Download as pdf or txt
Download as pdf or txt
You are on page 1of 157

Immobilized Enzymes and

their application in Medicine.


Dr. Gul Shahnaz
Immobilization of enzymes
Immobilization of enzymes (or cells) refers to the technique of confining/anchoring the enzymes (or cells) in or
on an inert support for their stability and functional reuse.
Immobilized enzymes possess higher resistance to environmental changes and can be recovered/recycled easily when compared to the
free forms.
Problems and Limitations Associated with Industrial Enzymes
Enzyme immobilization methods
 Immobilization strategies entail fixing or entrapping enzymes within solid support materials.
Researchers have suggested several support materials, besides many beneficial approaches for the
immobilization of enzymes.

 The main function of the support is to stabilize the structures of enzymes and accordingly preserve their
efficacy to a great extent by rendering them more resistant to the surrounding environments.

 Enzyme immobilization permits an easy recovery of both the used enzymes and their support materials,
and this is particularly beneficial in the food, medical, and pharmaceutical applications.

 Enzymes in their immobilized forms possess much higher stability and are also easier to handle when
compared to their free forms.

 Additionally, the enzymatic reaction can occur in a nonaqueous medium where the solid supports
preserve the enzyme’s constituents and make them stronger, which enhances their catalytic activity and
renders them reusable for several times.

 Another benefit of the immobilization process is that the catalysts can alter from homogeneous to
heterogeneous forms after the enzymatic binding, which assists in separating the enzymes, producing
products with high purity
Adsorption
• The enzyme here are adhere to carrier matrix due to hydrophobic
effects or by forming salt links.
• Binding is strong but may be weakened by substrate, pH etc.

• Commonly used matrices :-


ion exchange matrices, porous carbon,clays, hydrous metal oxides, polymeric
aromatic resins etc.
Covalent binding
• The enzyme here are attached to certain matrix by forming covalent
bonds
• Binding is very strong. It generally occurs with side chain of the
enzyme. Lysine is very useful, as they are very reactive, usually
exposed to surface and very rarely occur at active site of enzyme.
• Commonly used matrices :-
agarose, cellulose, poly acrylamides etc.
• Activation by CNBr
Cont….
• Activation by Ethyl chloroformate
Cont….
• Immobilization of enzyme using Glutaraldehyde
Methods for immobilization

c) Entrapment d) Membrane confinement


Entrapment

• The enzyme here are entrapped with in suitable gels or fibers,


with or with out covalent bonds.

• Commonly used matrices :-


Cellulose acetate fibers, calcium alginate etc.
Membrane confinement

• The enzyme molecule here are confined to semipermiable


membrane. Thus in a aqueous phase beg of membrane is free to
move with out allowing enzyme to come out.
Advantages
• Enzymes are costly, Immobilization permits their repeated use.
• The product can be easily separated without any additional cost.
• Immobilized enzymes can be used in nonaqueous system.
• Continuous production system can be used.
• Thermo stability of some enzyme can be increased by immobilization.
• Enzyme can be used at much higher concentration then free enzyme.
Disadvantages

• Additional cost.
• It sometime affects stability and activity of enzyme.
• This approach can not be used when one of the substrates is
insoluble.
• Some immobilization methods restricts the diffusion of substrate.
Applications of immobilized enzymes

Enzyme Substrate Product


Glucose isomerase D-glucose HFCS
Invertase Sucrose Invert sugar
Lipase Vegetable oil Cocoa butter substitute
Lactase Milk and whey Lactose free milk &whey
Nitrile hydratase Acrylonitrile Acrylamide
Raffinase Raffinose Raffinose free solution
Glucoamylase Dextrins D-glucose
Genetic Engineering and its
Application in Medicine

Dr. Gul Shahnaz


What is genetic engineering?

Genetic engineering is the direct modification of an


organism’s genome, which is the list of specific traits
(genes) stored in the DNA.
Changing the genome
enables engineers to give
desirable properties to
different organisms.
Organisms created by
genetic engineering
are called genetically modified organisms (GMOs).
History of GMO Development
1973: created first genetically
modified bacteria
1974: created GM mice
1982: first commercial
development of GMOs
(insulin-producing bacteria)
1994: began to sell
genetically modified food
2003: began to sell GMOs as
pets (Glofish)
What is the GMO process?
• All genetic changes affect the protein synthesis of the
organism.
• By changing which proteins are produced, genetic
engineers can affect the overall traits of the organism.
• Genetic modification can be completed by a number of
different methods:
• Inserting new genetic
material randomly or in
targeted locations
• Direct replacement of
genes (recombination)
• Removal of genes
• Mutation of existing genes
Gene Therapy
How is it done?
Growth hormone and therapy
• Growth hormone, also known as
somatotropin, is a protein hormone of about
190 amino acids that is synthesized and
secreted by cells called somatotrophs in the
anterior pituitary. It is a major participant in
control of several complex physiologic
processes, including growth and metabolism.
Growth hormone is also of considerable interest
as a drug used in both humans and animals.
Recombinant Protein
(Insulin)
Production of recombinant insulin

Attemps to produce insulin by recombinant DNA technology started in late 1970s.

The basic technique consisted of inserting human insulin gene and the promoter gene of
lac operon on to the plasmids of E. coli.

By this method human insulin was produced.

It was in July 1980, seventeen human volunteers were, for the first time, administered recombinant insulin for
treatment of diabetes at Guy's Hospital, London. And in fact, insulin was the first ever pharmaceutical product
of recombinant DNA technology administered to humans.

Recombinant insulin worked well, and this gave hope to scientists that DNA technology could be successfully
employed to produce substances of medical and commercial importance. An approval, by the concerned
authorities, for using recombinant insulin for the treatment of diabetes mellitus was given in 1982.

And in 1986, Eli Lilly company received approval to market human insulin under the trade name Humulin.
Production of recombinant insulin

Isolation of plasmid and Human insulin insert the human insulin gene Formation of r-DNA
gene into the plasmid.

Put the “recombinant” bacteria Return the plasmid to the bacteria


Harvest and purify the Recombinant bacteria use in large fermentation tanks (Transformation)
substance for use as a the gene to begin
medicine. producing human insulin.
Production of recombinant insulin

The orginal technique of insulin synthesis in E. coli has undergone


several changes, for improving the yield. e.g. addition of signal
peptide, synthesis of A and B chains separately etc.

The procedure employed for the synthesis of two insulin chains A


and B is illustrated in Fig.

The genes for insulin A chain and B chain are separately inserted to
the plasmids of two different E. coli cultures. The lac operon system
(consisting of inducer gene, promoter gene, operator gene and
structural gene Z for β-galactosidase) is used to express both the
genes.

The presence of lactose in the culture medium induces the synthesis


of insulin A and B chains in separate cultures.

The so formed insulin chains can be isolated, purified and joined


together to give a full-fledged human insulin.
Source: Adopted from Biochemistry (U. Satyanarayna)
What is Protein therapeutics?

It is currently estimated that there are 25,000–40,000 different


genes in the human genome, viewed from the perspective of
disease mechanisms, as disease may result
when any one of these proteins contains
mutations or other abnormalities, so it
gives a tremendous opportunity for
Protein therapeutics to alleviate
these disease.
Why protein therapeutics?

Proteins cannot be mimicked by simple chemical compounds.

There is often less potential for protein therapeutics to interfere

with normal biological processes and cause adverse effects.

It is often well tolerated and are less likely to elicit immune responses.

Provide effective replacement treatment without the need for gene therapy

Time of protein therapeutics may be faster


History and Development

2002 and beyond BIOTECHNOLOGY IMPROVEMENT

1992–1999 BIOTECHNOLOGY INDUSTRY

1986–1991 MORE BIOTECHNOLOGY SUCCESSES

Pre-1986 A STAR IS BORN


The Evolution of Protein Therapeutics : A Timeline

1953 First accurate model of DNA suggested

1982 Human insulin, created using recombinant DNA technology


1986 Interferon alfa and muromonab-CD3 approved

1993 CBER's Office of Therapeutics Research and Review (OTRR) formed


1997 First whole chimeric antibody, rituximab, and first humanized
antibody, daclizumab, approved

2002 Market for biotechnology products represents approximately $30


billion of $400 billion in yearly worldwide pharmaceutical sales

2006 An inhaled form of insulin (Exubera) approved, expanding protein


products into a new dosage form.
THE IMMUNE SYSTEM

The Latin term “IMMUNIS” means EXEMPT, referring to


protection against foreign agents.

DEFINITION: - The integrated body system of organs, tissues,


cells & cell products that differentiates self from non – self &
neutralizes potentially pathogenic organisms.
(The American Heritage Stedman's Medical Dictionary)

The Immune System consists of


1. Innate Immunity Primary Response
2. Acquired Immunity Secondary Response
CELLS OF THE IMMUNE SYSTEM
FUNCTIONING OF THE IMMUNE SYSTEM
HUMORAL (ANTIBODY MEDIATED) IMMUNE RESPONSE CELL MEDIATED IMMUNE RESPONSE
ANTIGEN (1ST EXPOSURE)
ENGULFED BY

MACROPHAGE ANTIGENS
FREE
ANTIGENS DISPLAYED
BECOMES BY
DIRECTLY
INFECTED
ACTIVATE APC CELLS
ACTIVATE
STIMULATES

HELPER CYTOTOXIC
B CELLS T CELLS
STIMULATES STIMULATES T CELL

MEMORY
GIVES RISE TO HELPER T GIVES RISE TO
CELLS
STIMULATES STIMULATES
STIMULATES

ANTIGEN (2nd EXPOSURE)


PLASMA MEMORY STIMULATES ACTIVE
MEMORY
CELLS B CELLS T CELLS CYTOTOXIC T
CELL

SECRETE ANTIBODIES
IMMUNOTHERAPY

Treatment of the disease by Inducing, Enhancing or


Suppressing the Immune System.

Active Immunotherapy: - Passive Immunotherapy: -


It stimulates the body’s own It does not rely on the body to
immune system to fight the attack the disease, instead they
disease. use the immune system
components ( such as
antibodies) created outside the
body.
What are antibodies
An antibody is a protein used by the immune system to identify and neutralize foreign objects
like bacteria and viruses. Each antibody recognizes a specific antigen unique to its target.

Monoclonal antibodies (mAb) are antibodies that are identical because they were produced
by one type of immune cell, all clones of a single parent cell.

Polyclonal antibodies are antibodies that are derived from different cell lines.

Isotypes
According to differences in their heavy chain constant domains, immunoglobulins are
grouped into five classes, or isotypes: IgG, IgA, IgM, IgD, and IgE.

IgG: IgG1 (66%), IgG2 (23%), IgG3 (7%) and IgG4 (4%) , blood and tissue liquid.
IgA:IgA1 (90%) and IgA2 (10%), stomach and intestines
IgM: normally pentamer, ocassionally hexamer, multiple immunoglobins linked with disulfide
bonds
IgD:1% of proteins in the plasma membranes of B-lymphocytes, function unknown
IgE: on the surface of plasma membrane of mast cells, play a role in immediate hypersensitive
and denfensive for parasite
How do monoclonal antibody drugs work

1- Make the cancer cell more visible to the immune


system.
The immune system attacks foreign invaders in your body, but it doesn't
always recognize cancer cells as enemies. A monoclonal antibody can be
directed to attach to certain parts of a cancer cell. In this way, the antibody
marks the cancer cell and makes it easier for the immune system to find.

The monoclonal antibody drug Rituximab attaches to a specific protein (CD20)


found only on B cells, one type of white blood cell. Certain types of lymphomas
arise from these same B cells. When Rituximab attaches to this protein on the
B cells, it makes the cells more visible to the immune system, which can then
attack. Rituximab lowers the number of B cells, including your healthy B cells,
but your body produces new healthy B cells to replace these. The cancerous B
cells are less likely to recur.
RITUXIMAB (Rituxan)

• A recombinant chimeric murine/human antibody directed against the


CD20 antigen, a hydrophobic transmembrane protein located on normal
pre-B and mature B lymphocytes.

• Following binding, rituximab triggers a host cytotoxic immune response


against CD20-positive cells.

• Rituximab is used in the treatment of B-cell Non-


Hodgkin's lymphomas.
• MECHANISM OF ACTION:

• Complement fixation
• ADCC
• Direct cytotoxicity via apoptotic pathway activated by binding to CD20.
FDA-APPROVED MONOCLONAL ANTIBODIES TARGETS and INDICATIONS
Antibody Trade Name Target Antigen FDA Indication

Rituximab Rituxan CD20 B-NHL

90Y-Ibritumomab Zevalin CD20 B-NHL

131I-tositumomab Bexxar CD20 B-NHL

Alemtuzumab Campath CD52 CLL

Gemtuzumab Mylotarg CD33 AML


ozogamicin
Cetuximab Erbitux EGFR metastatic
CRC
Panitumumab ABX-EGF EGFR metastatic
CRC
Trastuzumab Herceptin HER2/neu Metastatic
Br CA
Production of Recombinant Vaccine
Production of recombinant Vaccine

The recombinant vaccines are an important group of therapeutic products.

