application of bsAbs, with the approvalof emicizumab in 2017 as a landmark event. Emicizumabbridges factor IXa and X in spatially appropriate positionsto facilitate the factor IXa-catalyzed factor X activation,which is usually mediated by factor VIII in healthyindividuals, but is deficient in patients with hemophiliaA. Though the etiology of hemophilia A has been wellunderstood for a long period of time, the treatment optionsare still limited. Recombinant factor VIII and humanplasma-derived factor concentrates are the commonly usedpractices for hemophilia A. However, the short half-lifeof factor VIII and development of anti-drug antibody(ADA) remains the major challenges for factor replacementtherapy [126]. Emicizumab was intentionally designed tofunction as factor VIII with prolonged plasma half-life[127, 128]. Clinical results in hemophilia A patients withfactor VIII inhibitor showed that the weekly subcutaneous(SC) treatment of emicizumab significantly reduced thefrequency of bleeding episodes with no detectable anti-drug antibody [129]. Based on its promising efficacyand superior regimen schedule, emicizumab was initiallylaunched in the USA for hemophilia A patients with factorVIII inhibitor in 2017; and then its usage was quicklyexpanded to patient without factor VIII inhibitor and waslaunched in EU and Japan in 2018. Similar programs areunder preclinical development by Kymab and Shire.

Ocular. Excessive neovascularization, bleed, and fluidleakage from the abnormal blood vessels result in rapidvision loss or even blindness in patients with wet form

age-related macular degeneration (AMD). Anti-angiogenesistreatment, such as Lucentis, Eylea, and Beovu, has beenapproved for treatment of this condition and has shownsignificant improvement in visual acuity and prevention ofvision loss. Though over 90% of the patients can benefitfrom the treatment (i.e., maintain vision), eventually thesepatients become resistant to the treatment. New therapiesare needed to further improve the therapeutic efficacy. Aswe mentioned above, several bsAbs have been developedto block the process of angiogenesis for cancer indications.Similar strategy has also been exploited for the treatmentof wet AMD and diabetic macular edema (DME). Roche’sfaricimab is an Ig-like bsAb targeting VEGF and ANGPT2using CrossMab technology. During clinical tests, faricimabhas shown superior efficacy and safety in patients withDME, as compared to Lucentis [130]. Phase III studiesto evaluate faricimab’s therapeutic efficacy are initiated inearly 2019; and the filing for BLA is expected in 2021.

Neurology. Despite the tremendous efforts in developingbiological therapeutics for neurodegeneration diseases,effective treatment remains elusive. One of the obstaclesin developing biological drugs for neurological disease is toeffectively deliver the large molecule into the central neuronsystem. “Trojan horse” bsAb has one binding specificityresponsible for the transportation of the antibody tothe location that otherwise cannot be reached naturally,whereas the other binding specificity fulfills its function. Byusing this approach, a group of bsAbs have been developedto cross the blood-brain barrier (BBB). These antibodiesusually have one binding arm recognizing the receptorsin the receptor-mediated transcytosis system, such asinsulin receptor, transferrin receptor, and lipoproteintransport receptors [131], and the other arm targetingthe pathogenic molecules (Table 6). Bifunctional fusionproteins or antibody-drug conjugates are also under activedevelopment as therapeutic drugs or diagnostic reagentsfor central nervous system diseases, but they are not underthe scope of this review.

Infectious diseases. Due to the high frequency of escapemutations and development of drug resistance to single-agent treatment, combinatory treatment with mixture ofmAbs or by bsAbs to broaden the protection spectrum andto decrease the chance to establish drug resistance is beingdeveloped to fight against infections. MEDI3902 was orig-inally designed to achieve broader protection against Pseu-domonas aeruginosa by combining two clinically provenanti-PcrV and anti-Psl antibodies into one molecule. Psland PcrV are present in ∼90% of the P. aeruginosa clinicalisolates, respectively. Theoretically, the bsAb targeting Psland PcrV simultaneously can protect the host from theinfection of 97–100% of the isolates, which express eitheror both targets. Surprisingly, when compared to the mixtureof the parental antibodies in preclinical studies, MEDI3902showed enhanced efficacy. By further dissecting the MOA,it was found that the format of MEDI3902 rendered a high-avidity low-affinity binding to Psl, which led to the accu-mulation of MEDI3902 around the bacterium and moreefficient blocking of PcrV-mediated cytotoxicity [132].

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 49

The “Trojan horse” strategy is also employed by somebsAbs with elegant design for infectious diseases treatment.During filoviruses (e.g., Ebola virus) infection, the mem-brane envelope glycoprotein (GP) is responsible for the cellattachment and membrane fusion. A unique feature aboutthe GP of filoviruses is that it first binds to the receptoron the cell surface which induces the internalization ofthe virus particle, and then in the late endosome, GP iscleaved to expose the highly conserved receptor-bindingsite (RBS) for Niemann-Pick C1 (NPC1), which mediatesthe membrane fusion and cell entry [133]. Therefore, toprovide broad protection against filoviruses, bsAbs weredesigned to block the intracellular GPCL-NPC1 interac-tion by coupling the GPCL-NPC1 blocking arm with adelivering arm targeting a broadly conserved epitope inuncleaved GP. The delivering arm binds to the virus par-ticles and goes into the endosome together with the virus,where the blocking arm functions to abrogate the GPCL-NPC1 interaction when it is exposed and prevents theviral entry [134].

Diabetes. Fibroblast growth factor 21 (FGF21) playskey roles in stimulating metabolism and has shown somepreliminary clinical benefits in obese patients with diabetes.However, the poor PK profile and potential adverse effectsassociated with long-term usage of recombinant FGF21limit its usage. RG7992 (FGFR1 × KLB) was thereforedesigned to mimic the function of FGF21 but selectivelyactivate FGFR1/KLB complex in the liver, adipose, andpancreas tissues, where KLB is present, to avoid safetyconcern associated with broad FGFRs activation, but stillbe able to provide clinical benefit in obesity and diabetes[135].

MATCH BIOLOGY WITH AN OPTIMAL BISPECIFICFORMAT

As discussed in the early section, format diversity is essen-tial to serve the plethora of applications of bsAbs defined byTPPs. Variances in affinity, valency, epitope, and geometryof their binding domains, linkers, as well as in size- and Fc-mediated distribution and pharmacokinetic properties tofulfill a particular clinical application define a bsAb format.In practice, many variances or attributes for selecting anoptimal format are intertwined and must be addressed forselecting the right molecule. Therefore, we will discuss theseattributes below.

Antigen-binding affinity and valency

Affinity. Even though one of the advantages of usingantibody-based therapeutics is that they may interactwith their antigens with substantially high affinities,higher affinity does not always translate into betterefficacy. Unlike antagonistic molecule, whose potency isusually associated with its affinity, agonistic molecule ismore difficult to predict and to optimize its potency byincreasing the binding affinity. Based on different modesthe receptor uses for activation, different binding kineticsof the agonistic bsAb to reach optimal receptor activation

are required. For receptors depending on clustering toactivate, fast-on fast-off binding kinetics is preferred toensure efficient recruitment of receptors [136, 137]. Onthe contrary, for receptors activated by ligand binding-induced conformational change, the slow off bindingkinetics would endorse more durable activating efficacy[138]. Furthermore, there are evidences that the affinityto CD3 may significantly affect the function and safetyprofile for TRBAs. It has been suggested that T cells requirelower threshold for mediating cytotoxic killing than forcytokine production perhaps due to different number ofITAM motifs of TCR complex being phosphorylated,it may be possible to dissociate TRBAs’ potency fromtoxicity by modulating the CD3 affinity of the bsAbs.As shown by Leong et al., by lowering the affinity toCD3, the CD3 × CLL1 bsAb with low affinity to CD3exhibited better safety profile and retained equivalent invivo efficacy, as compared to the ones with high affinity toCD3 [139] when net impact on T cell activation, receptorinternalization, and PK all combined. Similar results werealso shown by Zuch de Zafra et al. By comparing a series ofCD38 × CD3 bsAbs with different affinities to CD3, theyfound that lowering the affinity to CD3 can dramaticallydecrease the cytokine release, but still maintain potencyin mediating cytotoxic killing [140]. In November 2019,AMG-424, the final lead from the aforementioned study,was granted with orphan drug designation for multiplemyeloma by the FDA.

As for the affinities of TRBAs to TAAs, due to the differ-ent expression profile of the TAAs in normal tissues versusin tumors, and the tolerability and the ability of regener-ation of TAA-positive cell populations in normal tissues,the TRBAs targeting different TAAs may require differentbinding kinetics. For low-expression, tumor-specific anti-gens, a TRBA with high affinity to the antigen wouldbe required to elicit efficient tumor cell killing. However,for TAA with low expression on essential normal tissues/organs, to spare the normal cells and avoid on-target off-tumor toxicity, low-affinity high-avidity TRBAs would bepreferred, which can be achieved by modulating the valency(see below).

Moreover, for a bsAb, difference in affinities of twodifferent antigen-binding specificities may determine whicharm drives tissue distribution, tissue penetration, and reten-tion of a therapeutic molecule at the site of MOA. Forexamples, high affinity to TAA and low affinity to CD3may enable the preferential binding of TRBAs to the targetcells and implement serial killing of the target cells by asingle T cell [141]; and as mentioned above, APLP2 × Her2bispecific ADC with high affinity to Her2 and low affinityto APLP2 preferentially binds to Her2-positive cells andthen bridges APLP2 on the cell surface to mediate efficientendocytosis to avoid the toxicity associated with the panexpression of APLP2.

For BBB crossing bsAbs, along with other consider-ations, careful selection of the transport receptor andselection of a molecule with appropriate binding kinetics tothe transport receptor is critical for success of this strategy.As reported by the scientists from Genentech, to ensurethe effectiveness of the transcytosis, the “Trojan horse”antibody using the TfR pathway needs to have low affinity

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

50 Antibody Therapeutics, 2020

to TfR [142]. Later, another study by the University ofWisconsin-Madison showed that TfR bsAb with highbinding affinity to TfR at pH 7.4 but low affinity atpH 5.5 can effectually release the bsAbs from BBB into thebrain and avoid the degradation of bsAb in the endosome[143].

Valency. The valency for each target can dramaticallyaffect the function of the bsAbs. For the TRBAs, mono-valency of anti-CD3 arms may help to avoid non-specificactivation of the T cells without engagement of tumor cells,as shown by Bardwell et al. [144]. Interestingly, Y-mAband Abpro have CD3 scFv fused to the C-terminus ofthe light chain. Even though the format ends up with twobinding sites for CD3, both companies claimed that thisformat was actually functional monovalent toward CD3.Additionally, Aptevo and Affimed also developed TRBAsusing bivalency to CD3. Preclinical evidence has suggestedthat the adoption of the ADAPTIR format can inducepotent T cell activation and target cell killing, but low levelsof cytokine release [145]. AFM-11 (CD19 × CD3) alsoshowed more potent T cell activation than BiTE controland strict CD19-dependent T cell activation preclinically[146]. However, due to one death and two life-threateningevents in clinical trial, AFM-11 was placed on clinicalhold.

The valence for the TAA may vary based on the prop-erties of the TAA, such as tumor specificity, antigen size,expression level on the tumor versus normal tissue, and thetolerance of complete elimination of TAA-positive cells. Inthe case of some types of hematopoietic tumors, the deple-tion of both normal and malignant cells expressing TAAs,such as CD19 and/or CD20, can be tolerated. However, formost of the other TAAs, the expression levels may be lowon normal tissues, but the killing of these low-expressionnormal cells can lead to deleterious consequences. To dis-tinguish the targethigh tumor cells from the targetlow normaltissue, RG7802 (CEA × CD3) was optimized to have low-affinity high-avidity 2 + 1 format in appropriate geometryto facilitate the selection of CEAhigh cells with a threshold of∼10 000 CEA-binding sites/cell [105].