A number of vaccines are now available for animals, and human which is going to have a major impact in the
healthcare industry.

One of the initial vaccines produced by rDNA method involves the cloning of the surface antigen of the
hepatitis B virus (HBsAg) in the yeast S. cerevisiae under the control of the alcohol dehydrogenase promoter.

A number of recombinant vaccines are now commercially prepared by the recombinant DNA technology,
where only the outside coat protein of the microorganism is expressed in the host to create the vaccine.

The expressed protein can then be purified from the recombinant host and used for inoculation.

This method has the advantage of safe delivery of antigen without transferring the actual disease-causing
microbe to the host. Currently recombinant vaccines for the hepatitis B virus, herpes type 2 viruses, and
malaria is under trial for use in future.
Table: list of diseases along with
the pathogenic organisms for
which recombinant vaccines are
developed

Source: Adopted from Biochemistry (U. Satyanarayna)


Pharmaceutical
Biotechnology
Dr. Gul Shahnaz
What Is Biotechnology and What Does It Mean to You?

• Biotechnology – using living organisms, or the products


of living organisms, for human benefit to make a product
or solve a problem

• Historical Examples
– Fermentation
– Selective breeding
– Use of antibiotics
Why are new drugs needed?

 unmet medical need; new diseases (Corona Virus, Swine flu; AIDS,
Alzheimer’s; obesity); low efficacy (dementia, cancer); side effects
(antidepressants, antipsychotics)
 downstream health costs; (Alzheimer’s; spinal injury)
 cost of therapy; (Interleukins)
 sustain industrial activity; pharmaceutical industry employs
thousands and makes a massive contribution to overseas earnings);
patent expiry
 Drug resistance:
The Long Road to a New Medicine

* 10-15 Year Process


Registration
New
Full Clinical
Data Medicine
Development Analysis

Candidate Medicine Tested in


Studies in 100-300 3-10,000 Patients (Phase III)
Patients – POC
(Phase II) Large Amounts of
Candidate Medicine
Synthesized Extensive
Safety
Studies
Studies in Healthy
Volunteers – safety Candidate
and POM (Phase I) Formulations
Exploratory Developed

Development

Early
Safety
Project Team Synthesis of Studies
and Plans Screening
Compounds

Discovery
Idea
Involves high cost and time

~100 Discovery Approaches

7,000,000
Compounds Screened

Preclinical
Pharmacology

Preclinical Safety
1-2
Clinical Pharmacology
& Safety
Products

Discovery Exploratory Development Full Development


Phase I Phase II Phase III

0 5 10 15

Idea 10 - 15 Years Drug


Structure Based Drug
Design

Determine Protein Structure

Identify Interaction Sites


Discovery or design of De Novo Design 3D Database
molecules that interact
with biochemical targets Evaluate Structure
of known 3D structure
Synthesize Candidate
Test Candidate

Lead Compound
Drug Targets
Molecule or structure within the organism linked to a particular disease, whose activity can be
modified by a drug.
Currently used drug targets

J. Drews Science 287, 1960 -1964


(2000)
What Is Biotechnology and What Does It
Mean to You?
• Example of Biotechnology – Selective Breeding
(a) (b)

Normal zebrafish "Casper" zebrafish – made


by selective breeding
• What feature of Casper makes it a "model organism" to study migration
of cancer cells compared to wildtype fish?
What Is Biotechnology and What Does It
Mean to You?
• Based on this tree,
can you become
successful in the
biotech industry
only studying
biology?
What Is Biotechnology and What Does It Mean to You?

• Modern Examples
– Gene cloning
– Genetic engineering
– Recombinant DNA technology
– Human Genome Project
1.1 What Is Biotechnology and What Does It
Mean to You?
• Example of "modern" biotechnology:
– recombinant DNA technology started modern biotech as an industry
• Examples of applications
– development of disease-resistant plants
– food crops that produce greater yields
– "golden rice" engineered to be more nutritious
– genetically engineered bacteria that can degrade environmental pollutants
• Work in groups to come up with more examples of
applications
1.1 What Is Biotechnology and What Does It
Mean to You?
• Use genetically modified cultured cells to make protein of
interest
• Products of Modern Biotechnology
– Example of proteins created by gene cloning called
recombinant proteins
Functional Genomics and Proteomics
Proteomics studies biological systems based
on global knowledge of protein sets
(proteomes).
Functional genomics studies biological
functions of proteins, complexes, pathways
based on the analysis of genome sequences.
Includes functional assignments for protein
sequences.
Genome Transcriptome Proteome Metabolome
“-ome”
CGTCCAA
CTGACGT
DNA Genome “Genomics”
CTACAAG
TTCCTAA DNA sequencing
GCT

Transcriptome
RNA
cDNA arrays
Cell
functions

Proteins Proteome “Proteomics”


2D PAGE, HPLC

Reactome, the chemical reactions involving a nucleotide


Protein Chemistry/Proteomics

Protein Chemistry Proteomics


• Individual proteins • Complex mixtures
• Complete sequence analysis • Partial sequence analysis
• Emphasis on structure and • Emphasis in identification by
function database matching
• Structural biology • System biology
Why are we studying proteins?

Proteins are the mediators of functions in the cell

Deviations from normal status denotes disease

Proteins are drug/therapeutic targets


Proteomics and biology
/Applications
Protein Expression Profiling
Identification of proteins in a particular
Proteome Mining sample as a function of a particular
Identifying as many as state of the organism or cell
possible of the proteins in Post-translational
your sample modifications
Identifying how and
where the proteins are
modified
Functional
proteomics

Protein-protein
interactions Protein-
Protein quantitation network mapping
Determining how the
or differential proteins interact with
Structural
analysis each other in living
Proteomics systems
Tools of Proteomics

Protein separation technology


Simplify complex protein mixtures
Target specific proteins for analysis

Mass spectrometry (MS)


Provide accurate molecular mass measurements
of intact proteins and peptides

Database
Protein, EST, and complete genome sequence
databases

Software collection
Match the MS data with specific protein
sequences in databases
The Proteome

The proteome in any cell represents a subset of all possible gene


products
Not all the genes are expressed in all the cells.
It will vary in different cells and tissue types in the same organism and
between different growth and developmental stages
The proteome is dependent on environmental factors, disease, drugs,
stress, growth conditions.

• Cycle of Proteins
• Proteins as Modular Structures – motifs, domains
• Functional Families
• Genomic Sequences
• Protein Expression /Protein level
Life cycle of a protein
Information found in DNA is used for
synthesis of the proteins

mRNA Protein Folding Translocation


to specific subcellular or
extracellular compartments

Posttranslational Processing
Proteolytic Cleaveage
Degradation Acylation
Methylation
Damage
-free radicals Phosphorylation
Sulfation
Selenoproteins
Environmental Ubiquination
-chemicals Glycolisation
radioactiivty
Molecular Structures
Primary structure a chain of amino acids Amino acids vary in their ability
to form the various secondary
Secondary structure three dimensional form, formally structure elements.
defined by the hydrogen bonds of the polymer

Amino acids that prefer to adopt helical conformations in


-helices proteins include methionine, alanine, leucine, glutamate
and lysine ("MALEK" in amino acid 1-letter codes)

The large aromatic residues (tryptophan, tyrosine and


-sheets phenylalanine) and Cβ-branched amino acids (isoleucine,
valine and threonine) prefer to adopt -strand
conformations.

Confer similar properties or functions when


they occur in a variety of proteins
Sequence alignment

Sequence alignment is a way of arranging primary sequences (of


DNA, RNA, or proteins) in such a way as to align areas sharing
common properties.

The degree of relatedness, similarity between the A software tool used for general
sequences is predicted computationally or sequences alignment tasks is
statistically ClustalW
General workflow of proteomics
analysis
separation
proteins digestion
MALDI, MS/MS

digestion

Identification
peptides
(LC)-MS/MS

ESI-MS
Electrospray Ionization tandem MS

MALDI-TOF
Matrix Assisted Laser Desorption
Ionization –Time of Flight
Transcriptomes
• Genome: all of hereditary information encoded in
the DNA (or RNA)
• Transcriptome: set of all mRNAs ("transcripts”)
produced from a genome
• Term can be applied to:
complete set of transcripts for a given organism
specific subset of transcripts present in a
particular cell type or under specific growth
conditions
• Transcriptome varies because it reflects genes that
are actively expressed at any given time
Genomics, ‘Omics & Technology
• Molecular biology: major scientific discipline for
past ~50 years
• Genomics = “analysis of genomes”: became
important science during 1990’s
• Analyses of various other biological molecules have
developed into their own scientific disciplines; e.g.
Metabolomics = “analysis of metabolites”, etc.
• Transcriptomics/Proteomics: developed during past
10-15 years
• Bioinformatics: has developed as major branch of
science - enables efficient analysis of data from
“omics” experiments
Genomics & Technology
• Significance of “omics” coincides with dramatic
improvements in different technologies:
molecular biology: increased range of
approaches for purification and manipulation of
proteins and nucleic acids
computers: required for gathering and analysis
of data
internet: allows data to be shared, quickly and
easily
• All developments have increased speed and cost-
effectiveness - available to much wider audience
Transcriptomics

• Transcriptomics uses high-


throughput techniques
based on DNA microarrays
• For further details about
microarrays see Lucchini et
al., Microbiology, 147, 1403-
1414 (2001)

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328
Transcriptomics

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328

SYLICA 'Omic Technologies – Bowater Feb 2013


Transcriptomics

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328

SYLICA 'Omic Technologies – Bowater Feb 2013


Transcriptomics

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328

SYLICA 'Omic Technologies – Bowater Feb 2013


Transcriptomics

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328
Transcriptomics

Nelson & Cox, “Lehninger, Principles of


Biochemistry”, 4th edn, 2004, p. 328
Transcriptomics
•Experiments performed
under different
conditions
•Determines effect of
conditions on
expression

•Produces huge amount


of data
•Lots of repeats required
- expensive
Nelson & Cox, “Lehninger, Principles of
Biochemistry”, 4th edn, 2004, p. 328
Polymerase Chain Reaction (PCR)
• Used to amplify DNA in the test tube
– Can amplify regions of interest (genes) within DNA
– Can amplify complete circular plasmids
• Mix together
– Target DNA
– Primers (oligonucleotides complementary to target)
– Nucleotides: dATP, dCTP, dGTP, dTTP
– Thermostable DNA polymerase
• Place the mixture into thermocycler
– Melt DNA at ~95°C
– Cool to ~ 50–60°C, primers anneal to target
– Polymerase extends primers in 5’3’ direction
– After a round of elongation is done, repeat steps
General Steps of PCR

SYLICA 'Omic Technologies – Bowater Feb 2013


General Steps of PCR
•Repeat steps 1–3 many times:
Photolitographic Synthesis of DNA
DNA Microarrays: Applications

DNA microarrays allow simultaneous screening of


many thousands of genes: high-throughput screening
• Genome-wide genotyping
– Which genes are present in this individual?
• Tissue-specific gene expression
– Which genes are used to make proteins?
• Mutational analysis
– Which genes have been mutated?

SYLICA 'Omic Technologies – Bowater Feb 2013


Adaptations to PCR
• Reverse Transcriptase PCR (RT-PCR)
– Used to amplify RNA sequences
– First step uses reverse transcriptase to convert
RNA to DNA
• Quantitative PCR (Q-PCR)
– Used to show quantitative differences in gene
levels
qPCR
Proteomes
• Proteome: set of all proteins produced under a given
set of conditions
• Term can be applied to:
complete set of proteins for a given organism
specific subset of proteins present in a particular
cell type or under specific growth conditions
• Proteome varies because it reflects genes that are
actively expressed at any given time
• Proteomics analyses many samples using 2D-
electrophoresis and mass spectrometry
• High-throughput, but less than transcriptomics
Separation techniques
Separation techniques used with intact proteins

1D- and 2D-SDS PAGE


Separating intact proteins to take
Preparative IEF isoelectric advantage of their diversity in physical
focusing properties

HPLC

Separation techniques for peptides

MS-MS
HPLC (MudPIT)
SELDI

Differential display proteomics


Difference gel electrophoresis (DIGE)
Isotope-coded affinity tagging (ICAT)
Enrichment /Fractionation

For the detection of low-abundance proteins, a separation of


complex mixtures into fractions with fewer components is
necessary

•Enrichment from larger volumes Selective precipitation


Selective centrifugation
Preparative approaches

•Combination of 2DE with LC

•Multi-dimensional LC
Protein extraction
Detergents: solubilize membrane proteins-
separation from lipids

Reductants: Reduce S-S bonds

Denaturing agents: Disrupt protein-protein


interactions-unfold proteins

Enzymes: Digest contaminating molecules (nucleic


acids etc)

Protease inhibitors

Aim: High recovery-low contamination-compatibility with separation method


Protein digestion
Trypsin
Why digest the protein?
Cleaves at lysine and
Accuracy of mass measurements
arginine, unless either is
Suitability followed by proline in C-
terminal direction
Sensitivity

The ideal protein digestion approach


would cleave proteins at certain
specific amino acid residues to yield
fragments that are most compatible Good activity both in gel digestion and
with MS analysis. in solution

Peptide fragments of between Other enzymes with more or


6 – 20 amino acids are ideal less specific cleavage:
for MS analysis and database Chymotrypsin
comparisons. Glu C (V8 protease)
Lys C
Asp N
Separation by LC
Salt
gradient UV detector

column

EC detector
waste

Number of peaks indicates the complexity of starting material

Peak position (i.e. elution time) may provide qualitative information about the sample
(comparison with standards)

Peak area may provide information on relative concentration of components.