Based on the lessons learned from the initial mAb devel-opment for cMET treatment, bivalency to cMET mayelicit agonistic, instead of antagonistic, effect resulting fromthe mAb-mediated dimerization of cMET. Though mono-valent binding to cMET can function as an antagonist,it can only block the HGF-mediated cMET activation.Later, Wang et al. demonstrated that ABT-700, a trulyantagonistic mAb against cMET, can bind to a uniqueepitope on cMET. The bivalency to cMET of ABT-700 andstringent hinge region was essential to inhibit both HGF-dependent and HGF-independent activation of cMET andinduce cMET downregulation [147]. Interestingly, half ofthe cMET bsAb programs are using monovalency againstcMET to avoid agonistic effect, while the other half choosebivalency. EMB-01 (EGFR × cMET) has two binding sitesfor cMET, and has no obvious cMET activation in theabsence of ligands. Furthermore, it can effectively induceEGFR and cMET degradation, therefore preventing thecMET activation [62].

Epitope, geometry, and distance between differentantigen-binding domains

Epitope. In respect of antagonistic bsAb, the bindingepitope of the corresponding binding units are requiredto prevent the receptor/ligand engagement, or the receptorsignal complex formation, or any step that is crucial for theinitiation or passage of signaling cascade into the cells toplay its biological function.

In general, the receptor-binding epitope for agonisticmolecules is not as predictive as it is for antagonisticmolecules. However, there is evidence to suggest thatthe binding epitopes do contribute significantly to thebsAb efficacy. It was found that anti-CD3 binding armsrecognizing different epitopes on CD3-activated T celldifferently. TeneoBio, therefore, identified a dozen ofanti-CD3 antibodies with different binding epitopes anddifferent binding kinetics to CD3 molecules to disassociatethe capabilities of TRBAs in inducing cytotoxic killingfrom promoting cytokine production post T cell activation.They identified a clone (F2B) that recognizes a uniqueepitope on CD3δε, but not CD3 γ ε, at a low affinity(34 nM). By comparing to another clone (F1F), whichbinds to both CD3δε and CD3 γ ε with high affinity(<1 pM), they found that BCMA × CD3 bsAb usingF2B arm (CD3_F2BxBCMA) can induce moderate levelsof cell killing but very weak cytokine production, ascompared to the one using F1F arm (CD3_F1FxBCMA)in vitro. Moreover, the in vivo efficacy study showedthat CD3_F2BxBCMA exhibited antitumor activity in awide dose range (0.01–10 μg), while CD3_F1FxBCMAcompletely lost its therapeutic efficacy at the high dose(10 μg) [148].

As we mentioned above, to effectively redirect T cellkilling, the TRBAs must be able to induce the IS formationbetween the T cells and target tumor cells. Besides theformat of the TRBAs, the tumor antigen selection, the sizeof the antigen, antigen surface density, as well as the dis-tance between the TRBAs binding epitope to target cellmembrane, all can influence the formation of the IS. Com-paring to large antigens or antigens with protruding struc-ture, the small antigens or antigens with structure closeto the cell membrane can more effectively promote the ISformation [106]. When the selected tumor antigen is largein size, such as melanoma chondroitin sulfate proteoglycan(MCSP) [149] and FcRH5 [150], the membrane-proximalepitope is desired. For cell surface targets that can be shedinto the bloodstream, to avoid antigen sink, the bsAbsshould recognize the membrane-bound but not the solubleform of the antigen [151].

Geometry. Besides the distance between the epitope tothe target cell membrane, the distance between the twotargets engaged by TRBAs also plays a crucial role in deter-mining whether it can effectively promote IS formation andT cell activation. Considering the distances between theTAA and CD3 epitopes to target cell and T cell, respec-tively, the format of the TRBAs needs to bring TAA andCD3 to a close proximity much less than 14 nm. Moreover,the whole molecule has to be able to physically fit into thesmall junction between the two cells in a density to effec-

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 51

tively form a cluster with several engaged target pairs toinitiate TCR signaling. Despite its short serum half-life, thesmall size of BiTE format with two binding units locatingin opposite sides is extremely potent in redirecting T cellcytotoxicity by inducing serial killing of tumor cells at aneffector-to-target ratio as low as 1:5 [141]. In another case,Aptevo fused the scFvs against the TAA and CD3 at the N-and C-terminus of Fc (scFv + scFv with Fc, 2 + 2), whichended up with longer distances between the two bindingdomains. The in vitro studies showed that this molecule hadmore potent target cell killing, but less cytokine release,as compared to the BiTE format [145]. Unfortunately, theclinical development for this molecule was discontinueddue to high frequency of anti-drug antibody development.

The same situation also applies to T cell co-stimulatoryand co-inhibitory receptors, which co-cluster with TCRduring IS formation and regulate T cell activation. PD-1and PD-L1 interaction leads to the accumulation of PD-1microclusters at cSMAC and destabilizes the IS. When theextracellular domain of PD-1 was elongated by insertingextra Ig domains, the inhibitory role of PD-1 decreasedalong with the increase of the number of Ig domainsinserted [152]. Though current anti-PD-1 molecules allblock the PD-1 signaling by inhibiting the PD-1/PD-L1interaction, in theory, the designs that can prevent the PD-1 colocalization to cSMAC should also be able to diminishthe inhibitory role of PD-1 in T cell responses. On thecontrary, bsAbs to activate the co-stimulatory receptorsuch as 4-1BB must exert its function at the site of IS[93]; therefore, a format that can meet these criteria isnecessary. As reported by Pieris, the geometry of the 4-1BBanticalin attachment significantly affected the function ofthe Her2 × 4-1BB bispecific anticalins. PRS-343 with 4-1BB anticalin attached at the C-terminus of the heavy chainshowed the most effective T cell activation, as compared toother formats. One possible explanation is that the bindingsites for Her2 and 4-1BB are approximately 15 nm apart,which is close to the distance of the IS. However, aftermeasuring the distances from the binding epitopes to thecell membrane, the distance between the target cell and theeffector cell might be much longer than 15 nm. On the otherhand, ND-021 (PD-L1 × 4-1BB × HSA) is an Fc-lackingscFv-VHH-based molecule. With its small size and flexiblestructure, it may have better potential in colocalizing atcSMAC and enhance TCR signaling. It will be interestingto see how it will perform in the clinical trials.

Cases are also shown in bsAbs programs developed forother conditions. For example, when the scientists at Med-Immune tested their Psl × PcrV bsAbs, they examined sev-eral different formats with varying intramolecular distancesbetween the two binding components. After comparisonof these formats in both in vitro and in vivo efficacy stud-ies, BiS4aPa, with an intermediate distance, exhibited themost effective protection against P. aeruginosa infectionand therefore was selected as the final therapeutic candidateformat [132].

Linker design

As reviewed by Brinkmann and Kontermann, various con-necting linkers have been explored [3]. Similar to the hinge

region of the IgG subclass, the length, flexibility, and aminoacid composition of the linkers used to connect the buildingblocks (scFv, Fab, etc.) may determine the correct for-mation, functionality, and developability of the resultingbispecific molecules, as shown by Le Gall et al. [153] andDiGiammarino et al. [154].

Size

The bsAbs have made significant impact on hematologicmalignancy treatments. However, the therapeutic benefitsdelivered by bsAb for solid tumor are still waiting to beunveiled. One of the concerns using bsAbs for solid tumortreatment is how to increase the drug tumor penetrationand accumulation. Though molecules with smaller sizewould have better chance entering the tumor site byincreased tumor penetration, the molecules with sizesmaller than the threshold of renal clearance of proteins arerapidly cleared from the blood and therefore have decreasedflux into the tumor [155]. Using a compartmental model,Schmidt and Wittrup predicted that molecules with thesize of 150 kDa would have the best tumor localization,whereas molecules with the size of 25 kDa would havethe worst tumor uptake [156]. However, due to their largesize, molecules at the size of ∼150 kDa have decreasedextravasation and normally take days to reach maximumtumor uptake. On the other hand, molecules of smaller sizereach the maximum tumor uptake within a short period oftime. The fast tumor penetration and systemic clearanceof small-sized molecule therefore lead to high tumor/bloodlocalization ratio, which is preferred for some applications,such as imaging [157], as well as safety management todecrease the systemic drug exposure-associated toxicity.

To improve the serum half-life, while still retaining thefast extravasation property, Harpoon developed the Tri-TAC platform, which targets TAA, CD3, as well as humanalbumin for extended half-life with a total size of ∼50 kDa.It is believed that with its improved drug exposure andsmall size, TriTAC would enable faster and better tumorpenetration, compared to large-sized bsAbs.

Fc region

The Fc region can substantially influence the bsAbs’ func-tion. It was found that the properties of IgG subclass hingeregion, such as length, sequences, flexibility, and disulfidebond structures, can influence the variable region presen-tation and thereby affect the functionality of an antibody[158]. While it is not always desired, the format with Fc canprolong the bsAb serum half-life through FcRn-mediatedrecycling and may provide Fc effector function through theinteraction with Fcγ Rs.

IgG subclass. Recently, Kapelski et al. reported theinfluence of the IgG subclass on TRBAs. They found thatdue to its short and rigid hinge region, IgG2 cannot effec-tively promote the IS formation. However, by replacing thehinge region of the IgG2 with the hinge region of IgG4 orIgG1, the function of IgG2 chimeric bsAb can effectivelyinduce IS formation and redirect T cell killing [159].

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

52 Antibody Therapeutics, 2020

Similarly, the Fc region also showed significant influenceon the factor VIII-mimetic activity of emicizumab. Aftercomparison between different IgG subclasses, interchaindisulfide bonds, and mutations in hinge region and CH2domain, IgG4 was selected as it presented with the mostpotent factor VIII-mimetic activity [160].

Fc effector function. As mentioned above, severalstrategies have been developed to enhance the bindingbetween Fc and Fcγ Rs to increase the Fc effector function.This could be important for bsAbs against TAAs foreffective killing tumor cells or for bsAbs against infectiousagents for pathogen uptake and clearance. However, the Fceffector function and Fcγ R binding are usually abrogatedfor TRBAs and some other agonistic bsAbs to avoidthe Fcγ R-mediated cross-linking, which may cause non-specific activation of T cells and the targeted receptors,respectively. Advances in Fc engineering allow tailoredmodification of Fc effector functions for specific need. Forexample, Xencor developed a series of TRBAs using theXmAb platforms, including AMG-424 (CD38 × CD3,Fab + scFv with Fc, 1 + 1), and used a combinationof mutations (E233P/L234V/L235A/G236del/S267K) tocompletely eliminate the binding of IgG1 Fc to Fcγ Rs [55].

Because IgG4 only binds to Fcγ R1 with high affin-ity and mediates weaker effector function than IgG1,it is commonly used for antagonistic antibodies tar-geting immune cells, to avoid Fc effector function-mediated cell elimination. However, the research by Zhanget al. showed that the anti-PD-1/IgG4 antibody caninduce the phagocytosis of PD-1+ T cells by activatingFcγ RI+ macrophages. By introducing five additionalmutations (E233P/F234VL235A/D265A/R409K), BGB-A317 showed no binding to Fcγ R1, and more efficientpreclinical antitumor activity, as compared to the anti-PD-1/IgG4 control [161]. The recently reported results ofthe pivotal study of BGB-A317 also exhibited its superiorantitumor efficacy in patients with relapsed/refractoryclassical Hodgkin lymphoma, with an overall response rate(ORR) of 87% and 63% complete response rate (CRR).

As we discussed above, the format contains many com-ponents that can be tweaked, their final impact on pharma-cological properties of a bsAb is intertwined, and here weonly mentioned some of them. The fine-tuned parts work inconcert with each other to determine the success of bsAbs.To obtain the optimal therapeutic candidate, the selectionof any component in the final format should be carefullyevaluated for specific target pairs; and the matched formatwill not only facilitate the bsAbs to elicit biological functionbut also may enable a molecule for further product devel-opment, which otherwise may not be suitable for clinicalapplication.

SELECT A RIGHT MOLECULE TO MEET BOTHFUNCTION AND DEVELOPABILITY REQUIREMENT

As illustrated in the early section, the six criteria criticalfor clinical development and commercial manufacturing(Fig. 1) define a good bsAb. Identification of a goodtherapeutic bispecific molecule usually requires starting

with good therapeutic and molecular design definedby MPP that is developed based on TPP, followed byvigorous in vitro and in vivo screening and characterizations.Below we will discuss these six criteria and how they canhave significant impact on the outcome of the resultingbsAbs: physiochemical properties, manufacturability,immunogenicity, PK/PD property, and, most importantly,efficacy and safety.