If coupled to MS protein identification (MW) can be provided

modified:www.dcu.ie/chemistry/ssg/images/Te
chni7.gif
Multidimensional HPLC

Mud PIT
Multidimensional Protein Identification Techniques or Tandem HPLC
the combination of dissimilar separation modes will allow a greater resolution
of peptides in mixture.

Ion-exchange Reversed phase

•Reversed phase, hydrophobicity


•Ion exchange, net positive/negative charge
•Size exclusion, peptide size, molecular weight
•Affinity chromatography, interaction with specific
functional groups
Gel Electrophoresis
• Electrophoresis separates molecules by size
• Resolution is limited

Berg, Tymoczko & Stryer, “Biochemistry”,


6th edn, 2006, p. 71
Gel electrophoresis
Classical process
High resolving power: visualization of thousands of protein forms
Quantative
Identifying proteins within proteome
Up/ down regulation of proteins
Detection of post-translational modifications
Coomassie blue stained gels

Protein fixing and staining or blotting


General detection methods (staining)
Organic dye – and silver based methods Coomassie blue, Silver
Radioactive labeling methods
Reverse stain methods
Fluorescence methods (Supro Ruby)
Ruby red
Gel scanning
(storage of image in a database)
Silver stained
Isoelectric point

•Proteins are amphoteric molecules


i.e. they have both acidic and basic functional groups

•pI= isoelectric point, is where the protein does not have any net
charge

•The protein charge depends on the pH of the solution.


Isoelectric Focusing
• Electrophoresis across a pH gradient
• Proteins migrate to their isoelectric pH

Berg, Tymoczko & Stryer, “Biochemistry”,


6th edn, 2006, p. 73
1st dimension
IsoElectric Focusing, IEF
Immobilized pH gradients (IPGs)
A pH gradient is generated by a limited
number of well defined chemicals
(immobilines) which are co-
polymerized with the acrylamide
matrix.

Migration of proteins in a pH
gradient: protein stop at pH=pI

Loading quantities (18 cm strip)


Analytical run: 50-100 μg
Individual strips: Use narrow range IPG strips to
Micropreparative runs: 0,5 – 10 mg
focus on particular pI range
24,18,11,7 cm long
3 mm wide
0,5 mm thickness
2nd dimension
pI
The strip is
loaded onto a
SDS gel
pH 10 Mw
pH 3

Staining !

Proteins that were


separated on IEF gel
are next separated in
the second dimension
based on their
molecular weights.
Two-dimensional Gel
Electrophoresis
• Protein sample
initially fractionated
in one dimension by
isoelectric focusing
• SDS-PAGE
performed
perpendicular to
original direction
• Separates proteins
according to pI and
mass Berg, Tymoczko & Stryer, “Biochemistry”,
6th edn, 2006, p. 74
SYLICA 'Omic Technologies – Bowater Feb 2013
Two-dimensional Gel
Electrophoresis

• Proteins from E.
coli separated by
2D-electrophoresis
• >1,000 proteins
can be resolved

Berg, Tymoczko & Stryer, “Biochemistry”,


6th edn, 2006, p. 74
Limitations/difficulties with the 2D gel
Reproducibility
Samples must be run at least in triplicate to rule out effects from
gel-to-gel variation (statistics)

Small dynamic range of protein staining as a detection


technique- visualization of abundant proteins while less abundant
might be missed.

Posttranscriptional control
Co-migrating spots forming a complex mechanisms
region
Incompatibility of some proteins with the
first dimension IEF step (hydrophobic
proteins)
Streaking and smearing
Marginal solubility leads to protein
precipitation and degradation- smearing
(Glycolysation, oxidation)
Weak spots and background
A Mass Spectrometer
source analyzer detector

The sample has to be introduced into the ionization source of the instrument. Once inside the
ionization source the sample molecules are ionized, because ions are easier to manipulate than
neutral molecules.

These ions are extracted into the analyzer region of the mass spectrometer where they are separated
according to their mass (m)-to-charge (z) ratios (m/z).

The separated ions are detected and this signal sent to a data system where the m/z ratios are stored
together with their relative abundance for presentation in the format of a m/z spectrum.

The analyzer and detector of the mass spectrometer, and often the ionization source too, are
maintained under high vacuum to give the ions a reasonable chance of traveling from one end of the
instrument to the other without any hindrance from air molecules.
..consists of..

source analyzer detector

Source -produces the ions from the sample (vaporizationMALDI, Matrix-Assisted Laser Desorption and
/ionization) Ionisation

ESI, ElectroSpray Ionisation


Mass Anlyzer - resolves ions based on their
mass/charge (m/z) ratio
Generate different, but
Detector –detection of mass separated ions complementary information
Mass Spectrometry
• MALDI-TOF mass spectrometry
• Protein sample is ionized and exposed to electrical field
• Ions travel according to size

Berg, Tymoczko & Stryer, “Biochemistry”,


6th edn, 2006, p. 94

SYLICA 'Omic Technologies – Bowater Feb 2013


MALDI
Matrix Assisted Laser Desorption and Ionisation
laser
ions
Peptides co-crystallised with matrix
++
+
+
- +
+ Produces singly charged protonated molecular
-
-
ions
High throughput
Single proteins

Rapid procedure, high rate of sample


throughput
large scale identification (“first look at a
sample”)
TOF
Time of flight

Measures the time it takes for the ions to


fly form one end to other and strike the
detector.
The speed with which the ions fly down
the analyzer tube is proportional to their
m/z values.
The greater the m/z the faster they fly
Separate ions o f different m/z based on
flight time
Fast
Requires pulsed ionization
MALDI-TOF
Matrix-assisted laser desorption ionization-time of flight

++
+ +
+ - +
-
-
TOF analyzer

Quick, easy, inexpensive Low reproducibility and repeatability of


single shot spectra (Averaging)
Highly tolerant to contaminents
Low resolution
High sensitivity
Matrix ions interfere in the low max range
Good accuracy in mass determination
Compatible with robotic devices for
high-throughput proteomics work
Best suited to measuring peptide masses
MALDI-TOF Mass Spectrum
• MALDI-TOF gives good estimates of molecular weights
• Can be used to identify a few proteins within a mixture

Berg, Tymoczko & Stryer, “Biochemistry”,


6th edn, 2006, p. 94
MALDI-TOF data
Every peak corresponds to the exact mass (m/z) of a peptide ion

112.1
234.4

Peak List = List of masses


890.5
1296.9
1876.4
= fingerprint

1987.5
…….

Modified from http://plantsci.arabidopsis.info/pg/day3practical1.ppt


Proteomic Analysis by Mass
Spectrometry
• Proteins separated by 2D
electrophoresis
• Single proteins eluted
• Digestion with trypsin
will give fragments with
unique set of sizes
• Sizes identified by mass
spectrometry and
matched to database
• Allows identification of
unknown proteins Berg, Tymoczko & Stryer, “Biochemistry”,
6th edn, 2006, p. 95
mAbs

ISSN: (Print) (Online) Journal homepage: www.tandfonline.com/journals/kmab20

High concentration formulation developability


approaches and considerations

Jonathan Zarzar, Tarik Khan, Maniraj Bhagawati, Benjamin Weiche, Jasmin


Sydow-Andersen & Alavattam Sreedhara

To cite this article: Jonathan Zarzar, Tarik Khan, Maniraj Bhagawati, Benjamin Weiche, Jasmin
Sydow-Andersen & Alavattam Sreedhara (2023) High concentration formulation developability
approaches and considerations, mAbs, 15:1, 2211185, DOI: 10.1080/19420862.2023.2211185

To link to this article: https://doi.org/10.1080/19420862.2023.2211185

© 2023 Genentech, Inc. Published with


license by Taylor & Francis Group, LLC.

Published online: 16 May 2023.

Submit your article to this journal

Article views: 6070

View related articles

View Crossmark data

Citing articles: 4 View citing articles

Full Terms & Conditions of access and use can be found at


https://www.tandfonline.com/action/journalInformation?journalCode=kmab20
MABS
2023, VOL. 15, NO. 1, 2211185
https://doi.org/10.1080/19420862.2023.2211185

REVIEW

High concentration formulation developability approaches and considerations


Jonathan Zarzara, Tarik Khanb, Maniraj Bhagawatic, Benjamin Weichec, Jasmin Sydow-Andersenc,
and Alavattam Sreedharaa
a
Pharmaceutical Development, Genentech Inc, South San Francisco, CA, USA; bPharma Technical Development Europe, F. Hoffmann-La Roche Ltd,
Basel, Switzerland; cLarge Molecule Research, Pharma Research and Early Development (pRED), Roche Diagnostics GmbH, Penzberg, Germany

ABSTRACT ARTICLE HISTORY


The growing need for biologics to be administered subcutaneously and ocularly, coupled with certain Received 27 January 2023
indications requiring high doses, has resulted in an increase in drug substance (DS) and drug product (DP) Revised 21 April 2023
protein concentrations. With this increase, more emphasis must be placed on identifying critical physico- Accepted 2 May 2023
chemical liabilities during drug development, including protein aggregation, precipitation, opalescence, KEYWORDS
particle formation, and high viscosity. Depending on the molecule, liabilities, and administration route, Aggregation; developability;
different formulation strategies can be used to overcome these challenges. However, due to the high high concentration
material requirements, identifying optimal conditions can be slow, costly, and often prevent therapeutics formulations; in-silico;
from moving rapidly into the clinic/market. In order to accelerate and derisk development, new experi­ physical instability;
mental and in-silico methods have emerged that can predict high concentration liabilities. Here, we subcutaneous
review the challenges in developing high concentration formulations, the advances that have been made administration; viscosity
in establishing low mass and high-throughput predictive analytics, and advances in in-silico tools and
algorithms aimed at identifying risks and understanding high concentration protein behavior.

Introduction
a formulation that does not meet the intended shelf life, especially
Monoclonal antibodies (mAb), and other protein-based thera­ in the case where there is a need to balance multiple competing
peutics, have become a huge success, as exemplified by the more degradation pathways. Furthermore, formulation development
than 100 different antibodies approved in the United States.1,2 can only improve high concentration behavior of biomolecules
The growing need for biologics to be administered subcuta­ to a certain extent. While some liabilities can be addressed effi­
neously and ocularly, which limits the total volume of drug that ciently (e.g., agitation-induced aggregation, particle formation),
can be delivered, coupled with certain indications requiring high others such as aggregation due to thermal unfolding and viscosity
doses, has resulted in a growing need for high concentration (>50 require the selection of suitable molecules for the intended pur­
mg/mL) formulations. While chemical liabilities such as deami­ pose before technical development is started. As such, the second
dation and isomerization are independent of the protein concen­ approach is to use a developability assessment program during the
tration, physical instabilities such as aggregation are protein candidate selection phase to identify potential liabilities.4–6 Early
concentration dependent. Due to this, high concentration mAb identification of such liabilities provides the opportunity to select
formulations can pose substantial manufacturing, stability, and a different variant or to attempt to fix the liability using protein
delivery challenges.3 As protein concentrations have increased, so engineering approaches. However, screening high concentration
has the occurrence of physical instability (e.g., opalescence, aggre­ liabilities requires large amounts of protein and therefore limits
gation, particles) and viscosity challenges.4 In addition, as mole­ the number of candidates that can be screened within a reasonable
cule formats have become more complex, high concentration time period and with limited resources.7 In addition, while there
instabilities have proven to be more challenging. has been success at identifying and removing chemical liabilities
Two general approaches are used to mitigate physical instabil­ (such as deamidation and isomerization), mitigating physical
ities and viscosity issues. The first is to optimize the protein instabilities is more complex. In these cases, there is generally no
formulation during development by finding conditions that mini­ single amino acid that needs to be replaced, as physical instabilities
mize potential liabilities (for example by changing the formulation are the result of protein-protein surface interactions. As such, the
pH and/or testing various excipients), thereby ensuring acceptable mechanisms behind aggregation and viscosity and approaches to
drug product manufacturing, administration conditions, and shelf predict, and therefore mitigate, such behavior have been the sub­
life. However, this approach does not allow the use of platform ject of much interest. Here, we review some of the recent work
formulations, as each molecule would require its own unique aimed at understanding, measuring, predicting, and mitigating
formulation, nor does it help with challenges during manufactur­ high concentration instabilities and viscosity challenges. For
ing where the protein may not be in its final formulation. The a comprehensive summary of the other aspects of developability,
process to optimize the formulation is often slow and may result in we refer you to a recent review.6

CONTACT Jonathan Zarzar zarzar.jonathan@gene.com Pharmaceutical Development, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
This article has been corrected with minor changes. These changes do not impact the academic content of the article.
© 2023 Genentech, Inc. Published with license by Taylor & Francis Group, LLC.
This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial License (http://creativecommons.org/licenses/by-nc/4.0/), which permits
unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited. The terms on which this article has been published allow the
posting of the Accepted Manuscript in a repository by the author(s) or with their consent.
2 J. ZARZAR ET AL.

Aggregation reported increased aggregation of an IgG2-trehalose formula­


tion when stored at −20°C. The glass transition temperature
Antibody aggregation is a substantial concern, as aggregates
(Tg’) of the matrix was determined to be −29°C. Interestingly,
are generally viewed as immunogenic, may be hyper-potent,
aggregation in this formulation increased with time at −20°C
and conversely can reduce overall efficacy.8 Aggregation
but not at −40°C or −10°C. This is most likely because of the
observed on stability may limit product shelf-life, which com­
crystallization of freeze-concentrated trehalose from the frozen
plicates supply chains. Aggregation occurs when a protein
solute at temperatures higher than the Tg’ of the matrix. Lack
forms high molecular weight species either through weak
of aggregation at −10°C was attributed to lower cryoconcen­
nonspecific interactions (self-association) or via covalent
tration of the protein at these temperatures, as well as higher
bonds (covalent-aggregation).9,10 In either case, it can lead to
mobility of the formulation that allows for any unfolded pro­
the formation of soluble (e.g., dimers, trimers) or insoluble
teins to refold back into native state. Connolly and others16
aggregates that can manifest as particulates. Protein aggrega­
expanded on the frozen instability studies and found that faster
tion can be influenced by a number of factors, such as mole­
cooling rates (>100°C/min) result in trehalose crystallization
cular properties; formulation composition, including active
and protein aggregation. They also reported that at lower
pharmaceutical ingredient (API) concentration; agitation;
cooling rates of≤1°C/min, trehalose remains predominantly
temperature; light; and contact with certain materials.11
amorphous and there is no effect on protein stability.
While controlling processing conditions, storage, shipment,
Interestingly, the authors found that phase distribution of
and handling may help reduce aggregation, it may not fully
amorphous and crystalline trehalose dihydrate in frozen solu­
mitigate aggregation. In those cases, other factors such as
tions depends on the ratio of trehalose to mAb. Based on these
molecular properties and formulation composition are impor­
observations, the authors found that an optimum ratio of 0.2
tant tools in reducing stability risks.
to 2.4 trehalose-mAb (w/w) ratio is ideal for frozen storage.
Optimization of formulations to prevent protein aggrega­
Bluemel et al.17 report an interesting cryoconcentration
tion is typically performed by leveraging prior experience
effect on mAb instability. Samples with lower mAb concentra­
combined with trial and error. In a typical study, the protein
tion showed increased formation of high molecular weight
is formulated in a variety of buffers, surfactants, excipients,
species (i.e., aggregates). In contrast, higher concentrated sam­
and pH levels and stressed via elevated temperature, agitation,
ples led to more subvisible particles (SVPs) when the samples
and freeze/thaw conditions to identify the optimal formulation
were stored above Tg’, that is, at −20°C or at −10°C. The
with minimum aggregation propensity. However, the propen­
authors went on to show that the protein and small excipients
sity of a molecule to aggregate at elevated temperatures may
freeze-concentrate differently, resulting in different local pro­
not be predictive of the propensity to aggregate at the intended
tein to stabilizer ratios within a container, ultimately leading to
storage temperature due to differences in degradation path­
mAb instability under these storage conditions.18
ways. Recently, Bunc et al. were able to predict the aggregate
Interestingly, Bluemel et al.19 have recently reported on the
fractions after three years of storage at 5°C using accelerated
use of computational fluid dynamic (CFD) simulations of
temperature degradation.12 The authors used a branched
mAb solutions. They conclude that CFD simulations are
kinetic model that breaks the stability behavior into a low
a promising tool to describe large-scale freezing and thawing
temperature and a high temperature pathway. Using this
of mAb solutions and could potentially save time and
model, the authors were able to predict the aggregation of six
resources. In addition to these instability pathways,
antibodies and resolved the curvature that is typically observed
a relatively new hypothesis on the interactions of proteins
when applying an Arrhenius model. The data also suggested
with air bubbles leading to aggregation has been proposed by
that at low temperature, aggregation is triggered by chemical
Salnikova et al.20 Authelin et al. recently published a detailed
modifications, while unfolding is the main driver at high
perspective covering these topics.21 In summary, higher pro­
temperatures.
tein concentrations in the DS can result in frozen instabilities
and have to be carefully evaluated.
Aggregation during frozen storage
Viscosity
The challenges of protein aggregation are not limited to hand­
ling at drug product (DP) storage temperature of 2–8°C and/or Self-association of mAbs increases with concentration and can
handling during typical production (e.g., 18–25°C). Frozen result in high viscosity, which challenges the capability of the
storage (</= −20°C) is a standard practice for protein drug ultrafiltration diafiltration (UFDF) unit operation during
substance (DS), and the storage temperature and storage mate­ manufacturing, resulting in slow operation and/or an unac­
rial must be carefully selected. Various stress factors that con­ ceptable pressure in the system. If the viscosity is only slightly
tribute to protein instability during DS processes have recently elevated, it is possible to mitigate it using single pass tangential
been reviewed by Das et al.13 and will not be reviewed herein. flow filtration (TFF) or increasing the recirculation tempera­
However, while the general assumption is that frozen storage ture to decrease the viscosity. However, increasing the recircu­
increases the stability profile, high concentration mAb formu­ lation temperature can also result in an increase in aggregation
lations have been found to be susceptible to instabilities such as and chemical degradation. High viscosity can also affect the
increased aggregation during frozen storage. Lazar et al.14 have filling operation, including the accuracy and fill rate. If the fill
shown that mAbs have a potential to undergo cold denatura­ rate is slowed enough, or stopped due to an interruption,
tion at storage temperatures near −20°C at pH 6.3. Singh et al.15 drying at the filling needles could occur, which can lead to
MABS 3

aggregates/particulates in the final drug product or yield loss is used in the 200 mg/mL polyclonal human IgG drug product
due to the need to flush the filling lines. During administra­ Hizentra.26 Other amino acids and excipients that have hydro­
tion, high viscosity can prevent the use of a syringe and needle, phobic and charged properties have also been shown to lower
in particular for subcutaneous administration with a pre-filled viscosity in certain cases, furthering the understanding that
syringe. Above a certain viscosity, the injection force required disrupting API network formation is key to lowering
to expel the solution might be too high for certain populations viscosity.27,28
(e.g., geriatric populations) and/or dose volumes. It is impor­ Other viscosity-lowering approaches use the formation of
tant that instructions for use and other training material nanoclusters or microparticles that restrict the viscosity effects
clearly highlight any product needs for warming the DP to to interparticle interactions, rather than intermolecular inter­
room temperature prior to injection, if needed to reduce visc­ actions. This approach creates two separate dominant forms of
osity to a level that is convenient for administration. Although molecular interactions, those within the particles, which do
the use of autoinjectors or other devices can overcome some not have a major influence on viscosity, and between the
limitations imposed by high viscosity, they can add complex­ surfaces of the particles, which is less than that of soluble
ity, lengthen development time if an autoinjector must be monomers. In one example, nanoclusters were formed by
available during pivotal clinical trials, and increase the cost of lyophilizing a mAb formulation containing trehalose as
goods. Therefore, substantial effort has gone into the develop­ a crowding agent and reconstituting it in a buffer close to the
ment of high concentration protein formulations with low mAb’s isoelectric point using a lower volume than the pre-
viscosity. One approach is to use excipients that can help lyophilization volume. This induced macromolecular crowd­
lower the viscosity. The other approach is to use experimental ing and led to the formation of nanoclusters that revert back to
and computational approaches to measure and predict viscos­ monomer upon injection.29,30 Other particle-based technolo­
ity in order to help select molecules with low viscosity. gies, using techniques such as electrospraying and microglas­
sification offered by companies like Elektrofi and Lindy
Biosciences, claim concentrations >400 mg/mL may be
Viscosity reducing formulations
reached. However, these microparticle approaches may require
Viscosity is the result of noncovalent intermolecular interac­ substantial process development and require the use of non-
tions that result in the formation of a molecule network. As aqueous solvents for the DP to be administered.
a result, any molecule or excipient that has the ability to
disrupt the network can decrease the viscosity of the sample.
Experimental measurement and prediction of
Charge-based interactions tend to have the biggest impact, as
viscosity, solubility, and aggregation
they affect molecules over longer ranges compared to hydro­
phobic interactions. Sodium chloride can lower the viscosity of High concentration behavior is influenced by a complex
a formulation by shielding protein charge and therefore dynamic of different molecular interactions (prominently
decreasing electrostatic driven protein-protein interactions hydrophobic and electrostatic). Proximal energy effects caused
(PPI). In contrast, it has been hypothesized that arginine by molecular crowding in high concentrated protein solutions
works by direct binding to the protein surface.22 This can further complicate the prediction of thhe behavior based on
result in both charge shielding, as well as a reduction in single parameters.31 One of the major liabilities for molecules
hydrophobic forces by interacting with aromatic residues. intended for high concentration formulation is molecular self-
While typically salts such as sodium chloride decrease the interaction. Depending on the exact nature of those interac­
viscosity, there have been reports of it increasing the tions and the underlying cause, a strong tendency to self-
viscosity.23,24 As such, each salt has to be evaluated on a case- interact can lead to high viscosity, opalescence, aggregation,
by-case basis, making evaluation a very slow and tedious and/or particle formation. Colloidal interactions in a native,
process. Caffeine has also shown promise at reducing viscosity, chemically modified, or (partially) unfolded state can have
and in some cases performed better than both sodium chloride distinct consequences for a protein in solution. Furthermore,
and arginine.24 Results obtained by Zeng et al.24 suggested that molecules are exposed to different unfavorable conditions
formulating with caffeine reduces the attractive PPIs, leading during manufacturing and administration that can trigger
to lower viscosity. Using rigid body docking simulations, the degradation via distinct pathways. Given the complexity of
authors found that caffeine may preferentially bind to posi­ underlying molecular mechanisms of self-interaction and the
tively charged groups and aromatic groups on the protein variety of external stressors, it is clear that there is no simple
surface. one-size-fits-all solution for the prediction of high concentra­
Other excipients have also been evaluated alone, or in tion behavior in biopharmaceutical development. Even small-
combination, to address viscosity and stability issues. scale experimental evaluation of viscosity, solubility, and
Recently, Banik et al.25 reported that the use of excipients aggregation under specific conditions requires large mass
such as carnitine HCl, (S)-(+)-camphorsulfonic acid, and amounts of protein due to the need to formulate at high
ornithine HCl resulted in varying effects to viscosity and self- concentration and the volume required by the analytics. For
interaction parameters that were also pH dependent. These example, the accepted method for evaluating the viscosity
findings again highlight how the charge distribution on mole­ behavior of a liquid dosage form at high concentration
cules can have a substantial impact on what excipients are involves formulating the protein at multiple concentrations
most effective in reducing viscosity. Proline is an example of and generating a viscosity curve using a cone and plate rhe­
another excipient that has been shown to reduce viscosity and ometer. Due to the high mass requirements (~25–75 mg),
4 J. ZARZAR ET AL.