Physiochemical properties and manufacturability. Asaforementioned, many strategies have been exploredto solve the CMC quality issues, such as mispairing,stability, aggregation, segmentation, solubility, viscosity,purification, etc. A good bispecific clinical candidateshould (1) be easily expressed with high percentage ofcorrectly assembled product in manufacturing scale; (2)display no significant aggregation or low percentage ofaggregation that can be easily removed, as aggregation mayaffect the therapeutic efficacy and increase immunogenicityrisk; (3) have good solubility, high stability, and lowviscosity to meet drug product formulation needs forintended clinical dosage and route of administration; and(4) have low manufacturing cost for economical reason.The stringency of those requirements may differ based onvarious clinical applications. For instance, reconstitutedlyophilized formulation for intravenous infusion (IV) isgenerally acceptable for oncology applications, while liquidformulation developed for SC administration may bepreferred for most of autoimmune indications. For oculardisease, the high solubility, high stability, and low viscosityare imperative for a competitive product. With the advanceof the bsAb technology, more and more reported bsAbformats can be expressed and purified with reasonableyield and meet the reasonable physiochemical propertiesfor a given clinical application and are scalable for large-scale manufacture, although some of the formats do requiresignificant CMC optimization and longer developmenttimeline than the others.

Immunogenicity. Immunogenicity is one of the criticalfactors limiting clinical use of biological therapeutics, as thegeneration of ADA may lead to fast drug clearance, neutral-ization of therapeutic effect, and even severe adverse eventsin clinic. The duration of the ADA response can be cate-gorized into transient and persistent ADAs. The persistentADA requires the T cell help and commonly leads to moredeleterious consequences. The nature and the levels of theADA generated are influenced by both the patients’ physi-cal conditions (autoimmune-prone vs. immunosuppressive,pre-existing ADAs, etc.) and the intrinsic properties of anantibody (i.e., sequences, impurities, format, MOAs, dos-ing regimens, etc.) [162]. For example, the cancer patientsare usually immunosuppressive, while the patients withautoimmune diseases are prone to develop auto-reactiveantibodies and ADAs. The antibodies that contain strongT cell epitopes have high risk to induce T cell-dependentpersistent ADA. Compared to bsAbs that deplete B cells,the bsAbs that enhance immune system response may havea higher chance to induce ADA. And the bsAbs dosed bySC and intramuscular (IM) administration may be easier

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 53

to be picked up by dendritic cells (DCs) and present bsAb-derived peptides to T cells, as compared to the ones givenby IV infusion.

As reviewed by Davda et al. [163], using the approvedmAb clinical results, they found that although both ate-zolizumab and durvalumab were Fc-engineered anti-PD-L1 mAbs, only atezolizumab showed higher rate of ADA,as compared to the other anti-PD(L)1 mAbs. The combi-national treatment of the anti-PD(L)1 and anti-CTLA-4could increase the rate of ADA: the ADA rates againstnivolumab were increased from ∼12% (monotherapy) to24–38% (combo therapy with ipilimumab). Furthermore,the antibodies mediating B cell depletion usually hadlow ADA rates. As most of the reported bsAb formatsare heavily engineered and with non-native Ig sequencesintroduced, it is very likely that the bsAbs have higherimmunogenicity risk than regular mAbs. However, mostof the bsAb programs are still at early clinical stages, andonly very limited information and results are available toevaluate the immunogenicity issue for bsAbs. As mentionedabove, Aptevo developed APVO-414 (PSMA × CD3)using the ADAPTIR platform. In the initial Phase Idose escalation study, 58% of the patients developedADA with the titers as high as 1:250 000, leading to fastdrug clearance from the blood. By modifying the doseregimen from weekly IV to continuous IV infusion, theADA titers were decreased dramatically to the range of1:160–1:320. However, there were still 50% of the patientsdeveloped ADAs. Later, this program was discontinued asno therapeutic benefit was observed.

Due to its critical impact on the clinical outcomes,methods to minimize the immunogenicity risk have beenexploited at early discovery phase. Firstly, more and moreantibody therapeutics are utilizing humanized or evenfully human antibodies or fragments to decrease the non-human sequences, thereby decreasing the immunogenicityrisk. Secondly, in silico approaches are being employedto identify the immunogenic epitopes, especially T cellepitopes can be removed to prevent the generation of Tcell-dependent persistent ADAs. Though these approachesstill need to be validated in clinical practice, various in silicoalgorithms have been developed to predict the presenceof potential T cell epitopes. To compliment the in silicoprediction, in vitro assays are utilized, which include HLAbinding assays, primary peripheral blood mononuclear cell(PBMC) assays, mixed lymphocyte reaction assays, and 3Dmodels to mimic the conditions in specific tissues [164].The integrated results from in silico algorithm and in vitroassays can provide some suggestive information and helpthe bsAb development. For example, during rounds ofengineering and optimization, emicizumab adopted a de-immunization strategy that the effects of each mutation onimmunogenicity were evaluated by using algorithm, andany mutation that may increase immunogenicity risk wasavoided [127]. The clinical data suggested that there werelow level or no ADA observed in treated patients.

Pharmacokinetic and pharmacodynamic properties. PK,described as what the body does to a drug, refers to thedrug absorption, distribution, metabolism, and excretion.

PD, described as what the drug does to the body, involvesthe target binding and the following effect. The PK/PDprofiles play an important role in effecting the drug efficacyand safety and therefore are critical for the developmentof bsAbs. Many factors of the bsAbs can influence the PKprofiles, including molecule format, size, physicochemicalproperties, Fcγ R binding, as well as target binding affin-ity. For example, Harpoon is developing a novel protease-activated T cell engager platform, ProTriTAC based on theaforementioned TriTAC platform. By modifying the non-CDR region, the anti-albumin SDA can bind and mask theanti-CD3 arm while maintaining its binding to albumin.Furthermore, a tumor-associated protease cleavage site isintroduced to the linker between the anti-CD3 bindingdomain and anti-albumin SDA. In the circulation, theanti-albumin keeps anti-CD3 arm inactive and imparts themolecule long serum half-life. Once it enters into the TME,ProTriTACs are cleaved by tumor-associated proteases tolose the anti-albumin SDA and expose the anti-CD3 bind-ing site to function. If the cleaved molecules enter intothe circulation again, they will be rapidly cleared from thesystem due to its small size. By using this strategy, theydeveloped a ProTriTAC targeting EGFR, which was noteasy to be targeted by TRBAs due to its wide expression inthe normal tissue.

As long serum half-life may increase tissue penetrationand therapeutic efficacy, as well as require lower dosageand less frequent drug administration, sometimes, a longerserum half-life is preferred. IgGs and albumin are bothabundant in plasma with long half-life due to the bindingto FcRn, which rescues them from degradation in theendo/lysosomal compartment. Therefore, enhancing the Fcbinding to FcRn, or by adding a HSA binding domaininto the format (without Fc), is commonly used by bsAbsto improve the serum PK. Many mutations in the CH2-CH3 region have been tested to increase the Fc binding toFcRn, yet only the YTE and LS mutation combinations(YTE = M252Y/S254T/T256E; LS = M428L/N434S) havebeen clinically validated [165]. YTE mutations can increasethe antibody serum half-life ∼4-fold in human, but alsodecrease the ADCC activity of the antibody. VRC01LScontaining the LS mutations also showed more than 4-foldincrease in serum half-life in human [166]. Unlike YTEmutations, LS mutations have no impact on antibody’sADCC activity. On the other hand, in some applications,when the prolonged half-life is undesired, mutations todecrease the Fc to FcRn binding can also be applied.Detailed methods in modulating FcRn binding to modifyPK were reviewed by Leipold [167]. Though the effect ofFcRn on influencing serum half-life has been well studied,it still remains controversial on how it affects the drugmetabolism in other tissues, such as the brain and eyes. Asmatter of fact, Lucentis (Fab) and Eylea (Fc fusion protein)only showed slightly different ocular half-life in humans,suggesting FcRn binding may not play a major role indetermining ocular half-life, while the molecular size mayplay some, but not determining roles on PK properties ofmolecules in retina.

Due to the high binding affinity to the target, target-mediated drug disposition (TMDD) is common forantibody-based therapeutics, especially for those targeting

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

54 Antibody Therapeutics, 2020

surface antigens. As mentioned in the previous section,decreasing the target binding affinity, in some cases, canprolong the drug half-life and therefore improve the thera-peutic efficacy. As shown by Leong et al., the relationshipbetween CD3 affinity of the CD3 × CLL1 TRBA to itsactivity, PK and safety are quite complicated. During invitro characterization, they found that the one with lowaffinity to CD3 (CLL1/CD3L) showed decreased potency,but had more favorable safety profiles, as compared tothe one with high affinity to CD3 (CLL1/CD3H). Moreimportantly, when they tested these molecules in vivo, theyfound that CLL1/CD3L had slower drug clearance (50%)and increased drug exposure, which led to more durableantitumor responses, as compared to CLL1/CD3H [139].A similar case was also observed in an IL-15/Rα × PD-1 bifunctional protein. The fusion protein was engineeredto decrease the IL-15/Rα potency, thereby decrease theantigen sink, and increase half-life. Several variants withdecreased potency were generated and compared in vivo. Asthey predicted, the low potency variants showed dramatichalf-life extension from 0.5 day (wild type) to 9 days[US20180118828].

Besides affecting the serum PK, target binding affinitymay also influence the tumor/tissue distribution of thebsAbs. For example, the affinity of the bsAb to the tumorantigen can influence the tumor penetration. BsAbs withextremely high affinity to tumor antigen get stuck at theentrance and therefore have poor tumor penetration [168,169]. While low-affinity bsAbs distribute further into thetumor, but bsAbs with small sizes may have decreasedretention time in the tumor. The distribution of TRBAsfor solid tumors, as predicted by Friedrich et al., may besignificantly affected by the distribution of T cells, andmodifying the affinity to CD3 or TAA may not be sufficientto accumulate TRBAs and T cells into the tumor [170].Other methods may be used to affect the bsAb distributioninside the tumor tissue and include target selection, Fc, andutilizing of transcytosis [155].

The ultimate goal of all previously discussed strategies tomodulate PK was to enhance the overall clinical efficacyand/or to minimize the toxicity of the therapeutic bsAbs.Similarly, improving the PD profiles can also be achieved bymodifying the antigen-binding activity and by modifyingthe Fc-mediated effector function to further increase clini-cal potential of the bsAbs. Thus, the PK/PD profiles can bemodified by adjusting multiple factors, while most of thesefactors are interdependent, which highlight the inherentchallenges in therapeutic antibody design, and improvingone property can sometimes affect the others. Therefore,we should bear in mind that due to the complexity of theMOAs of bsAbs, the PK/PD profiles may not be the same aswe expected (hoped). Robust technologies and tools (bothexperimental and in silico) are critically needed to advancethe understanding of structural determinants of the bsAbsthat can impact the PK/PD properties and to guide theoptimization of bsAbs.

Efficacy and safety. A reasonable efficacy/safety win-dow is fundamental for a good clinical candidate; andPK/PD profiles, efficacy, and safety profiles commonly

influence each other. It is common that the drug showinghigh potency in discovery stage tends to be selected as thetherapeutic candidate. However, highly potent drug thatinduces toxicity at low dose leaves no or very limited ther-apeutic window, which may significantly hinder its clinicalapplication. On the other hand, the drug with a reasonablepotency but better safety profile may have wide therapeuticwindow, and the therapeutic efficacy may be improved byreadily increasing the dose without inducing significanttoxicity. Increasing drug exposure may be another wayto enhance the efficacy and prolong the response dura-tion, as we discussed above. However, increased systemicexposure may also increase the chance and the severityof adverse event. It is hard to predict which compositionof the MPP can translate into an optimal TPP in clinicalapplication.