screening multiple variants can be slow and/or cost characterization of antibodies was affinity-capture SINS
prohibitive.4 As such, there is much interest in methods that (AC-SINS).36,37 In a seminal work, Lie et al.37 optimized
can increase throughput with lower mass requirements the experimental steps of AC-SINS and used it to measure
(Table 1). the self-interaction propensity of more than 400 mAbs.
Molecular self-interaction in the formulation is determined Using the known high concentration properties of a small
by three major factors: 1) the inherent physicochemical char­ subset of these mAbs, the authors claimed to predict the
acteristics of the protein, 2) the concentration of the protein in developability of the entire panel. However, no attempt was
solution, and 3) the presence of other components in the made to correlate these predictions with actual analysis at
solution (e.g., excipients). While the desired final concentra­ high protein concentration for the entire panel. The biggest
tion is mostly determined by the required clinical dose and the drawback of AC-SINS is that it is only applicable under
route of administration, both molecule selection and formula­ physiological buffer conditions (PBS, pH 7.4), which prohi­
tion development (or early evaluation of formulation suitabil­ bits testing of proteins in formulation buffers at pH 5–6.
ity) can be guided using assays predicting high concentration Attempts to overcome this limitation have focused on engi­
behavior. In the following section, we discuss small scale (low neering the surface properties of the AuNPs by functionaliz­
mass) options of these assays and give an overview of available ing them with polymers that sterically or electrostatically
tools for the characterization of observed instabilities promi­ stabilize the nanoparticles. A recently developed method in
nent in these solutions. this area, termed PEG-stabilized SINS (PS-SINS), uses poly­
ethylene glycol (PEG) functionalization to extend the applic­
ability of the method to a wider pH range.38
Viscosity and self-interaction
A complementary method, which uses poly-L-lysine (PLL)
The first set of methods for assessing self-interaction propen­ functionalization (termed charge stabilized SINS, CS-SINS)
sity of proteins at low concentrations is based on assessing to achieve a similar extension in applicability, was used to
the second virial coefficient (termed A2 or B22), which analyze the behavior of a panel of 56 antibodies composed of
describes the weak pairwise interaction between two solute molecules with IgG1, IgG2, and IgG4 frameworks.39 The
molecules in a solvent. A small-scale study of three mAbs authors showed that the behavior of IgG1 and IgG2 mAbs
demonstrated that A2 can be an effective low concentration in the CS-SINS assay correlated very well with their high
(<20 mg/ml) predictor of protein aggregation and viscosity at concentration viscosity and opalescence properties.
high concentrations.32 A less tedious alternative to measuring However, this correlation was much weaker for IgG4
A2, depending on the available instruments, is to measure its mAbs, which shows that different molecular pathways con­
component, the diffusion interaction parameter, kD, which is tribute to high concentration liabilities even for closely
amenable to high-throughput assessment using dynamic light related molecule types.
scattering (DLS).33,34 Using a panel of 29 mAbs (28 IgG1 and 1 An alternative method to study protein self-interaction
IgG4) Connolly et al.33 demonstrated that kD is a useful pre­ in high throughput is based on biolayer interferometry
dictor of viscosity in buffer systems of both high and low ionic (BLI). Initially described by Sun et al.,40 Domnowski et al.41
strengths. Kingsbury et al.34 expanded on this study by mea­ implemented the method on an Octet HTX system, thereby
suring kD for 59 mAbs (44 IgG1, 4 IgG2, and 11 IgG4) and substantially increasing the throughput. This allows the
trying to correlate the outcome with their high concentration ranking of molecules in a standard buffer system, as well
opalescence and viscosity behavior. The authors found that kD as the early evaluation of different formulations and their
measured under dilute concentrations in a low ionic strength impact on self-interaction. The broad buffer compatibility
His-HCl buffer could very well predict high concentration of the self-interaction (SI-) BLI technology and the com­
opalescence and viscosity liabilities. They could also set a kD mercial availability of sensor tips with different capture
cutoff of 20 mL/g as a threshold below which most of the functionalities are advantages of this approach. However,
antibodies in their panel tended to show high concentration only indirect correlations to high concentration properties
opalescence or viscosity. have been established.
A second family of methods that have proven useful for One early approach for the determination of self-
measuring the self-interaction between protein molecules interaction parameters, such as the second virial coefficients,
using extremely low amounts of protein fall under the is self-interaction chromatography (SIC). Using
umbrella of self-interaction nanoparticle spectroscopy a chromatographic resin functionalized with a protein of inter­
(SINS)-based techniques. These approaches take advantage est, the retention time of the protein can be correlated to its
of the extreme sensitivity of the localized surface plasmon self-interaction propensity.42,43 While both SINS and SI-BLI
resonance spectrum of gold nanoparticles (AuNP) to the are assays designed with high-throughput applications in
inter-nanoparticle distance to report on the self-interaction mind, SIC can be used to characterize the self-interaction of
of test proteins that are immobilized on the AuNP-surface.35 a protein in more detail, detecting heterogeneous behavior and
The extremely low protein concentrations (1–100 μg/ml) and even sampling fractions of broad peaks to better understand
mass (μg scale) needed, the applicability with unpurified the heterogeneity. Furthermore, an in-depth characterization
samples, and amenability to high-throughput analysis make of the influence of solution components is possible to guide
these techniques suitable for application at very early project formulation or process development for a low number of
phases, including during binder screening. One of the first molecules.44 However, the preparation of a specifically func­
reported variants of a SINS method used for the tionalized column resin for SIC is both skill and labor intensive
MABS 5

and requires relatively large amounts of protein. Therefore, the above mentioned self-interaction assays, and similar to
SIC is less suitable for ranking many molecule candidates light scattering-based kD determination, PEG/AS precipita­
during developability assessment. tion does not require the immobilization of the interacting
In contrast, cross-interaction chromatography (CIC) cir­ molecules and thus avoids potential artifacts arising from this.
cumvents the need for a specific column preparation for each
individual mAb.44,45 Attachment of a polyclonal IgG mixture
Aggregation and characterization of observed species
or unrelated mAbs to the resin allows a higher throughput of
mAb candidates to be evaluated. The relatively large amount of Low volume and/or low concentration stability studies are
sample needed to specifically functionalize a column resin for a standard part of the developability assessment for protein
SIC is also omitted, making CIC a more suitable option for therapeutics. Aggregation liabilities can be identified using
early molecule assessment. Nevertheless, a good inverse corre­ a combination of freeze-thaw stress, mechanical stress, and
lation between retention time on the column and protein high-temperature stress conditions using DP storage and phy­
solubility was shown for a series of mAbs evaluated by Jacobs siological conditions. Interestingly, in a study of 137 mAbs
et al.45 Potential association of test molecules with the general from different stages of clinical development, aggregation was
structural features of IgGs caused by Fab-Fc or Fc-Fc interac­ observed at 40°C (stress stability) only weakly correlated to
tions can explain these observations. However, it must be other parameters such as self-interaction.50 Therefore, the
considered that a specific case of self interaction, caused by specific evaluation of stability in small-scale experiments is
mutual association of features within the complementarity- still necessary to cover this important aspect of developability
determining regions (CDRs), will be missed using CIC. This assessment. The design of such studies requires careful con­
disadvantage can become more relevant when evaluating new sideration since, at high concentration, molecular crowding
formats, especially when the constant part of the molecules (or molecular proximity) effects lead to a shift in prominence
compared to the variable part comprises a smaller percent of of different forces that influence molecular interactions. Short-
the overall construct (e.g., Fab fragments). While SIC is range forces such as van der Waals and hydrophobic interac­
a powerful tool to characterize one or few final candidates tion become more important factors of protein-protein inter­
with regards to the exact influence of the solvent, CIC can be actions at high protein concentrations. In contrast, protein-
used during early selection of candidates in molecule protein interactions at lower concentrations (and therefore
discovery. longer molecular distances) are dominated by longer range
charge or dipole effects.31 As such, choosing suitable condi­
tions and a relevant concentration for accelerated stability
Solubility
assessment is crucial to avoid missing molecule liabilities that
Solubility, or propensity to precipitate, is an important factor only become apparent at higher concentrations (>100 mg/mL).
in the formulation of stability and API behavior upon admin­ Furthermore, specific routes of administration can result in
istration to patients. Determining a molecule’s solubility in a high local concentration under physiological conditions, and
a low volume assay can be a useful tool for increasing the efforts should be made to assess the potential aggregation risk
number of molecules that can be screened in early project under physiological conditions. Given the complexity and
stages. This can be accomplished in a high-throughput manner diversity of the physiological environment to which a protein
by inducing precipitation at low concentration by the addition therapeutic can be exposed, small-scale stability conditions
of ammonium sulfate (AS) or polyethylene glycol (PEG). tailored to reflect this could become a prerequisite for devel­
While PEG mostly acts via the excluded volume effect (“mole­ opability assessments in the future.51 The relevance of confor­
cular crowding”), AS potentially alters the interaction behavior mational stability (routinely determined by thermal unfolding
of molecules.46 Therefore, PEG is considered to be the pre­ and the calculation of the denaturation midpoint temperature,
ferred precipitant in such experiments. The general principle is Tm) as compared to colloidal stability for protein aggregation,
to add different concentrations of the precipitant and measure is controversial. Jain et al.50 did not report any correlation
the free soluble protein concentration after separation of solu­ between Tm and accelerated stability results, indicating that
ble and insoluble fractions.47 The resulting midpoint concen­ under common small-scale stress conditions (and in the range
tration of PEG is a good indicator for protein solubility and of Tm values represented in the mAb test-set), this parameter
correlates well with other predictors such as kD.48 An alter­ has no significant impact on aggregation.
native study describes using microscopy to qualitatively assess Regardless of the study design, detailed characterization of
precipitation and other effects such as phase separation or the observed impurities/degradation products (prominently
using turbidimetry as a direct readout for precipitation with­ high molecular weight species and/or particles) is required.
out the need for prior separation of insoluble fractions.49 This endeavor can be challenging and requires the use of
Advantages of this method include the low protein concentra­ a specialized toolkit of analytical methods. Apart from the low-
tion required (in the range of 1 mg/mL for IgGs) and the resolution biophysical methods used for investigation of pro­
potential for high-throughput application with an optimized tein oligomerization and complex formation, such as DLS or
small-scale assay and a suitable readout.49 Scannell et al.48 also more time-consuming experimental techniques like analytical
stated that PEG precipitation might be superior to DLS-based ultracentrifugation, the most established and representative
kD determination when working with ionic excipients. method for assessment of species of different molecular
Therefore, early formulation screening for challenging mole­ weights in the biopharmaceutical industry is size exclusion
cules could benefit from this approach. In contrast to most of chromatography (SEC). While SEC alone does not provide
6 J. ZARZAR ET AL.

an accurate size of the observed species, coupling it to a multi- molecule formats (e.g., Fabs, DutaFabs, fusion proteins,
angle light scattering (MALS) detector enables determination Contorsbodies).56,57 In fact, Starr et al.39 observed differences
of the molar mass and concentration, which are proportional in the correlation between CS-SINS score and diffusion inter­
to the light scattered by the particles.52 Despite the wide and action parameter [kD] when evaluating different IgG sub­
validated applicability of the SEC method for release and classes. It would not be surprising if similar differences are
stability monitoring, further investigation of aggregation phe­ observed for more diverse formats. Similarly, Domnowski et ­
nomena may require orthogonal techniques to SEC to gain al.,41 while showing a good correlation between response rate
additional or more precise data that is typically not necessary in SI-BLI to kD by DLS, observed exceptions that indicate that
for release and stability monitoring. the exact method setup (e.g., molecules immobilized vs free in
Such orthogonal techniques include native mass spectro­ solution) could bias the obtained results. While for early mole­
metry (MS) coupled to liquid chromatography for separation. cule selection, these exceptions are acceptable, a late develop­
Modern ultra-high-mass-range (UHMR) mass spectrometers ability assessment of a limited number of candidates could
allow acquisition of high-quality mass spectra in order to benefit from a confirmation using a second orthogonal
determine molar masses of up to several MDa with superior method. Additional studies are needed to determine which
mass resolution and accuracy.53 Nevertheless, native MS is assays have broader general applicability or can be quickly
a gas-phase technique, which requires volatile buffers and has adjusted to format needs and which approaches might be too
a low tolerance for salts and detergents.52 specific to keep pace with a rapidly evolving drug discovery
Another orthogonal method, mass photometry (MP), can pipeline.
be used for the analysis of proteins and protein complexes
under native buffer conditions using very low sample quanti­
In-silico approaches for viscosity and aggregation
ties. This is achieved by the use of interferometric scattering
prediction
microscopy, which enables detection and quantification of
light scattered by single particles.52 The free choice of buffers In addition to the development of high-throughput analytics to
for investigation of observed impurities, as well as its high measure and understand high concentration liabilities, there
sensitivity are advantages of this technology. MP provides have been many advances in the use of computational
quantitative, label-free detection and mass determination of approaches. Studies have suggested that electrostatic and
biomolecules. In addition, MP makes studying stoichiometry, hydrophobic interactions both play a role in viscosity, solubi­
energetics and dynamics of protein complexes possible.54 lity, and aggregation of proteins.58–61 In particular, it has been
While the low sample consumption of MP is an advantage, shown that negative charge on the variable fragment (Fv) of
MP is only applicable at nanomolar or lower concentrations a IgG1 mAb correlates with high viscosity, presumably due to
and is thus unable to detect unstable high molecular weight intermolecular interactions with corresponding positive
species that dissociate at such low concentrations. charge patches on Fc regions.62 This has led to the develop­
Furthermore, the resolution of MP is lower than that of MS ment of various methods that quantify the charge and hydro­
and, while sufficient to detect aggregation/multimerization, phobicity of proteins and then attempt to correlate those
can fail to resolve species originating from degradation of results to viscosity and aggregation. The first generation of
the API. methods relied on protein sequence to calculate properties of
To complement these methods and cover the full size range interest, such as the isoelectric point or hydrophobic content,
of possible aggregates, techniques for investigation of visible and predict high concentration behavior. One such approach
and subvisible particles such as optical microscopy, light correlated the viscosity of 14 antibodies with the net charge at
obscuration, membrane microscopy, flow imaging, conductiv­ the formulation pH, charge symmetry, and the hydrophobicity
ity-based particle counter, and fluorescence microscopy, are index.62 In that study the authors showed that hydrophobicity
available.55 Additionally, laser diffraction, as well as DLS, and charge distribution affect the solution viscosity, while
nanoparticle tracking analysis, or turbidimetry/nephelometry increased charge decreases it. Similar sequence-based
can be used to assess the size of particles. Den Engelsman approaches, including methods such as AGGRESCAN,
et al.55 have made a comprehensive comparison of these meth­ PASTA, TANGO, and Zyggregator, have been used to predict
odologies regarding their power of observations, their advan­ protein aggregation.63–67 However, these methods are not anti­
tages, and disadvantages. body specific, tend to have high levels of false positives for
mAbs, and ignore the contribution of neighboring residues.
This led to the creation of structure-based prediction methods
Current state of the experimental toolbox for high
that take into account the three-dimensional (3D) protein
concentration behavior prediction
structure to gain a better representation of surface patches
While high-throughput predictive assays with impressive cor­ that could result in protein-protein interactions. One such
relations to unwanted high concentration solution behavior of method is AGGRESCAN3D (A3D), which combines the
proteins have been developed, it is currently impossible for aggregation propensity of amino acids obtained from
a single assay to predict which liability will ultimately be AGGRESCAN with structural information from the 3D pro­
manifested.38,39,41,49 Furthermore, most assays have been tein structure to identify aggregation prone regions.68,69 By
developed for standard IgG formats. Due to the complexity taking into account only residues that are surface exposed
of the underlying molecular mechanism of self-association, the and within a spherical region around the residue’s central
limitation of these assays becomes relevant for alternative carbon, A3D is able to lower the false positive rate, as it ensures
MABS 7

only residues that are able to interact are taken into account. surface hydrophobicity, positive and negative CDR charge and
Another method known as the spatial charge map (SCM) uses charge asymmetry. The authors then demonstrated that TAP
homology modeling to build the 3D structure of the Fv region was able to flag both an affinity-matured molecule that exhib­
and calculates the spatial summation of charge of the surface ited high levels of aggregation due to a large surface hydro­
exposed residues.70 While SCM has been shown to differenti­ phobic patch, as well as another antibody that exhibited poor
ate between high and low viscosity molecules using three expression. Using such tools, residues of interest could be
different datasets, the cutoff between the two populations identified and used to introduce mutations that either add or
varied each time. This could result from differences in experi­ remove charge or change a hydrophobic patch and thereby
mental conditions between datasets or the method picking up improve a molecule’s developability.
differences in molecule frameworks. In addition, SCM does Beyond calculating the charge and hydrophobicity of
not account for the effect of buffer on viscosity, and thus this a molecule, it has become increasingly common to calculate
method cannot be used to determine if the high viscosity can additional descriptors that can correlate with high concentra­
be fixed via formulation changes. tion liabilities. For example, Li and coworkers modeled the Fv
Similar approaches can be taken to calculate hydrophobi­ regions and calculated molecular descriptors to understand
city of an antibody with one common method being the spatial and predict the viscosity of antibodies.75 The authors used
aggregation propensity (SAP). SAP measures the hydrophobi­ a panel of 11 antibodies, all formulated under-standardized
city of a region by taking into account the surface exposure and conditions and found that the charge, zeta-potential, and pI of
hydrophobic nature of particular amino acids and those prox­ the Fv region correlated with the viscosity of highly concen­
imal to that residue.7 Compared with purely sequence-based trated solutions. In addition, at 150 mg/mL, molecules with
approaches, SAP is able to determine hydrophobic patches high viscosity had negatively charged patches on the Fv region,
created by multiple hydrophobic residues that are near each which could interact with the positively charged IgG1 Fc
other in the folded state. To demonstrate the applicability of regions. This was similar to Vikas et al.’s results showing Fv
SAP, Chennamsetty et al. used two antibodies with high charge distribution affected viscosity.62 However, using only
hydrophobic scores, identified the regions responsible for the the Fv to understand high concentration liabilities disregards
hydrophobicity, and introduced single or multiple point muta­ the effect of the constant regions, which have been shown to
tions to lower the hydrophobic nature.7 After incubating the engage in transient intermolecular networks.76,77 It is also
mutants at elevated temperatures, they demonstrated that the possible that the introduction of Fc mutations (for example
new variants had higher thermal stability as measured by for half-life extension) can destabilize the Fc and alter protein-
the percent monomer still present after thermal stress. In protein interactions.78 To account for this, there has been
addition, functional analysis demonstrated that they preserved a shift to modeling the whole antibody. In one example,
the antigen-binding activity when mutations were not in the Tomar et al. predicted the concentration-dependent viscosity
CDR. Such a technique can prove useful for selecting as well as curve from the combination of experimental data and homol­
optimizing therapeutic candidates.71 Another method known ogy-based structural models.79 Using molecular descriptors
as Aggscore takes into account the distribution of hydrophobic calculated on 16 mAbs, the authors found that the hydropho­
and electrostatic patches on the protein surface.72 As opposed bic surface area of the full-length antibody, as well as the
to SAP, Aggscore has a scoring function that takes into account charges on the VH, VL, and hinge region could be used to
the intensity and relative orientation of the respective surface predict the viscosity curves. Similar to other works, the robust­
patches. However, Aggscore was not trained on antibodies and ness of the method still needs to be determined since the
while the authors correlated Aggscore to hydrophobic interac­ results are limited to a small dataset and a single formulation
tion chromatography (HIC), CIC, and standup mono-layer condition.
adsorption chromatography (SMAC), the method was not While homology modeling the whole antibody further
evaluated against high concentration properties such as visc­ improves model accuracy, molecules are inherently dynamic
osity, aggregation, or opalescence. and that motion can result in different descriptor values when
Building on SAP, Lauer et al. devised the Developability compared to those from a single static structure. To account
Index (DI), which incorporates the competing effects of elec­ for this, it is becoming common to use molecular dynamics
trostatic and hydrophobic interactions.73 Using a dataset of 12 (MD) to capture the side chain fluctuations and understand
antibodies of different subtypes, the authors were able to gen­ the impact of conformational dynamics on antibody proper­
erally predict the aggregation at elevated temperatures and ties. In addition, MD can be used to identify key residues that
across a range of pH. While the method provides a general may be involved in protein-protein interactions and propose
trend of aggregation propensity, the validation dataset is lim­ mutations to disrupt such interactions.80 Furthermore, MD
ited, and it does not give insight into other high concentration can also identify additional types of interactions such as
liabilities such as viscosity or opalescence. Similarly, Raybould cation-π and/or π-π, which may contribute to high concentra­
et al. developed the Therapeutic Antibody Profiler (TAP) to tion behavior.80 However, doing full atom molecular dynamics
provide developability rules similar to the Lipinski rules for on a complete antibody is computationally intensive and hence
small-molecule development.74 To develop TAP, Raybould limits the speed and throughput as a screening tool. With the
et al.74 modeled the variable domain of clinical stage introduction of even larger complex structures, the limitation
antibodies50 and used them to determine five metrics that becomes even more pronounced. One way to overcome this
were thought to be important in developability. The metrics limitation is to coarse grain (CG) model the molecule, which
include the length of the CDRs, the extent and magnitude of allows the simplification of an antibody to a reduced system
8 J. ZARZAR ET AL.

composed of 8–16 beads, with each bead containing a single (SASA) of the CDR-H2/H3 region correlated with the aggre­
charge representing the summation of all partial charges in gation rate. The SAP value of the CDR-H3 was also identified
that domain.76,77,81–83 Not only does this allow the modeling of as being important, which agreed with previous studies that
the whole antibody, but also the study of interactions between charge and hydrophobicity are key factors. Comparison of the
multiple copies of a protein or even multiple proteins. In one two-factor linear model with the dynamic mode of A3D
study, a 12 and 26 bead model was constructed to model two showed that the new model performed much better for this
antibodies that had different viscosity behavior at high protein limited dataset (prediction coefficient of 0.71 vs 0.28). In
concentrations.77 Using such an approach, the authors studied a subsequent study, Lai et al. measured the viscosity of 27
the impact of domain-level charge-charge interactions on the commercially approved mAbs and used the experimental
two antibodies and visualized the type of interaction (Fab-Fab data, along with computationally derived descriptors, to
or Fab-Fc). Interestingly, the antibody that had high viscosity develop a decision tree to classify antibodies into high and
at high concentration formed dense clusters, whereas the anti­ low viscosity.85 The authors found that antibody net charge
body with low viscosity at high concentration did not. To and Fv amino acid composition drive viscosity. In particular,
further understand the mechanism behind network formation, they found that the number of hydrophilic and hydrophobic
the authors investigated the impact of charge swap mutants on residues in the Fv region are important. Expanding the dataset
the viscosity of the two antibodies and found differences in the to predict the aggregation propensity of 20 pre-clinical and
number of Fab-Fab interactions across the variants.82 Similar clinical stage antibodies gave mixed results.88 In the case of
results were found by Buck et al. when they studied four aggregation, the authors found that positive SCM values and
different antibodies using CG models.76,77,81–83 In this case, the solvent-accessible surface area of hydrophobic residues on
the authors used an asymmetrical model that is more repre­ the variable fragment were the most important, which agreed
sentative of the structure in solution and found that molecules with their previous results. However, for viscosity, the authors
with higher viscosity also had a higher number of pairwise found that the model developed with commercial antibodies
interactions.76 While these studies highlight the importance of did not adequately predict the viscosity of the clinical-stage
electrostatic interactions in network formation, hydrophobic antibodies.88 A potential explanation is that the original model
interactions will likely play an important role when the inter­ was trained on too small of a dataset, resulting in feature
molecular distance decreases. Izadi et al. expanded the pre­ selection that is not representative of a large population of
vious work and developed a CG model that not only accounts antibodies. The smaller the dataset, the higher the risk of
for higher-order electrostatic multipole moments, but also for overfitting ML models, resulting in models that are not pre­
hydrophobicity.83 After tuning the model with data from three dictive when applied to a new dataset. Since datasets of high
antibodies, the authors predicted the viscosity behavior of 12 concentration molecules are limited, this becomes a recurring
different antibodies and demonstrated that the model could be problem in predicting high concentration liabilities. In addi­
useful in predicting the effect of ionic strength on the viscosity tion, given the expense of producing such data, it is likely that
of solutions. Such an approach could be useful to determine this will remain a bottleneck for some time. To try to overcome
the likelihood that a high concentration formulation could be this, the authors combined both commercial and clinical-stage
developed in the case where no low viscosity variants exist. antibodies to produce a model trained on 47 molecules. In that
In addition to the use of MD and coarse grain modeling, model, the two most important parameters are the number of
there has been an increased interest in using machine learning hydrophobic residues and net charges on the light chain vari­
(ML) to predict high concentration liabilities as well as for­ able region. More data will be required to determine if the new
mulation fixes for those liabilities.84–91 After removing high model accurately predicts high concentration behavior.
correlated descriptors, Lai et al. used a dataset of 21 approved Another model developed by Cloutier et al. used an elastic
antibodies to develop an aggregation model.87 In that model, net (EN) and support vector machine (SVM) model to predict
the positive SCM values and the solvent-accessible surface area antibody-excipient interactions for various excipients