For example, despite its extreme potency in eliminatingthe tumor cells, the life-threatening adverse effect associ-ated with the treatment of blinatumomab, as well as shortserum half-life, both significantly limit the application ofblinatumomab [171]. To improve the therapeutic efficacyand prolong the serum half-life, Affimed developed AFM-11 (Fv + Fv, 2 + 2), a tetravalent CD19 × CD3 bsAb [146].In vitro characterization studies showed that AFM-11 wasmore potent than BiTE molecule to elicit target cell killing.Though bivalent for CD3, AFM-11 showed stringenttarget-dependent activation of T cells. Using a NOD/SCIDxenograft model, AFM-11 showed favorable PK profileswith preferential tumor localization over normal tissue anda half-life of ∼20 h. In Phase I dose escalation study, AFM-11 was dosed by continuous infusion (Week 1, 0.7 ng/kg/wkto 130 ng/kg/wk; Week 2+, 2 ng/kg/wk to 400 ng/kg/wk).During the study, among the 14 patients who completedthe dose limiting toxicity observation period, 3 patientsshowed complete response (CR), but 2 were transient andpatients relapsed after cycle 2. Serum half-life was rangedfrom 7.14 to 10.6 h in four evaluable patients. Although nocytokine release syndrome (CRS) was observed, two Grade3 neurotoxicity and one fatal event were recorded in the twohighest dose groups. AFM-11 was placed on clinical hold,due to the severe adverse events.

TRBAs in formats containing Fc may have improvedstability and manufacture profile, as well as prolongedserum half-life. The long-term drug exposure may provideimproved efficacy and more flexible dosing strategy, butmay be more difficult to handle if undesired effect is expe-rienced. Regeneron developed REGN-1979 (Fab + Fabwith Fc, 1 + 1), a CD20 × CD3 bsAb. In vitro assaysshowed that REGN-1979 can effectively and specificallymediate the killing of CD20+ cells. The preclinical phar-macology studies using cynomolgus monkeys showed thatREGN-1979 can cause durable and deep B cell depletionwith a serum half-life of ∼14 days [31]. In June 2019,Regeneron reported the early-stage dose escalation trialresults of REGN-1979: 93% ORR and 71% CRR in 14patients with follicular lymphoma treated with REGN-1979 (5–320 mg); and 57% ORR in 7 patients with diffuselarge B cell lymphoma (DLBCL) treated with REGN-1979(80–160 mg), which were all CR. Among the total of 81evaluable patients, 7% experienced Grade 3 or higher CRS,and at least 10% of patients experienced Grade 3 or higher

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 55

adverse event. The incidence and severity of CRS can bemitigated by optimized premedication. Recently, in 2019ASH annual meeting, similar results were also reportedfor mosunetuzumab (CD20 × CD3, Roche) with ORRand CRR of 62.7 and 43.3%, respectively, in patients withslow-growing non-Hodgkin lymphoma. Both REGN-1979and mosunetuzumab showed benefit to patients who haddisease progressed post CAR-T therapies.

The comprehensive review regarding TRBAs publishedby Ellerman made a perfect case of how complex it canbe to optimize a TRBA, and the change of a factor of thebsAb may influence multiple profiles of the molecule, anda molecule profile can be modulated by multiple factors.For example, to uncouple the capabilities of TRBAs toinduce cytotoxic killing and cytokine production by theT cells, the TRBAs can be modified by (1) decreasing theaffinity to CD3, as T cell cytotoxic killing requires a loweractivation threshold; (2) using a different CD3 bindingepitope, as based on the “permissive geometry” model,different binding epitope may lead to different CD3 confor-mational change and T cell signaling; and (3) switching toanother format. In another case, to distinguish the antigen-overexpressing tumor cells and the low-expression normalcells, one can (1) decrease the binding affinity and usemultivalency to the antigen and (2) increase the distance ofthe IS, by either choosing a membrane distal epitope on theantigen or using a format with longer distance between thetwo binding domain. From another aspect, decreasing theaffinity of CD3 may diminish the target cell killing potencyin vitro; it may also increase the PK profile and tumor accu-mulation which ends up with comparable or even improvedin vivo efficacy and therapeutic window [108].

Another group of bsAbs that represents with challengesin leveraging the safety and efficacy is the agonistic bsAbtargeting co-stimulatory receptors, such as 4-1BB. Recently,the results reported for PRS-343 showed first sign of hopefor development anti-4-1BB treatment (see above). Numabdeveloped ND-021, a monovalent trispecific antibody tar-geting PD-L1, 4-1BB, and HSA. The in vitro efficacy testssuggested that the ultrahigh affinity (2 × 10−12 M) to PD-L1determined the potency of the molecule; binding to a distalepitope on 4-1BB can promote the 4-1BB clustering moreeffectively; and when the affinity to 4-1BB was way lowerthan the affinity to PD-L1, the effective dose range can besignificantly extended. As compared to the combinationsof mAbs, ND-021 showed superior activity in enhancingactivated T cell responses. Due to the monovalency and lackof Fc region, ND-021 displayed strictly PD-L1-dependent4-1BB activation and spared antigen-presenting cells fromdepletion. In in vivo efficacy tests, ND-021 showed higherantitumor activity than combined treatment with mAbs inmice. Most importantly, ND-021 did not induce liver toxi-city, and systemic T cell activation in cynomolgus monkeyposts a single-dose IV injection [172] although it remainselusive how this may translate into safety in humans. Cur-rently, this program is at IND-enabling study stage, and weare looking forward to see its clinical results.

On the basis of the strong biological rationale, empow-ered by the carefully harmonized format, and with themeticulously selected binding units, bispecific moleculesjust finish the first step to its final success. A good bispecific

clinical candidate not only needs to show promising thera-peutic potential but also needs to have good physiochemicalproperties and scalable manufacturability. Furthermore,favorable PK properties and low immunogenicity are alsocritical to assure the success of the candidate. Besides allthe above mentioned factors, the efficacy/safety ratio isone of the major determinants whether a bsAb moves intodevelopment stages in the end.

KEY CHALLENGES THE FIELD STILL FACING

Though bsAbs development has made significant progressand several strategies have been exploited to solve some ofthe challenges, many still remain. We would like to reviewthese challenges in two categories: technical challenges andmechanistic or biology challenges.

Technical challenges

Discovery. Compared to mAbs, bsAbs display signifi-cant complexity in the research and development stages.Special testing systems are needed to characterize thepotential therapeutic efficacy, toxicity, and PK/PD profilesof the bsAb therapeutic candidates, and many of thesesystems may be quite complicated, as compared to thesystems used to evaluate mAbs.

For example, artificial cell line used to evaluate bsAbfunction needs to overexpress both targets and include bothsignaling pathways, and the generation of such cell line mayhave huge technical challenges. Also, the expression leveland temporal order of the two targets on the artificial cellline may not reflect the disease situation in human. Forprimary cell-based efficacy tests, a specific population ofcells may need to be isolated and cultured ex vivo to inducethe expression of both targets, which makes the assaysextremely time- and cost-consuming and low throughput.Furthermore, even though researchers try to mimic thereal situation under which the bsAb plays its functionalroles, the in vitro assay system cannot completely reflect theimmune system, and therefore the effect of the bsAb cannotbe accurately evaluated in vitro.

The selection of species and relevant disease model forefficacy, pharmacology, and toxicology studies can be com-plicated, with considerations for the properties of both tar-gets, such as the cross-species specificity, the functionalityof the bsAb, as well as the expression and function of thetargets. Although, transgenic animals and animals graftedwith human immune systems are developed for bsAbs with-out cross-species binding, it is still doubtful how closelythese models can reflect the actual clinical conditions andhow accurately they predict the therapeutic efficacy, safetyrisk, and PK/PD profiles of a bsAb.

CMC. With the advanced protein engineering technol-ogy and elegantly designed bispecific formats, the physio-chemical properties and manufacturability are no longersignificant hurdles in developing bispecific clinical candi-dates. However, different formats do vary in the degreesof difficulty in Chemistry, Manufacturing and Controls(CMC) development, and the ones that fulfill developabil-

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

56 Antibody Therapeutics, 2020

ity criteria no doubt would significantly lower developmentrisk and shorten development timeline.

Preclinical pharmacology and toxicology. The preclinicalpharmacology and toxicology studies are very critical forthe development of bsAbs, as the results from these studiesnot only support the scientific rationale of the bsAbs butalso provide valuable information for selecting the FIHdose. Though the scope of the bsAb preclinical studies maybe similar to that for mAbs, the selection of the relevantspecies may be more challenging for bsAbs due to theadditional target. The relevant species should be selectedbased on the following: (1) both targets should have similarexpression profiles and biological functions as the targetsin human, respectively, and (2) the bsAb should bind toboth targets with similar properties as it binds to the humantargets. In the case that Fc effector function is required,especially the ones with modified binding to Fcγ Rs, theselected species should also be able to predict the Fc func-tion in human. If such a species is available, the FIHdose may be selected based on the no-observed-adverse-effect level (NOAEL). If a relevant species is not available,in vitro pharmacology studies and in vivo pharmacologystudies using surrogate bsAb or transgenic animals maybe required to provide supporting information. The FIHdose may be selected by using the minimum anticipatedbiological effect level (MABEL) approach if no relevanttoxicity species are available, especially for molecules withagonistic activities. Several case studies of the bsAb preclin-ical studies were reviewed by Prell et al. [173] and by Trivediet al. [174] to illustrate the complexity and challenge duringbsAb preclinical development.

Clinical development. Based on the draft guidance forbsAb development programs published by the FDA inApril 2019, several factors should be considered duringbsAb clinical development: (1) scientific rationale (e.g.,MOA, therapeutic advantages over standard of care); (2)mode of action (e.g., bridge two target cells, simultaneousor sequential binding); (3) binding kinetics to each target;(4) special pharmacology studies (e.g., PK/PD assessmentfor active form of the bsAb, immunogenicity assessment foreach domain of the bsAb); and (5) in certain cases, factorialdesign of clinical trials to inform risk/benefit ratio.

TGN1412, an anti-CD28 agonistic antibody case, alertedus that cautions must be taken in regard to clinical develop-ment of bsAbs with novel MOAs, especially for agonisticmolecules. Therefore, it is recommended that for bsAbsplaying agonistic function, especially for unprecedentedtarget pairs, the selection of the initial dose of the FIHtrial should use MABEL approach. Additionally, agonis-tic bsAbs may have a bell-shaped dose-response that thetherapeutic efficacy peaks at a dose that receptor occu-pancy is not saturated and then decreases along with theincreased drug dose [175]. Therefore, an agonistic bsAbwith a narrowed bell-shaped dose-response curve may besignificantly difficult for researchers to select the optimaldoses for different patients.

Comprehensive examinations for anti-drug antibodiesmay be required to evaluate the immunogenicity risk of

different domains of the bsAbs, as some of the bsAbsare heavily engineered with potential immunogenic epitopeintroduced. Special attention needs to be taken on ADAagainst TRBAs and agonistic bsAbs using the tumor/tissuelocalization strategy, as the presence of ADA may break theTAA dependency of these bsAbs and lead to non-specificactivation of immune cells and unpredictable severe adverseevent. One should always follow FDA outlined and rec-ommended adoption of a risk-based approach to evaluateand mitigate immune responses or adverse immunologi-cally related responses associated with therapeutic proteinproducts that affect their safety and efficacy during clinicaldevelopment of a bsAb.

Furthermore, in some instances, combinational therapyprovides the flexibility in adjusting the dosing regimen,which cannot be achieved by bsAbs. Although variousbispecific formats can provide some degree of flexibility inadjusting affinity and valency of a binding specificity tosuit different needs, once the format is determined, the ratioagainst two targets is fixed, and it cannot be adjusted basedon the clinical results, which may pose clinical developmentchallenge for a drug. Moreover, an optimal treatment mayrequire sequential target intervention. For example, theconcurrent treatment of anti-PD-1 with anti-OX40 treat-ment leads to substantial increase in serum cytokines andthe expression of inhibitory receptors on T cells, as well asdecreased T cell proliferation, thereby attenuating the anti-tumor efficacy of anti-OX40 treatment. However, delayingthe PD-1 treatment can increase the antitumor activity ofanti-OX40 treatment [176]. In another case, NK cells canbe activated and upregulate 4-1BB expression by exposingto rituximab-coated CD20+ tumor cells or trastuzumab-coated Her2-overexpressing breast cancer cells. The anti-4-1BB treatment following the treatment of rituximab ortrastuzumab can enhance the ADCC effect of NK cellsto antibody-coated tumor cells [177, 178]. In such cases,the combinational therapy with mAbs offers the flexibilitywhich cannot be accomplished by current bsAb strategies.