Table 1. Overview of small-scale assays to prediction solution behavior of proteins in high concentration formulations.
Method Parameter Predictive for Material need Throughput References
33
AUC ks viscosity high low
25,33,34,98
DLS/SLS Kd/A2 (self-interaction) Viscosity, aggregation/ medium low/medium
particle formation
36,39
SINS Wavelength shift (self-interaction) Viscosity, opalescence low high
42–45,99
SIC/CIC Retention time (self-interaction) medium/high low/medium
40,41
SI-BLI Binding response rate (self-interaction) Viscosity, opalescence low high
100,101
Scissor turbidity Precipitation upon high low
injection
48,49
PEG/ammonium sulfate Equilibrium protein concentration as function low/medium medium/high
precipitation of precipitant concentration (solubility)
Small scale stability (F/T, Aggregation, particle medium/high low
temperature, mechanical formation (*)
stress)
(*) relevant concentration of proteins has to be used, long term high concentration exposure under physiological conditions becomes more relevant in e.g.,
Ophthalmology.
MABS 9

including sucrose, arginine hydrochloride, and sodium formulations. Not only is the number of potential format
chloride.91 In that study, the aggregation and viscosity beha­ combinations increasing,94 but in-silico modeling of such for­
vior of antibodies was correlated with the relative strength and mats still remains relatively unexplored.
weakness of the protein-excipient interactions. The authors Building on the theme of more data, it is becoming
proposed that such an approach can be used in combination increasingly common to use multi-attribute monitoring
with SAP and SCM to determine when to mutate the antibody (MAM) methods for the residue level quantification of
to correct such liabilities. antibodies.95–97 The use of such methods provides deeper
In summary, many advances in predicting molecule aggre­ characterization of product variants and could be useful in
gation and viscosity have been made. Although the approach furthering our understanding of protein aggregation, in
behind each method varies, charge and hydrophobicity are particular during long-term and accelerated storage.
typically found to play a role in these high concentration When coupled with automation and low volume analytics,
liabilities. In addition, the limited datasets in combination the resulting data could be used to build various models
with the small number of problematic molecules in those that filter molecules with unfavorable properties from
datasets, have resulted in models that fail to have wide applic­ entering development. Eventually, the use of computational
ability. However, computational models are expected to tools could increase the number of variants that are eval­
improve and eventually help identify high risk molecules uated, further increasing the probability of technical suc­
before they enter development. In the cases where no viable cess. Ultimately, a fundamental understanding of high
alternative exists, computational approaches can provide concentration liabilities will be required, especially as mole­
insight into possible engineering solutions. Two recent reviews cule formats become more complex.
provide an in-depth discussion of some of the recent advances
in computational assessments for developability.92,93
Acknowledgments
The authors would like to thank Jasper Lin and Trevor Swartz for their
Conclusion and future outlook careful review and comments.
With the focus on higher protein concentrations and the
introduction of more complex molecule formats, high concen­ Disclosure statement
tration instabilities have been observed more frequently. If
predictive assessments, whether experimentally or computa­ No potential conflict of interest was reported by the authors.
tionally, are to be applied, additional data to build and validate
models will be required. However, due to the high protein
masses and the labor-intensive process needed to generate Funding
high concentration data, most studies conducted to date have The author(s) reported that there is no funding associated with the work
been carried out at low concentration. For example, the exten­ featured in this article.
sive survey conducted by Jain et al.50 looked at biophysical
properties found in clinical stage and commercial antibodies
with the assumption that antibodies that reached that stage of Acronyms
clinical trials have characteristics amenable to therapeutic A3D AGGRESCAN3D
development. However, most of those molecules were not AC-SINS affinity-capture SINS
developed at high concentration and thus the high concentra­ API active pharmaceutical ingredient
tion data do not exist. It is possible that, as the protein con­ AS ammonium sulfate
AUC analytical ultracentrifugation
centration of those molecules increases, additional factors AuNP gold nanoparticles
relevant to developability will be uncovered while documented BLI biolayer interferometry
thresholds for each property will need recalibrating. CDR Complementarity-determining regions
Building a deeper understanding of high concentration CFD computational fluid dynamics
liabilities will require systematic data of molecules at high CG coarse grained
CIC cross-interaction chromatography
concentration. While automation can help decrease the experi­ CS-SINS charge stabilized SINS
mental burden, many limitations exist, including the high DI developability index
protein masses required by the standard analytics. DLS dynamic light scattering
Identification of low volume high-throughput analytics could DS drug substance
decrease this burden, but additional work is required to estab­ DP drug product
EN elastic network
lish correlations between low mass, high-throughput assay Fv Fragment variable
results, and high concentration aggregation. As such, the kD diffusion interaction parameter
need for large amounts of protein remains. In addition, auto­ MALS multi-angle light scattering
mation of the sample preparation becomes difficult due to the MAM multi-attribute monitoring
increase in viscosity that is often observed at higher protein MD molecular dynamics
ML machine learning
concentrations, and sample-to-sample variation in viscosity MP mass photometry
adds to the complexity. The move to novel formats also adds MS mass spectrometry
complexity to the process of developing high concentration PEG polyethylene glycol
10 J. ZARZAR ET AL.

PEG-SINS PEG-stabilized SINS 13. Das TK, Narhi LO, Sreedhara A, Menzen T, Grapentin C,
PLL poly-L-lysine Chou DK, Antochshuk V, Filipe V. Stress factors in mab drug
PPI Protein-protein interactions substance production processes: critical assessment of impact on
QSAR quantitative structure-activity relationship product quality and control strategy. J Pharm Sci. 2020;109
SAP spatial aggregation propensity (1):116–33. doi:10.1016/j.xphs.2019.09.023.
SASA solvent accessible surface area 14. Lazar KL, Patapoff TW, Sharma VK. Cold denaturation of monoclo­
SCM spatial charge map nal antibodies. MAbs. 2010;2(1):42–52. doi:10.4161/mabs.2.1.10787.
SEC size exclusion chromatography 15. Singh SK, Kolhe P, Mehta AP, Chico SC, Lary AL, Huang M.
SI-BLI self-interaction biolayer interferometry Frozen state storage instability of a monoclonal antibody: aggrega­
SIC self-interaction chromatography tion as a consequence of trehalose crystallization and protein
SINS self-interaction nanoparticle spectroscopy unfolding. Pharmaceut Res. 2011;28(4):873–85. doi:10.1007/
SLS static light scattering s11095-010-0343-z.
SMAC standup mono-layer adsorption chromatography 16. Connolly BD, Le L, Patapoff TW, Cromwell MEM, Moore JMR,
SVM support vector machine Lam P. Protein aggregation in frozen trehalose formulations:
SvP subvisible particle effects of composition, cooling rate, and storage temperature.
TAP therapeutic antibody profiler J Pharm Sci. 2015;104(12):4170–84. doi:10.1002/jps.24646.
TFF tangential flow filtration 17. Bluemel O, Anuschek M, Buecheler JW, Hoelzl G, Bechtold-
Tg’ glass transition temperature Peters K, Friess W. The effect of mAb and excipient cryoconcen­
UFDF ultrafiltration diafiltration tration on long-term frozen storage stability – Part 1: higher
UHMR ultra-high-mass-range molecular weight species and subvisible particle formation.
Int J Pharm X. 2022;4:100108. doi:10.1016/j.ijpx.2021.100108.
18. Bluemel O, Buecheler JW, Hauptmann A, Hoelzl G, Bechtold-
Peters K, Friess W. The effect of mAb and excipient cryoconcen­
References
tration on long-term frozen storage stability – part 2: aggregate
1. Kaplon H, Chenoweth A, Crescioli S, Reichert JDSM. Antibodies formation and oxidation. Int J Pharm X. 2022;4:100109. doi:10.
to watch in 2022. MAbs. 2022;14(1):2014296. doi:10.1080/ 1016/j.ijpx.2021.100109.
19420862.2021.2014296. 19. Bluemel O, Pavlišič A, Likozar B, Rodrigues MA, Geraldes V,
2. The antibody society. Therapeutic monoclonal antibodies Bechtold-Peters K, Friess W. Computational fluid dynamic simu­
approved or in regulatory review. [cited 2023 May 1]; Available lations of temperature, cryoconcentration, and stress time during
from: https://www.antibodysociety.org/antibody-therapeutics- large-scale freezing and thawing of monoclonal antibody
product-data/ solutions. Eur J Pharm Biopharm. 2022;177:107–12. doi:10.1016/
3. Shire SJ, Shahrokh Z, Liu J. Challenges in the development of high j.ejpb.2022.06.007.
protein concentration formulations. J Pharm Sci. 2004;93 20. Salnikova M, Varshney D, Shalaev E. Heterogeneity of Protein
(6):1390–402. doi:10.1002/jps.20079. Environments in Frozen Solutions and in the Dried State. In:
4. Sharma VK, Biobetters KR. Molecular Assessment of Monoclonal Varshney D, Singh M, editors. Lyophilized Biologics and
Antibody-Based Therapeutics Enabling Lead Selection for Clinical Vaccines. NY: Springer New York; 2015. p. 11–24.
Development. In: Rosenberg A, Demeule B, editors. Biobetters: 21. Authelin J-R, Rodrigues MA, Tchessalov S, Singh SK, McCoy T,
Protein Engineering to Approach the Curative. AAPS Advances in Wang S, Shalaev E. Freezing of biologicals revisited: scale, stability,
the Pharmaceutical Sciences Series. NY: Springer New York; 2015. p. excipients, and degradation stresses. J Pharm Sci. 2020;109
153–80. (1):44–61. doi:10.1016/j.xphs.2019.10.062.
5. Jarasch A, Koll H, Regula JT, Bader M, Papadimitriou A, 22. Borwankar AU, Dear BJ, Twu A, Hung JJ, Dinin AK, Wilson BK,
Kettenberger H. Developability assessment during the selection Yue J, Maynard JA, Truskett TM, Johnston KP. Viscosity reduc­
of novel therapeutic antibodies. J Pharm Sci. 2015;104 tion of a concentrated monoclonal antibody with arginine·HCl
(6):1885–98. doi:10.1002/jps.24430. and arginine·Glutamate. Ind Eng Chem Res. 2016;55
6. Xu Y, Wang D, Mason B, Rossomando T, Li N, Liu D, Cheung JK, (43):11225–34. doi:10.1021/acs.iecr.6b02042.
Xu W, Raghava S, Katiyar A, et al. Structure, heterogeneity and 23. Lilyestrom WG, Yadav S, Shire SJ, Scherer TM. Monoclonal anti­
developability assessment of therapeutic antibodies. MAbs. body self-association, cluster formation, and rheology at high
2019;11(2):239–64. doi:10.1080/19420862.2018.1553476. concentrations. J Phys Chem B. 2013;117(21):6373–84. doi:10.
7. Chennamsetty N, Voynov V, Kayser V, Helk B, Trout BL. Design 1021/jp4008152.
of therapeutic proteins with enhanced stability. Proc National 24. Zeng Y, Tran T, Wuthrich P, Naik S, Davagnino J, Greene DG,
Acad Sci. 2009;106(29):11937–42. doi:10.1073/pnas.0904191106. Mahoney RP, Soane DS. Caffeine as a viscosity reducer for highly
8. Bessa J, Boeckle S, Beck H, Buckel T, Schlicht S, Ebeling M, concentrated monoclonal antibody solutions. J Pharm Sci.
Kiialainen A, Koulov A, Boll B, Weiser T, et al. The immunogenicity 2021;110(11):3594–604. doi:10.1016/j.xphs.2021.06.030.
of antibody aggregates in a novel transgenic mouse model. Pharmaceut 25. Banik N, Braun S, Brandenburg JG, Fricker G, Kalonia DS,
Res. 2015;32(7):2344–59. doi:10.1007/s11095-015-1627-0. Rosenkranz T. Technology development to evaluate the effective­
9. Roberts CJ. Protein aggregation and its impact on product quality. ness of viscosity reducing excipients. Int J Pharmaceut.
Curr Opin Biotech. 2014;30:211–17. doi:10.1016/j.copbio.2014.08. 2022;626:122204. doi:10.1016/j.ijpharm.2022.122204.
001. 26. Hung JJ, Dear BJ, Dinin AK, Borwankar AU, Mehta SK, Truskett TT,
10. Roberts CJ. Therapeutic protein aggregation: mechanisms, design, Johnston KP. Improving viscosity and stability of a highly concen­
and control. Trends Biotechnol. 2014;32(7):372–80. doi:10.1016/j. trated monoclonal antibody solution with concentrated proline.
tibtech.2014.05.005. Pharmaceut Res. 2018;35(7):133. doi:10.1007/s11095-018-2398-1.
11. Mahler H, Friess W, Grauschopf U, Kiese S. Protein aggregation: 27. Dear BJ, Hung JJ, Truskett TM, Johnston KP. Contrasting the
pathways, induction factors and analysis. J Pharm Sci. 2009;98 influence of cationic amino acids on the viscosity and stability of
(9):2909–34. doi:10.1002/jps.21566. a highly concentrated monoclonal antibody. Pharmaceut Res.
12. Bunc M, Hadzi S, Graf C, Boncina M, Lah J. Aggregation time 2017;34(1):193–207. doi:10.1007/s11095-016-2055-5.
machine: a platform for the prediction and optimization of 28. Dear BJ, Hung JJ, Laber JR, Wilks LR, Sharma A, Truskett TM,
long-term antibody stability using short-term kinetic analysis. J Med Johnston KP. Enhancing stability and reducing viscosity of
a monoclonal antibody with cosolutes by weakening
Chem. 2022;65(3):2623–32. doi:10.1021/acs.jmedchem.1c02010.
MABS 11