Mechanistic or biology challenges

The most fascinating applications of bsAbs are to enablenovel biological function and therapeutic MOA otherwiseimpossible by using mAbs alone or in combination. How-ever, the novel MOA may also impose unknown safety riskon bsAbs, which cannot be readily predicted or evaluatedin preclinical studies, and possibly result in severe or evenlife-threatening adverse events during the clinical stage.Therefore, the uncertainty in function and safety of thesebsAbs represents a major challenge for development ofbsAb therapeutics.

When selecting the target pair, researchers shouldconsider the spatial and temporal presence of both targets.Whether both targets are expressed at the same location atthe same time? Whether their levels are within a reasonablerange that can be effectively treated by a bsAb with fixedstoichiometry? Whether the two targets expressed on differ-ent cells or on the same cells? Whether the bsAb will medi-ate in-cis or in-trans engagement of the two targets? Willdifferent engagement models result in different outcomes

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 57

in efficacy and safety? Those are all important questionsone need to think through when embarking a bsAb project.

Bispecific antibodies engaging CD32B and FcεR weredesigned to employ the dominant negative role of CD32Band inhibit the activation of FcεR to alleviate IgE-mediateddiseases. The bsAb 9202.1/5411 with IgG1 format wasproduced using Escherichia coli cell line and therefore hadno Fc effector function due to lack of glycosylation. In vitroanalysis showed that this bsAb can inhibit IgE-mediatedactivation of mast cells and basophils. As mentioned by theauthors, several formats that were bivalent for FcεR mightcross-link FcεR in the absence of CD32B, thereby activat-ing rather than inhibiting FcεR [179]. One may speculatein the worst scenario in vivo, sometime may be inevitable,if such a molecule formed aggregates, it may function toactivate rather than inhibit FcεR as one initially designed.

This becomes even more complicated for agonistic bsAbsto activate receptors. As a receptor is co-evolved with itscognate ligand, the signaling upon ligand-receptor engage-ment is evolved to be tightly controlled under physiologicalconditions. Due to the plasticity of receptors, polygamywidely exists for ligand-receptor interaction. When usingantibody-based therapeutics to mimic the function of aligand, the antibody may bind to the site on the receptor dif-ferent from its cognate ligand binding site, which may elicitdifferent signals. The deviation from the cognate activationmay result in unexpected consequence, and their potentialsafety risk is unknown.

For example, as reported by Gu et al., a panel ofbiparatopic anti-Her2 antibodies in DVD-Ig formatgenerated from the same parental mAbs only differedby VD orientations or linker length. Surprisingly, DVD-Ig molecules with one VD orientation showed agonisticeffect and increased tumor cell proliferation, whereasmolecules with the opposite VD orientation remainedantagonistic. Further studies revealed that a particularVD orientation interrupted Her2/EGFR and Her2/Her3interaction, resulting in increased Her2 homodimerizationand activation [180]. Similarly, a biparatopic anti-CTLA-4 bsAb unexpectedly changed the signalosome assemblyon the cytoplasmic domain of CTLA-4 and completelyconverted the inhibitory receptor into a stimulatoryreceptor [181].

The preclinical and clinical development path havelargely paved for bsAbs with precedent mechanisms.However, the development of bsAbs with novel biologicalmechanisms still faces a few challenges and pitfalls. It mayrequire more preclinical studies and early discussion withregulatory agencies for clinical development plans. Webelieve that, in the future, biology will be the key driver fordesign and selection of a bsAb and the key considerationfor clinical development of bsAb drugs.

PERSPECTIVE

A growing number of recombinant bsAbs are now in clini-cal development. These bsAbs represent quite different for-mats. The number of the formats may reflect the diversityin desired features of therapeutic applications and may alsoreflect the different understanding of biology. For instance,

for T cell-redirected cytotoxicity, a variety of formats, withdifference in affinity, valency, domain geometry, Fc proper-ties, and pharmacokinetic properties, have progressed intoclinical development. It will be interesting to see clinicalvalidation of various preclinical rationales behind thedesign of those molecules in the coming years.

With the advent of gene therapy, RNA therapy, celltherapy, and various other new therapies, we should alwayscompare those different therapeutic options and pay closeattention to those new therapeutic modalities that mayhave disruptive potentials, for instance, both chimericantigen receptors T cell (CAR-T) therapy and TRBAs havedemonstrated dramatic effects in patients with hematologictumors. One TRBA and two CAR-T cell products havebeen approved by major regulatory agencies within thelast 10 years for the treatment of hematological cancers,and an additional approximately 60 TRBAs and 300 CARcell constructs are in clinical trials today. CAR-Ts aredesigned to activate T cells via intracellular T cell co-stimulatory signaling modules in tandem and to form acytolytic synapse with target cells that is very differentfrom the classical immune synapse both physically andmechanistically, whereas the TRBA-induced synapse issimilar to the classic immune synapse by bringing T cellsclose proximity to tumor cells via a bispecific molecule.As published in 2018 ASH annual meeting, in patientswith relapsed and refractory multiple myeloma (r/r MM),AMG-420 (BCMA × CD3, BiTE) showed 70% ORRand 40% CRR. Similarly, bb-2121 (BCMA CAR-T) andJCARH125 also demonstrated ∼80% ORR and ∼30%CRR. On the other hand, both TRBAs and CAR-Ttherapies showed similar adverse effect, which may bedue to their MOA in redirecting T cell cytotoxicity totumor cells. Blincyto and CAR-T therapies, Kymriah andYescarta, are all targeting CD19+ tumor cells and provedfor treatment of B cell lymphomas, and all of them have theblock box warnings for CRS and neurological toxicities.From the manufacturing aspect, due to the characteristicsof BiTE molecules, the manufacture of Blincyto still hasquite a few challenges, but this has been solved by thenext generation of TRBAs in the clinical development.For autologous CAR-T therapies, a complicated and time-consuming (3–4 weeks) manufacturing process is requiredfor each patient. Additionally, as the CAR-T therapies arelive cells, the regulatory requirements for CAR-T therapiesare more complicated and stringent than regular biologicaltherapeutics. Most CAR-T cells today are autologous,although significant strides are being made to developoff-the-shelf allogeneic CAR-based products. Therefore,in general comparing these two therapeutic platforms,TRBAs are the off-the-shelf products and may be moreconvenient and affordable to patients in the near futurewhen more TRBAs are available, while CAR-T therapymay be tedious but may have advantage to mobilize theentire T cell machinery in a very different mechanism tofight cancer cells. Both platforms currently are facing thesame moderate anticancer effects in solid tumor settings,probably due to inaccessibility of immune effector cells tosolid tumors and complex immunosuppressive mechanismsat TME. The knowledge learned from clinical trials foreither one will definitely help to improve the design of both

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

58 Antibody Therapeutics, 2020

therapies with additional immunomodulatory features toovercome the key challenges they are still facing.

Nevertheless, bsAbs and msAbs open up tremendousopportunities to explore previously unexplored therapeuticoptions. We believe that the next decade will witnessthe clinical success of bsAbs or msAbs employing somenovel MOAs in the applications in cancer and infectious,metabolic, ocular, and other diseases with significant unmetmedical needs.

DECLARATIONS

Siwei Nie, Zhuozhi Wang, Jianqing Xu, and Jijie Gu arecurrent employees of WuXi Biologics and may hold WuXiBiologics’ stocks.

CONFLICT OF INTEREST STATEMENT

Siwei Nie, Zhuozhi Wang, Jianqing Xu and Jijie Gu arecurrent employees of WuXi Biologics, and may hold WuXiBiologics’ stocks.

ABBREVIATIONS:bsAb bispecific antibodymAb monoclonal antibodyMPP molecular product profileTPP target product profileMOA mechanism of actionUMN unmet medical needsTMDD target-mediated drug dispositionSDA single-domain antibodyFv variable fragmentscFv single-chain variable fragmentFab antigen-binding fragmentscFab single-chain antigen-binding fragmentVH heavy chain variable domainVL light chain variable domainCH1 heavy chain constant domain 1CH2 heavy chain constant domain 2CH3 heavy chain constant domain 3CH4 heavy chain constant domain 4Fc fragment of crystallizable regionFD the heavy chain of a Fab, i.e. VH domain plus

CH1 domainPK/PD pharmacokinetic/pharmacodynamicsTRBA T cell-redirecting bispecific antibodyTAA tumor-associated antigen

REFERENCES1. Husain, B, Ellerman, D. Expanding the boundaries of

biotherapeutics with Bispecific antibodies. BioDrugs 2018; 32:441–64.

2. Labrijn, AF, Janmaat, ML, Reichert, JM et al. Bispecificantibodies: a mechanistic review of the pipeline. Nat Rev DrugDiscov 2019; 18: 585–608.

3. Brinkmann, U, Kontermann, RE. The making of bispecificantibodies. MAbs 2017; 9: 182–212.

4. Harmsen, MM, Van Solt, CB, Fijten, HPD et al. Prolonged in vivoresidence times of llama single-domain antibody fragments in pigs

by binding to porcine immunoglobulins. Vaccine 2005; 23:4926–34.

5. Zhu, X, Wang, L, Liu, R et al. COMBODY: one-domain antibodymultimer with improved avidity. Immunol Cell Biol 2010; 88:667–75.

6. Del Bano, J, Florès-Florès, R, Josselin, E et al. A bispecificantibody-based approach for targeting Mesothelin in triple negativebreast cancer. Front Immunol 2019; 10: 1593.

7. Wolf, E, Hofmeister, R, Kufer, P et al. BiTEs: bispecific antibodyconstructs with unique anti-tumor activity. Drug Discov Today2005; 10: 1237–44.

8. Kim, S, Kim, H, Jo, DH et al. Bispecific anti-mPDGFRβ ×cotinine scFv-Cκ-scFv fusion protein and cotinine-duocarmycincan form antibody-drug conjugate-like complexes that exertcytotoxicity against mPDGFRβ expressing cells. Methods 2019;154: 125–35.

9. Tietz, J, Spohn, G, Schmid, G et al. Affinity and potency ofRTH258 (ESBA1008), a novel inhibitor of vascular endothelialgrowth factor a for the treatment of retinal disorders. InvestOphthalmol Vis Sci 2015; 56: 1501.

10. Egan, TJ, Diem, D, Weldon, R et al. Novel multispecificheterodimeric antibody format allowing modular assembly ofvariable domain fragments. MAbs 2017; 9: 68–84.

11. Moore, PA, Zhang, W, Rainey, GJ et al. Application of dualaffinity retargeting molecules to achieve optimal redirected T-cellkilling of B-cell lymphoma. Blood 2011; 117: 4542–51.

12. Veri, M-C, Burke, S, Huang, L et al. Therapeutic control of B cellactivation via recruitment of Fcγ receptor IIb (CD32B) inhibitoryfunction with a novel bispecific antibody scaffold. Arthritis Rheum2010; 62: 1933–43.

13. Johnson, S, Burke, S, Huang, L et al. Effector cell recruitment withnovel Fv-based dual-affinity re-targeting protein leads to potenttumor cytolysis and in vivo B-cell depletion. J Mol Biol 2010; 399:436–49.

14. McAleese, F, Eser, M. RECRUIT-TandAbs®

: harnessing theimmune system to kill cancer cells. Futur Oncol 2012; 8:687–95.

15. Davé, E, Adams, R, Zaccheo, O et al. Fab-dsFv: a bispecificantibody format with extended serum half-life through albuminbinding. MAbs 2016; 8: 1319–35.

16. Bhatta, P, Humphreys, DP. Relative contribution of frameworkand CDR regions in antibody variable domains to multimerisationof Fv- and scFv-containing bispecific antibodies. Antibodies (Basel,Switzerland) 2018; 7: 35.

17. Schoonjans, R, Willems, A, Schoonooghe, S et al. Fab chains as anefficient heterodimerization scaffold for the production ofrecombinant bispecific and trispecific antibody derivatives. JImmunol 2000; 165: 7050–7.

18. Lin, L, Li, L, Zhou, C et al. A HER2 bispecific antibody can beefficiently expressed in Escherichia coli with potent cytotoxicity.Oncol Lett 2018; 16: 1259–66.

19. Harwood, SL, Alvarez-Cienfuegos, A, Nuñez-Prado, N et al.ATTACK, a novel bispecific T cell-recruiting antibody withtrivalent EGFR binding and monovalent CD3 binding for cancerimmunotherapy. Oncoimmunology 2017; 7: e1377874–4.

20. Ridgway, JBB, Presta, LG, Carter, P. ‘Knobs-into-holes’engineering of antibody CH3 domains for heavy chainheterodimerization. Protein Eng Des Sel 1996; 9: 617–21.

21. Atwell, S, Ridgway, JBB, Wells, JA et al. Stable heterodimers fromremodeling the domain interface of a homodimer using a phagedisplay library. J Mol Biol 1997; 270: 26–35.

22. Spiess, C, Bevers, J 3rd, Jackman, J et al. Development of a humanIgG4 bispecific antibody for dual targeting of interleukin-4 (IL-4)and interleukin-13 (IL-13) cytokines. J Biol Chem 2013; 288:26583–93.

23. Gunasekaran, K, Pentony, M, Shen, M et al. Enhancing antibodyFc heterodimer formation through electrostatic steering effects:applications to bispecific molecules and monovalent IgG. J BiolChem 2010; 285: 19637–46.

24. Moore, GL, Bautista, C, Pong, E et al. A novel bispecific antibodyformat enables simultaneous bivalent and monovalentco-engagement of distinct target antigens. MAbs 2011; 3: 546–57.

25. Von Kreudenstein, TS, Escobar-Carbrera, E, Lario, PI et al.Improving biophysical properties of a bispecific antibody scaffold

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 59

to aid developability: quality by molecular design. MAbs 2013; 5:646–54.

26. Davis, JH, Aperlo, C, Li, Y et al. SEEDbodies: fusion proteinsbased on strand-exchange engineered domain (SEED) CH3heterodimers in an Fc analogue platform for asymmetric binders orimmunofusions and bispecific antibodies. Protein Eng Des Sel2010; 23: 195–202.

27. Skegro, D, Stutz, C, Ollier, R et al. Immunoglobulin domaininterface exchange as a platform technology for the generation ofFc heterodimers and bispecific antibodies. J Biol Chem 2017; 292:9745–59.

28. Tustian, AD, Endicott, C, Adams, B et al. Development ofpurification processes for fully human bispecific antibodies basedupon modification of protein A binding avidity. MAbs 2016; 8:828–38.

29. Dimasi, N, Fleming, R, Sachsenmeier, KF et al. Guiding bispecificmonovalent antibody formation through proteolysis of IgG1single-chain. MAbs 2017; 9: 438–54.

30. Merchant, AM, Zhu, Z, Yuan, JQ et al. An efficient route tohuman bispecific IgG. Nat Biotechnol 1998; 16: 677–81.

31. Smith, EJ, Olson, K, Haber, LJ et al. A novel, native-formatbispecific antibody triggering T-cell killing of B-cells is robustlyactive in mouse tumor models and cynomolgus monkeys. Sci Rep2015; 5: 17943.

32. De, Nardis, C, Hendriks, LJA, Poirier, E et al. A new approach forgenerating bispecific antibodies based on a common light chainformat and the stable architecture of human immunoglobulin G(1).J Biol Chem 2017; 292: 14706–17.

33. Shiraiwa, H, Narita, A, Kamata-Sakurai, M et al. Engineering abispecific antibody with a common light chain: identification andoptimization of an anti-CD3 epsilon and anti-GPC3 bispecificantibody, ERY974. Methods 2019; 154: 10–20.

34. Fischer, N, Elson, G, Magistrelli, G et al. Exploiting light chainsfor the scalable generation and platform purification of nativehuman bispecific IgG. Nat Commun 2015; 6: 6113.

35. Lewis, SM, Wu, X, Pustilnik, A et al. Generation of bispecific IgGantibodies by structure-based design of an orthogonal Fabinterface. Nat Biotechnol 2014; 32: 191.

36. Froning, KJ, Leaver-Fay, A, Wu, X et al. Computational design ofa specific heavy chain/κ light chain interface for expressing fullyIgG bispecific antibodies. Protein Sci 2017; 26: 2021–38.

37. Mazor, Y, Oganesyan, V, Yang, C et al. Improving target cellspecificity using a novel monovalent bispecific IgG design. MAbs2015; 7: 377–89.

38. Vaks, L, Litvak-Greenfeld, D, Dror, S et al. Design principles forbispecific IgGs, opportunities and pitfalls of artificial disulfidebonds. Antibodies (Basel, Switzerland) 2018; 7: 27.

39. Litvak-Greenfeld, D, Vaks, L, Dror, S et al. “BIClonals”:production of bispecific antibodies in IgG format in transientlytransfected mammalian cells. In: Steinitz, M (ed). Methods inMolecular Biology. New York, NY: Springer, 2019, 431–54

40. Liu, Z, Leng, EC, Gunasekaran, K et al. A novel antibodyengineering strategy for making monovalent bispecificheterodimeric IgG antibodies by electrostatic steering mechanism. JBiol Chem 2015; 290: 7535–62.

41. Dillon, M, Yin, Y, Zhou, J et al. Efficient production of bispecificIgG of different isotypes and species of origin in single mammaliancells. MAbs 2017; 9: 213–30.

42. Bönisch, M, Sellmann, C, Maresch, D et al. Novel CH1:CLinterfaces that enhance correct light chain pairing in heterodimericbispecific antibodies. Protein Eng Des Sel 2017; 30: 685–96.

43. Seifert, O, Rau, A, Beha, N et al. Diabody-Ig: a novel platform forthe generation of multivalent and multispecific antibody molecules.MAbs 2019; 11: 919–29.

44. Schaefer, W, Regula, JT, Bähner, M et al. Immunoglobulin domaincrossover as a generic approach for the production of bispecific IgGantibodies. Proc Natl Acad Sci USA 2011; 108: 11187–92.

45. Metz, S, Panke, C, Haas, AK et al. Bispecific antibody derivativeswith restricted binding functionalities that are activated byproteolytic processing. Protein Eng Des Sel 2012; 25:571–80.

46. Shen, J, Vil, MD, Jimenez, X et al. Single variable domain-IgGfusion: a novel recombinant approach to Fc domain-containingbispecific antibodies. J Biol Chem 2006; 281: 10706–14.

47. Shen, J, Vil, MD, Jimenez, X et al. Single variable domain antibodyas a versatile building block for the construction of IgG-likebispecific antibodies. J Immunol Methods 2007; 318: 65–74.

48. Shi, SY, Lu, Y-W, Liu, Z et al. A biparatopic agonistic antibodythat mimics fibroblast growth factor 21 ligand activity. J Biol Chem2018; 293: 5909–19.

49. Wozniak-Knopp, G, Bartl, S, Bauer, A et al. Introducingantigen-binding sites in structural loops of immunoglobulinconstant domains: Fc fragments with engineeredHER2/neu-binding sites and antibody properties. Protein Eng DesSel 2010; 23: 289–97.

50. Everett, KL, Kraman, M, Wollerton, FPG et al. Generation ofFcabs targeting human and murine LAG-3 as building blocks fornovel bispecific antibody therapeutics. Methods 2019; 154:60–9.

51. Wu, C, Ying, H, Grinnell, C et al. Simultaneous targeting ofmultiple disease mediators by a dual-variable-domainimmunoglobulin. Nat Biotechnol 2007; 25: 1290–7.

52. Dickopf, S, Lauer, ME, Ringler, P et al. Highly flexible,IgG-shaped, trivalent antibodies effectively target tumor cells andinduce T cell-mediated killing. Biol Chem 2019; 400: 343.

53. Coloma, MJ, Morrison, SL. Design and production of noveltetravalent bispecific antibodies. Nat Biotechnol 1997; 15: 159–63.

54. Hoseini, SS, Guo, H, Wu, Z et al. A potent tetravalentT-cell-engaging bispecific antibody against CD33 in acute myeloidleukemia. Blood Adv 2018; 2: 1250–8.

55. Moore, GL, Bernett, MJ, Rashid, R et al. A robust heterodimericFc platform engineered for efficient development of bispecificantibodies of multiple formats. Methods 2019; 154: 38–50.

56. Yu, S, Zhang, J, Yan, Y et al. A novel asymmetricalanti-HER2/CD3 bispecific antibody exhibits potent cytotoxicity forHER2-positive tumor cells. J Exp Clin Cancer Res 2019; 38: 355.

57. Qi, J, Li, X, Peng, H et al. Potent and selective antitumor activity ofa T cell-engaging bispecific antibody targeting amembrane-proximal epitope of ROR1. Proc Natl Acad Sci USA2018; 115: E5467–76.

58. Robinson, HR, Qi, J, Cook, EM et al. A CD19/CD3 bispecificantibody for effective immunotherapy of chronic lymphocyticleukemia in the ibrutinib era. Blood 2018; 132: 521–32.

59. Bezabeh, B, Fleming, R, Fazenbaker, C et al. Insertion of scFv intothe hinge domain of full-length IgG1 monoclonal antibody resultsin tetravalent bispecific molecule with robust properties. MAbs2017; 9: 240–56.

60. Manikwar, P, Mulagapati, SHR, Kasturirangan, S et al.Characterization of a novel bispecific antibody with improvedconformational and chemical stability. J Pharm Sci 2019. doi:10.1016/j.xphs.2019.06.025.

61. Kim, HS, Dunshee, DR, Yee, A et al. Tethered-variable CLbispecific IgG: an antibody platform for rapid bispecific antibodyscreening. Protein Eng Des Sel 2017; 30: 627–37.

62. Gong, S, Ren, F, Wu, D et al. Fabs-in-tandem immunoglobulin is anovel and versatile bispecific design for engaging multipletherapeutic targets. MAbs 2017; 9: 1118–28.

63. Bostrom, J, Yu, S-F, Kan, D et al. Variants of the antibodyHerceptin that interact with HER2 and VEGF at the antigenbinding site. Science (80-) 2009; 323: 1610–4.

64. Schaefer, G, Haber, L, Crocker, LM et al. A two-in-one antibodyagainst HER3 and EGFR has superior inhibitory activitycompared with monospecific antibodies. Cancer Cell 2011; 20:472–86.

65. Lee, CV, Koenig, P, Fuh, G. A two-in-one antibody engineeredfrom a humanized interleukin 4 antibody through mutation inheavy chain complementarity-determining regions. MAbs 2014; 6:622–7.

66. Hu, S, Fu, W, Xu, W et al. Four-in-one antibodies have superiorcancer inhibitory activity against EGFR, HER2, HER3, andVEGF through disruption of HER/MET crosstalk. Cancer Res2015; 75: 159–70.

67. Strop, P, Ho, W-H, Boustany, LM et al. Generating bispecifichuman IgG1 and IgG2 antibodies from any antibody pair. J MolBiol 2012; 420: 204–19.

68. Labrijn, AF, Meesters, JI, de Goeij, BECG et al. Efficientgeneration of stable bispecific IgG1 by controlled fab-armexchange. Proc Natl Acad Sci USA 2013; 110: 5145–50.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

60 Antibody Therapeutics, 2020

69. Schanzer, JM, Wartha, K, Croasdale, R et al. A novelglycoengineered bispecific antibody format for targeted inhibitionof epidermal growth factor receptor (EGFR) and insulin-likegrowth factor receptor type I (IGF-1R) demonstrating uniquemolecular properties. J Biol Chem 2014; 289: 18693–706.

70. Castoldi, R, Schanzer, J, Panke, C et al. TetraMabs: simultaneoustargeting of four oncogenic receptor tyrosine kinases for tumorgrowth inhibition in heterogeneous tumor cell populations. ProteinEng Des Sel 2016; 29: 467–75.

71. LaFleur, DW, Abramyan, D, Kanakaraj, P et al. Monoclonalantibody therapeutics with up to five specificities: functionalenhancement through fusion of target-specific peptides. MAbs2013; 5: 208–18.

72. Yin, W, Zhu, J, Gonzalez-Rivas, D et al. Construction of a novelbispecific antibody to enhance antitumor activity against lungcancer. Adv Mater 2018; 30: 1805437.

73. Harper, J, Adams, KJ, Bossi, G et al. An approved in vitroapproach to preclinical safety and efficacy evaluation of engineeredT cell receptor anti-CD3 bispecific (ImmTAC) molecules. PLoSOne 2018; 13: e0205491–1.

74. Siegemund, M, Schneider, F, Hutt, M et al. IgG-single-chainTRAIL fusion proteins for tumour therapy. Sci Rep 2018; 8: 7808.

75. Gantke, T, Weichel, M, Herbrecht, C et al. Trispecific antibodiesfor CD16A-directed NK cell engagement and dual-targeting oftumor cells. Protein Eng Des Sel 2017; 30: 673–84.

76. Hanahan, D, Weinberg, RA. Hallmarks of cancer: the nextgeneration. Cell 2011; 144: 646–74.

77. Ridgway, J, Zhang, G, Wu, Y et al. Inhibition of Dll4 signallinginhibits tumour growth by deregulating angiogenesis. Nature 2006;444: 1083–7.

78. Yan, M. Therapeutic promise and challenges of targetingDLL4/NOTCH1. Vasc Cell 2011; 3: 17.

79. Li, Y, Hickson, JA, Ambrosi, DJ et al. Abt-165, a dual variabledomain immunoglobulin (dvd-ig) targeting dll4 and vegf,demonstrates superior efficacy and favorable safety profiles inpreclinical models. Mol Cancer Ther 2018; 17: 1039–50.

80. Gordon, MS, Nemunaitis, JJ, Ramanathan, RK et al. Phase 1,open-label, dose-escalation and expansion study of ABT-165, adual variable domain immunoglobulin (DVD-Ig) targeting bothDLL4 and VEGF, in patients (pts) with advanced solid tumors. JClin Oncol 2016; 34: 2507.

81. Wainberg, Z, Strickler, J, Gordon, M et al. P-234 Phase 1bopen-label study evaluating the safety, pharmacokinetics, andpreliminary efficacy of ABT-165 plus FOLFIRI in patients withsecond-line (2L) colorectal cancer (CRC). Ann Oncol 2018; 29. doi:10.1093/annonc/mdy151.233.

82. Moores, SL, Chiu, ML, Bushey, BS et al. A novel bispecificantibody targeting EGFR and cMet is effective against EGFRinhibitor-resistant lung tumors. Cancer Res 2016; 76: 3942–53.

83. MacDonald, BT, He, X. Frizzled and LRP5/6 receptors forWnt//b-catenin signaling. Cold Spring Harb Perspect Biol 2012; 4:a007880.

84. Zinzalla, V, Drobits-Handl, B, Savchenko, A et al. AbstractDDT01-01: BI 905677: a first-in-class LRP5/6 antagonist targetingWnt-driven proliferation and immune escape. Cancer Res 2019; 79:DDT01-01.

85. Schadendorf, D, Hodi, FS, Robert, C et al. Pooled analysis oflong-term survival data from phase II and phase III trials ofIpilimumab in unresectable or metastatic melanoma. J Clin Oncol2015; 33: 1889–94.

86. Kwok, G, Yau, TCC, Chiu, JW et al. Pembrolizumab (Keytruda).Hum Vaccin Immunother 2016; 12: 2777–89.

87. Rotte, A. Combination of CTLA-4 and PD-1 blockers fortreatment of cancer. J Exp Clin Cancer Res 2019; 38: 255.

88. Gros, A, Robbins, PF, Yao, X et al. PD-1 identifies thepatient-specific CD8+ tumor-reactive repertoire infiltrating humantumors. J Clin Invest 2014; 124: 2246–59.

89. Yang, Z-Z, Kim, HJ, Villasboas, JC et al. Expression of LAG-3defines exhaustion of intratumoral PD-1(+) T cells and correlateswith poor outcome in follicular lymphoma. Oncotarget 2017; 8:61425–39.

90. Woo, S-R, Turnis, ME, Goldberg, MV et al. Immune inhibitorymolecules LAG-3 and PD-1 synergistically regulate T-cell function

to promote tumoral immune escape. Cancer Res 2012; 72:917–27.

91. Ascierto, PA, Melero, I, Bhatia, S et al. Initial efficacy ofanti-lymphocyte activation gene-3 (anti-LAG-3; BMS-986016) incombination with nivolumab (nivo) in pts with melanoma (MEL)previously treated with anti-PD-1/PD-L1 therapy. J Clin Oncol2017; 35: 9520.

92. Chester, C, Sanmamed, MF, Wang, J et al. Immunotherapytargeting 4-1BB: mechanistic rationale, clinical results, and futurestrategies. Blood 2018; 131: 49–57.

93. Claus, C, Ferrara, C, Xu, W et al. Tumor-targeted 4-1BB agonistsfor combination with T cell bispecific antibodies as off-the-shelftherapy. Sci Transl Med 2019; 11: eaav5989.

94. Lan, Y, Zhang, D, Xu, C et al. Enhanced preclinical antitumoractivity of M7824, a bifunctional fusion protein simultaneouslytargeting PD-L1 and TGF-β. Sci Transl Med 2018; 10: eaan5488.

95. Strauss, J, Heery, CR, Schlom, J et al. Phase I trial of M7824(MSB0011359C), a bifunctional fusion protein targeting PD-L1and TGFβ, in advanced solid tumors. Clin Cancer Res 2018; 24:1287–95.

96. Chelius, D, Ruf, P, Gruber, P et al. Structural and functionalcharacterization of the trifunctional antibody catumaxomab. MAbs2010; 2: 309–19.

97. Linke, R, Klein, A, Seimetz, D. Catumaxomab: clinicaldevelopment and future directions. MAbs 2010; 2: 129–36.

98. Dufner, V, Sayehli, CM, Chatterjee, M et al. Long-term outcomeof patients with relapsed/refractory B-cell non-Hodgkin lymphomatreated with blinatumomab. Blood Adv 2019; 3: 2491–8.

99. Kantarjian, H, Stein, A, Gökbuget, N et al. Blinatumomab versuschemotherapy for advanced acute lymphoblastic leukemia. N EnglJ Med 2017; 376: 836–47.

100. Davis, SJ, van der Merwe, PA. The kinetic-segregation model: TCRtriggering and beyond. Nat Immunol 2006; 7: 803–9.

101. Strohl, WR, Naso, M. Bispecific T-cell redirection versus chimericantigen receptor (CAR)-T cells as approaches to kill cancer cells.Antibodies 2019; 8: 41.

102. Clynes, RA, Desjarlais, JR. Redirected T cell cytotoxicity in cancertherapy. Annu Rev Med 2019; 70: 437–50.

103. Trabolsi, A, Arumov, A, Schatz, JH. T cell–activating bispecificantibodies in cancer therapy. J Immunol 2019; 203: 585–92.

104. Wong, R, Pepper, C, Brennan, P et al. Blinatumomab inducesautologous T-cell killing of chronic lymphocytic leukemia cells.Haematologica 2013; 98: 1930–8.

105. Bacac, M, Fauti, T, Sam, J et al. A novel carcinoembryonic antigenT-cell bispecific antibody (CEA TCB) for the treatment of solidtumors. Clin Cancer Res 2016; 22: 3286–97.

106. Rossi, DL, Rossi, EA, Cardillo, TM et al. A new class of bispecificantibodies to redirect T cells for cancer immunotherapy. MAbs2014; 6: 381–91.

107. Offner, S, Hofmeister, R, Romaniuk, A et al. Induction of regularcytolytic T cell synapses by bispecific single-chain antibodyconstructs on MHC class I-negative tumor cells. Mol Immunol2006; 43: 763–71.

108. Ellerman, D. Bispecific T-cell engagers: towards understandingvariables influencing the in vitro potency and tumor selectivity andtheir modulation to enhance their efficacy and safety. Methods2019; 154: 102–17.

109. Castella, B, Melaccio, A, Foglietta, M et al. Vγ 9Vδ2 T cells asstrategic weapons to improve the potency of immune checkpointblockade and immune interventions in human myeloma. FrontOncol 2018; 8: 508.

110. Brandes, M, Willimann, K, Bioley, G et al. Cross-presentinghuman gammadelta T cells induce robust CD8+ alphabeta T cellresponses. Proc Natl Acad Sci USA 2009; 106: 2307–12.

111. Hoeres, T, Smetak, M, Pretscher, D et al. Improving the efficiencyof Vγ 9Vδ2 T-cell immunotherapy in cancer. Front Immunol 2018;9: 800.

112. Oberg, H-H, Peipp, M, Kellner, C et al. Novel Bispecific antibodiesincrease γ δ T-cell cytotoxicity against pancreatic cancer cells.Cancer Res 2014; 74: 1349–60.

113. Reusch, U, Burkhardt, C, Fucek, I et al. A novel tetravalentbispecific TandAb (CD30/CD16A) efficiently recruits NK cells forthe lysis of CD30+ tumor cells. MAbs 2014; 6: 728–39.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

Antibody Therapeutics, 2020 61

114. Bartlett, NL, Chen, RW, Domingo-Domenech, E et al. A phase 1bstudy investigating the combination of the tetravalent Bispecific NKcell engager AFM13 and pembrolizumab in patients withrelapsed/refractory Hodgkin lymphoma after brentuximab vedotinfailure: updated safety and efficacy data. Blood 2018; 132: 1620.

115. Sawas, A, Chen, P, Vlad, G et al. Clinical and biological evaluationof the novel Cd30/Cd16a tetravalent bispecific antibody (Afm13) inrelapsed or refractory Cd30-positive lymphoma with cutaneouspresentation: a biomarker phase Ib/Iia study (Nct03192202).Hematol Oncol 2019; 37: 314–6.

116. Chan, WK, Kang, S, Youssef, Y et al. A CS1-NKG2D bispecificantibody collectively activates cytolytic immune cells againstmultiple myeloma. Cancer Immunol Res 2018; 6: 776–87.

117. Corgnac, S, Perret, R, Derré, L et al. CD1d-antibody fusionproteins target iNKT cells to the tumor and trigger long-termtherapeutic responses. Cancer Immunol Immunother 2013; 62:747–60.

118. Van Der Horst, HJ, Oostindie, SC, Cillessen, SAGM et al.Targeting CD37 in B-cell malignancies using the novel therapeuticDuoHexaBody-CD37 results in efficient killing of tumor B-cells exvivo via complement-dependent cytotoxicity, even in relapsedand/or refractory patient samples. Blood 2018; 132: 4179.

119. Advani, R, Flinn, I, Popplewell, L et al. CD47 blockade byHu5F9-G4 and rituximab in non-Hodgkin’s lymphoma. N Engl JMed 2018; 379: 1711–21.

120. Hatterer, E, Barba, L, Noraz, N et al. Co-engaging CD47 andCD19 with a bispecific antibody abrogates B-cell receptor/CD19association leading to impaired B-cell proliferation. MAbs 2019; 11:322–34.

121. Garcia-Martinez, JM, Wernitznig, A, Rinnenthal, J et al. Abstract2051: BI 905711, a novel CDH17-targeting TRAILR2 agonist,effectively triggers tumor cell apoptosis and tumor regressionsselectively in CDH17-positive colorectal cancer models. Cancer Res2019; 79: 2051–1.

122. Pandey, P, Sliker, B, Peters, HL et al. Amyloid precursor proteinand amyloid precursor-like protein 2 in cancer. Oncotarget 2016; 7:19430–44.

123. Bay, AP, Kalsy, A, Tiwari, S et al. Abstract 233: bispecific HER2ADC: making more potent HER2 ADC by improving targetinternalization. Cancer Res 2019; 79: 233–3.

124. Ravetch, JV, Lanier, LL. Immune inhibitory receptors. Science(80-) 2000; 290: 84–9.

125. Chen, W, Shankar, S, Lohr, J et al. SAT0027 Immunomodulatory

effects of MGD010, a dart®

molecule targeting human B-CELLCD32B and CD79B. Ann Rheum Dis 2017; 76: 777–8.

126. Knight, T, Callaghan, MU. The role of emicizumab, a bispecificfactor IXa- and factor X-directed antibody, for the prevention ofbleeding episodes in patients with hemophilia A. Ther Adv Hematol2018; 9: 319–34.

127. Kitazawa, T, Igawa, T, Sampei, Z et al. A bispecific antibody tofactors IXa and X restores factor VIII hemostatic activity in ahemophilia A model. Nat Med 2012; 18: 1570–4.

128. Sampei, Z, Igawa, T, Soeda, T et al. Identification andmultidimensional optimization of an asymmetric bispecific IgGantibody mimicking the function of factor VIII cofactor activity.PLoS One 2013; 8: e57479.

129. Oldenburg, J, Mahlangu, JN, Kim, B et al. Emicizumab prophylaxisin hemophilia A with inhibitors. N Engl J Med 2017; 377: 809–18.

130. Sahni, J, Patel, SS, Dugel, PU et al. Simultaneous inhibition ofangiopoietin-2 and vascular endothelial growth factor-a withFaricimab in diabetic macular Edema BOULEVARD phase 2randomized trial. Ophthalmology 2019; 126: 1155–70.

131. Pulgar, VM. Transcytosis to cross the blood brain barrier. NewAdvancements and Challenges. Front Neurosci 2019; 12: 1019.

132. DiGiandomenico, A, Keller, AE, Gao, C et al. A multifunctionalbispecific antibody protects against Pseudomonas aeruginosa. SciTransl Med. 2014; 6: 262ra155.

133. Takada, A. Filovirus tropism: cellular molecules for viral entry.Front Microbiol 2012; 3: 1–9.

134. Holtsberg, W, Bakken, RR, Mittler, E et al. A “Trojan horse”bispecific-antibody strategy for broad protection againstebolaviruses. Science 2017; 354: 350–4.

135. Kolumam, G, Chen, MZ, Tong, R et al. Sustained brown fatstimulation and insulin sensitization by a humanized bispecific

antibody agonist for fibroblast growth factor receptor 1/βKlothocomplex. EBioMedicine 2015; 2: 730–43.

136. Chodorge, M, Züger, S, Stirnimann, C et al. A series of Fasreceptor agonist antibodies that demonstrate an inverse correlationbetween affinity and potency. Cell Death Differ 2012; 19: 1187–95.

137. Liu, Z, Stoll, VS, DeVries, PJ et al. A potenterythropoietin-mimicking human antibody interacts through anovel binding site. Blood 2007; 110: 2408–13.

138. Hothersall, JD, Guo, D, Sarda, S et al. Structure-activityrelationships of the sustained effects of adenosine A2A receptoragonists driven by slow dissociation kinetics. Mol Pharmacol 2017;91: 25–38.

139. Leong, SR, Sukumaran, S, Hristopoulos, M et al. Ananti-CD3/anti-CLL-1 bispecific antibody for the treatment of acutemyeloid leukemia. Blood 2017; 129: 609–18.

140. Zuch de Zafra, CL, Fajardo, F, Zhong, W et al. Targeting multiplemyeloma with AMG 424, a novel anti-CD38/CD3 bispecificT-cell–recruiting antibody optimized for cytotoxicity and cytokinerelease. Clin Cancer Res 2019; 25: 3921–33.

141. Hoffmann, P, Hofmeister, R, Brischwein, K et al. Serial killing oftumor cells by cytotoxic T cells redirected with aCD19-/CD3-bispecific single-chain antibody construct. Int JCancer 2005; 115: 98–104.

142. Yu, YJ, Zhang, Y, Kenrick, M et al. Boosting brain uptake of atherapeutic antibody by reducing its affinity for a transcytosistarget. Sci Transl Med 2011; 3: 84ra44.

143. Tillotson, BJ, Goulatis, LI, Parenti, I et al. Engineering ananti-transferrin receptor ScFv for pH-sensitive binding leads toincreased intracellular accumulation. PLoS One 2015; 10:e0145820–0.

144. Bardwell, PD, Staron, MM, Liu, J et al. Potent and conditionalredirected T cell killing of tumor cells using half DVD-Ig. ProteinCell 2018; 9: 121–9.

145. Hernandez-Hoyos, G, Sewell, T, Bader, R et al. MOR209/ES414, anovel bispecific antibody targeting PSMA for the treatment ofmetastatic castration-resistant prostate cancer. Mol Cancer Ther2016; 15: 2155–65.

146. Reusch, U, Duell, J, Ellwanger, K et al. A tetravalent bispecificTandAb (CD19/CD3), AFM11, efficiently recruits T cells for thepotent lysis of CD19(+) tumor cells. MAbs 2015; 7: 584–604.

147. Wang, J, Goetsch, L, Tucker, L et al. Anti-c-Met monoclonalantibody ABT-700 breaks oncogene addiction in tumors with METamplification. BMC Cancer 2016; 16: 105.

148. Trinklein, ND, Pham, D, Schellenberger, U et al. Efficient tumorkilling and minimal cytokine release with novel T-cell agonistbispecific antibodies. MAbs 2019; 11: 639–52.

149. Bluemel, C, Hausmann, S, Fluhr, P et al. Epitope distance to thetarget cell membrane and antigen size determine the potency of Tcell-mediated lysis by BiTE antibodies specific for a largemelanoma surface antigen. Cancer Immunol Immunother 2010; 59:1197–209.

150. Li, J, Stagg, NJ, Johnston, J et al. Membrane-proximal epitopefacilitates efficient T cell synapse formation by anti-FcRH5/CD3and is a requirement for myeloma cell killing. Cancer Cell 2017; 31:383–95.

151. Crawford, A, Haber, L, Kelly, MP et al. A Mucin 16 bispecific Tcell-engaging antibody for the treatment of ovarian cancer. SciTransl Med 2019; 11: eaau7534.

152. Yokosuka, T, Takamatsu, M, Kobayashi-Imanishi, W et al.Programmed cell death 1 forms negative costimulatorymicroclusters that directly inhibit T cell receptor signaling byrecruiting phosphatase SHP2. J Exp Med 2012; 209:1201–17.

153. Le Gall, F, Reusch, U, Little, M et al. Effect of linker sequencesbetween the antibody variable domains on the formation, stabilityand biological activity of a bispecific tandem diabody. Protein EngDes Sel 2004; 17: 357–66.

154. DiGiammarino, EL, Harlan, JE, Walter, KA et al. Ligandassociation rates to the inner-variable-domain of adual-variable-domain immunoglobulin are significantly impactedby linker design. MAbs 2011; 3: 487–94.

155. Thurber, GM, Schmidt, MM, Wittrup, KD. Antibody tumorpenetration: transport opposed by systemic and antigen-mediatedclearance. Adv Drug Deliv Rev 2008; 60: 1421–34.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022

62 Antibody Therapeutics, 2020

156. Schmidt, MM, Wittrup, KD. A modeling analysis of the effects ofmolecular size and binding affinity on tumor targeting. Mol CancerTher 2009; 8: 2861–71.

157. Goldenberg, DM, Chatal, J-F, Barbet, J et al. Cancer imaging andtherapy with bispecific antibody pretargeting. Update Cancer Ther2007; 2: 19–31.

158. Roux, KH, Strelets, L, Michaelsen, TE. Flexibility of human IgGsubclasses. J Immunol 1997; 159: 3372–82.

159. Kapelski, S, Cleiren, E, Attar, RM et al. Influence of the bispecificantibody IgG subclass on T cell redirection. MAbs 2019; 11:1012–24.

160. Sampei, Z, Igawa, T, Soeda, T et al. Non-antigen-contacting regionof an asymmetric bispecific antibody to factors IXa/X significantlyaffects factor VIII-mimetic activity. MAbs 2015; 7: 120–8.

161. Zhang, T, Song, X, Xu, L et al. The binding of an anti-PD-1antibody to Fcγ RI has a profound impact on its biologicalfunctions. Cancer Immunol Immunother 2018; 67: 1079–90.

162. Singh, SK. Impact of product-related factors on immunogenicity ofbiotherapeutics. J Pharm Sci 2011; 100: 354–87.

163. Davda, J, Declerck, P, Hu-Lieskovan, S et al. Immunogenicity ofimmunomodulatory, antibody-based, oncology therapeutics. JImmunother Cancer 2019; 7: 105.

164. Groell, F, Jordan, O, Borchard, G. In vitro models forimmunogenicity prediction of therapeutic proteins. Eur J PharmBiopharm 2018; 130: 128–42.

165. Saunders, KO. Conceptual approaches to modulating antibodyeffector functions and circulation half-life. Front Immunol 2019; 10:1296.

166. Gaudinski, MR, Coates, EE, Houser, KV et al. Safety andpharmacokinetics of the Fc-modified HIV-1 human monoclonalantibody VRC01LS: a phase 1 open-label clinical trial in healthyadults. PLoS Med 2018; 15: e1002493.

167. Leipold, D, Prabhu, S. Pharmacokinetic and pharmacodynamicconsiderations in the design of therapeutic antibodies. Clin TranslSci 2019; 12: 130–9.

168. Adams, GP, Schier, R, Mccall, AM et al. High affinity restricts thelocalization and tumor penetration of single-chain Fv antibodymolecules. Cancer Res 2001; 61: 4750–5.

169. Rudnick, SI, Lou, J, Shaller, CC et al. Influence of affinity andantigen internalization on the uptake and penetration of anti-HER2antibodies in solid tumors. Cancer Res 2011; 71: 2250–9.

170. Friedrich, SW, Lin, SC, Stoll, BR et al. Antibody-directed effectorcell therapy of tumors: analysis and optimization using aphysiologically based pharmacokinetic model. Neoplasia 2002; 4:449–63.

171. Goebeler, M-E, Knop, S, Viardot, A et al. Bispecific T-cell engager(BiTE) antibody construct blinatumomab for the treatment ofpatients with relapsed/refractory non-Hodgkin lymphoma: finalresults from a phase I study. J Clin Oncol 2016; 34:1104–11.

172. Gunde, T, Brock, M, Warmuth, S et al. Abstract 1532: a novel,monovalent tri-specific antibody-based molecule thatsimultaneously modulates PD-L1 and 4-1BB exhibits potentanti-tumoral activity in vivo. Cancer Res 2019; 79: 1532–2.

173. Prell, RA, Lee, DW, Halpern, WG et al. Chapter 14. Nonclinicaldevelopment of multi-targeting biopharmaceuticals. In: Plitnick,LM, Biosimilars, H (eds). Vaccines and Specialty BiologicsDJBT-ND of NB. San Diego: Academic Press, 2013, 343–71

174. Trivedi, A, Stienen, S, Zhu, M et al. Clinical pharmacology andtranslational aspects of bispecific antibodies. Clin Transl Sci 2017;10: 147–62.

175. Mayes, PA, Hance, KW, Hoos, A. The promise and challenges ofimmune agonist antibody development in cancer. Nat Rev DrugDiscov 2018; 17: 509–27.

176. Messenheimer, DJ, Jensen, SM, Afentoulis, ME et al. Timing ofPD-1 blockade is critical to effective combination immunotherapywith anti-OX40. Clin Cancer Res 2017; 23: 6165–77.

177. Kohrt, HE, Houot, R, Goldstein, MJ et al. CD137 stimulationenhances the antilymphoma activity of anti-CD20 antibodies.Blood 2011; 117: 2423–32.

178. Kohrt, HE, Houot, R, Weiskopf, K et al. Stimulation of naturalkiller cells with a 4-1BB-specific antibody enhances trastuzumabefficacy in xenotransplant models of breast cancer. J Clin Invest2012; 122: 1066–75.

179. Jackman, J, Chen, Y, Huang, A et al. Development of a two-partstrategy to identify a therapeutic human bispecific antibody thatinhibits IgE receptor signaling. J Biol Chem 2010; 285: 20850–9.

180. Gu, J, Yang, J, Chang, Q et al. Identification of anti-ErbB2 dualvariable domain immunoglobulin (DVD-IgTM) proteins withunique activities. PLoS One 2014; 9: e97292–2.

181. Madrenas, J, Chau, LA, Teft, WA et al. Conversion of CTLA-4from inhibitor to activator of T cells with a bispecific tandemsingle-chain Fv ligand. J Immunol 2004; 172: 5948–56.

Dow

nloaded from https://academ

ic.oup.com/abt/article/3/1/18/5739255 by guest on 30 M

ay 2022