protein-protein interactions. J Pharm Sci. 2019;108(8):2517–26. identify the stability of new antibody therapeutics. Eur J Pharm
doi:10.1016/j.xphs.2019.03.008. Biopharm. 2018;133:131–37. doi:10.1016/j.ejpb.2018.10.009.
29. Johnston KP, Maynard JA, Truskett TM, Borwankar AU, 45. Jacobs SA, Wu S-J, Feng Y, Bethea D, O’Neil KT. Cross-interaction
Miller MA, Wilson BK, Dinin AK, Khan TA, Kaczorowski KJ. chromatography: a rapid method to identify highly soluble mono­
Concentrated dispersions of equilibrium protein nanoclusters clonal antibody candidates. Pharmaceut Res. 2009;27(1):65. doi:10.
that reversibly dissociate into active monomers. Acs Nano. 1007/s11095-009-0007-z.
2012;6(2):1357–69. doi:10.1021/nn204166z. 46. Curtis RA, Ulrich J, Montaser A, Prausnitz JM, Blanch HW.
30. Miller MA, Khan TA, Kaczorowski KJ, Wilson BK, Dinin AK, Protein–protein interactions in concentrated electrolyte solutions.
Borwankar AU, Rodrigues MA, Truskett TM, Johnston KP, Biotechnol Bioeng. 2002;79(4):367–80. doi:10.1002/bit.10342.
Maynard JA. Antibody nanoparticle dispersions formed with mix­ 47. Toprani VM, Joshi SB, Kueltzo LA, Schwartz RM, Middaugh CR,
tures of crowding molecules retain activity and in vivo bioavailability. Volkin DB. A micro–polyethylene glycol precipitation assay as
J Pharm Sci. 2012;101(10):3763–78. doi:10.1002/jps.23256. a relative solubility screening tool for monoclonal antibody design
31. Laue T. Proximity energies: a framework for understanding con­ and formulation development. J Pharm Sci. 2016;105(8):2319–27.
centrated solutions. J Mol Recognit. 2012;25(3):165–73. doi:10. doi:10.1016/j.xphs.2016.05.021.
1002/jmr.2179. 48. Scannell MJ, Hyatt MW, Budyak IL, Woldeyes MA, Wang Y.
32. Saito S, Hasegawa J, Kobayashi N, Kishi N, Uchiyama S, Fukui K. Revisit PEG-Induced precipitation assay for protein solubility
Behavior of monoclonal antibodies: relation between the second assessment of monoclonal antibody formulations. Pharmaceut
virial coefficient (B2) at low concentrations and aggregation pro­ Res. 2021;38(11):1947–60. doi:10.1007/s11095-021-03119-4.
pensity and viscosity at high concentrations. Pharmaceut Res. 49. Chai Q, Shih J, Weldon C, Phan S, Jones BE. Development of a
2012;29(2):397–410. doi:10.1007/s11095-011-0563-x. high-throughput solubility screening assay for use in antibody
33. Connolly BD, Petry C, Yadav S, Demeule B, Ciaccio N, discovery. MAbs. 2019;11(4):747–56. doi:10.1080/19420862.2019.
Moore JMR, Shire SJ, Gokarn YR. Weak interactions govern the 1589851.
viscosity of concentrated antibody solutions: high-throughput 50. Jain T, Sun T, Durand S, Hall A, Houston NR, Nett JH, Sharkey B,
analysis using the diffusion interaction parameter. Biophys J. Bobrowicz B, Caffry I, Yu Y, et al. Biophysical properties of the
2012;103(1):69–78. doi:10.1016/j.bpj.2012.04.047. clinical-stage antibody landscape. Proc National Acad Sci.
34. Kingsbury JS, Saini A, Auclair SM, Fu L, Lantz MM, Halloran KT, 2017;114(5):944–49. doi:10.1073/pnas.1616408114.
Calero-Rubio C, Schwenger W, Airiau CY, Zhang J, et al. A single 51. Schuster J, Koulov A, Mahler H-C, Detampel P, Huwyler J,
molecular descriptor to predict solution behavior of therapeutic Singh S, Mathaes R. In Vivo stability of therapeutic proteins.
antibodies. Sci Adv. 2020;6(32):eabb0372. doi:10.1126/sciadv.abb0372. Pharmaceut Res. 2020;37(2):23. doi:10.1007/s11095-019-2689-1.
35. Ghosh SK, Pal T. Interparticle coupling effect on the surface 52. den BM, Lai S-H, Xue X, van KM, Bleijlevens B, Heck AJR.
plasmon resonance of gold nanoparticles: from theory to Comparative analysis of antibodies and heavily glycosylated
applications. Chem Rev. 2007;107(11):4797–862. doi:10.1021/ macromolecular immune complexes by size-exclusion chromato­
cr0680282. graphy multi-angle light scattering, native charge detection mass
36. Sule SV, Dickinson CD, Lu J, Chow C-K, Tessier PM. Rapid spectrometry, and mass photometry. Anal Chem. 2022;94
analysis of antibody self-association in complex mixtures using (2):892–900. doi:10.1021/acs.analchem.1c03656.
immunogold conjugates. Mol Pharmaceut. 2013;10(4):1322–31. 53. Füssl F, Strasser L, Cari S, Bones J. Native LC–MS for capturing
doi:10.1021/mp300524x. quality attributes of biopharmaceuticals on the intact protein
37. Liu Y, Caffry I, Wu J, Geng SB, Jain T, Sun T, Reid F, Cao Y, Estep P, level. Curr Opin Biotech. 2021;71:32–40. doi:10.1016/j.copbio.
Yu Y, et al. High-throughput screening for developability during 2021.05.008.
early-stage antibody discovery using self-interaction nanoparticle 54. Asor R, Kukura P. Characterising biomolecular interactions and
spectroscopy. MAbs. 2013;6(2):483–92. doi:10.4161/mabs.27431. dynamics with mass photometry. Curr Opin Chem Biol.
38. Phan S, Walmer A, Shaw EW, Chai Q. High-throughput profiling 2022;68:102132. doi:10.1016/j.cbpa.2022.102132.
of antibody self-association in multiple formulation conditions by 55. den EJ, Garidel P, Smulders R, Koll H, Smith B, Bassarab S, Seidl A,
PEG stabilized self-interaction nanoparticle spectroscopy. MAbs. Hainzl O, Jiskoot W. Strategies for the assessment of protein
2022;14(1):2094750. doi:10.1080/19420862.2022.2094750. aggregates in pharmaceutical biotech product development.
39. Starr CG, Makowski EK, Wu L, Berg B, Kingsbury JS, Gokarn YR, Pharmaceut Res. 2011;28(4):920–33. doi:10.1007/s11095-010-
Tessier PM. Ultradilute measurements of self-association for the 0297-1.
identification of antibodies with favorable high-concentration 56. Beckmann R, Jensen K, Fenn S, Speck J, Krause K, Meier A,
solution properties. Mol Pharmaceut. 2021;18(7):2744–53. doi:10. Röth M, Fauser S, Kimbung R, Logan DT, et al. DutaFabs are
1021/acs.molpharmaceut.1c00280. engineered therapeutic fab fragments that can bind two targets
40. Sun T, Reid F, Liu Y, Cao Y, Estep P, Nauman C, Xu Y. High simultaneously. Nat Commun. 2021;12(1):708. doi:10.1038/
throughput detection of antibody self-interaction by bio-layer s41467-021-20949-3.
interferometry. MAbs. 2013;5(6):838–41. doi:10.4161/mabs.26186. 57. Georges GJ, Dengl S, Bujotzek A, Hesse F, Fischer JAA, Gärtner A,
41. Domnowski M, Jaehrling J, Frieß W. Assessment of antibody Benz J, Lauer ME, Ringler P, Stahlberg H, et al. The contorsbody,
self-interaction by bio-layer-interferometry as a tool for early an antibody format for agonism: design, structure, and function.
stage formulation development. Pharmaceut Res. 2020;37(2):29. Comput Struct Biotechnology J. 2020;18:1210–20. doi:10.1016/j.
doi:10.1007/s11095-019-2722-4. csbj.2020.05.007.
42. Quigley A, Williams DR. The second virial coefficient as 58. Du W, Klibanov AM. Hydrophobic salts markedly diminish visc­
a predictor of protein aggregation propensity: a self-interaction osity of concentrated protein solutions. Biotechnol Bioeng.
chromatography study. Eur J Pharm Biopharm. 2015;96:282–90. 2011;108(3):632–36. doi:10.1002/bit.22983.
doi:10.1016/j.ejpb.2015.07.025. 59. Yadav S, Liu J, Shire SJ, Kalonia DS. Specific interactions in high
43. Tessier PM, Vandrey SD, Berger BW, Pazhianur R, Sandler SI, concentration antibody solutions resulting in high viscosity.
Lenhoff AM. Self‐interaction chromatography: a novel screening J Pharm Sci. 2010;99(3):1152–68. doi:10.1002/jps.21898.
method for rational protein crystallization. Acta Crystallogr Sect 60. Yadav S, Laue TM, Kalonia DS, Singh SN, Shire SJ. The influence
D. 2002;58(10):1531–35. doi:10.1107/S0907444902012775. of charge distribution on self-association and viscosity behavior of
44. Hedberg SHM, Rapley J, Haigh JM, Williams DR. Cross- monoclonal antibody solutions. Mol Pharmaceut. 2012;9
interaction chromatography as a rapid screening technique to (4):791–802. doi:10.1021/mp200566k.
12 J. ZARZAR ET AL.

61. Tomar DS, Singh SK, Li L, Broulidakis MP, Kumar S. In Silico 78. Majumdar R, Esfandiary R, Bishop SM, Samra HS,
prediction of diffusion interaction parameter (kD), a key indicator Middaugh CR, Volkin DB, Weis DD. Correlations between
of antibody solution behaviors. Pharmaceut Res. 2018;35(10):193. changes in conformational dynamics and physical stability in
doi:10.1007/s11095-018-2466-6. a mutant IgG1 mAb engineered for extended serum half-life.
62. Sharma VK, Patapoff TW, Kabakoff B, Pai S, Hilario E, Zhang B, MAbs. 2015;7:84–95.
Li C, Borisov O, Kelley RF, Chorny I, et al. In silico selection of 79. Tomar DS, Li L, Broulidakis MP, Luksha NG, Burns CT, Singh SK,
therapeutic antibodies for development: viscosity, clearance, and Kumar S. In-silico prediction of concentration-dependent viscosity
chemical stability. Proc National Acad Sci. 2014;111(52):18601–06. curves for monoclonal antibody solutions. MAbs. 2017;9:476–89.
doi:10.1073/pnas.1421779112. 80. Tilegenova C, Izadi S, Yin J, Huang CS, Wu J, Ellerman D,
63. Pallarés I, Ventura S. Advances in the prediction of protein aggre­ Hymowitz SG, Walters B, Salisbury C, Carter PJ. Dissecting the
gation propensity. Curr Med Chem. 2019;26(21):3911–20. doi:10. molecular basis of high viscosity of monospecific and bispecific
2174/0929867324666170705121754. IgG antibodies. MAbs. 2020;12:1692764.
64. Conchillo-Solé O, de GN, Avilés FX, Vendrell J, Daura X, 81. Chowdhury A, Bollinger JA, Dear BJ, Cheung JK, Johnston KP,
Ventura S. AGGRESCAN: a server for the prediction and evalua­ Truskett TM. Coarse-grained molecular dynamics simulations for
tion of “hot spots” of aggregation in polypeptides. BMC understanding the impact of short-range anisotropic attractions
Bioinform. 2007;8(1):65. doi:10.1186/1471-2105-8-65. on structure and viscosity of concentrated monoclonal antibody
65. Trovato A, Seno F, Tosatto SCE. The PASTA server for protein solutions. Mol Pharmaceut. 2020;17:1748–56.
aggregation prediction. Protein Eng Des Sel. 2007;20(10):521–23. 82. Chaudhri A, Zarraga IE, Yadav S, Patapoff TW, Shire SJ, Voth GA.
doi:10.1093/protein/gzm042. The role of amino acid sequence in the self-association of ther­
66. Fernandez-Escamilla A-M, Rousseau F, Schymkowitz J, Serrano L. apeutic monoclonal antibodies: insights from coarse-grained
Prediction of sequence-dependent and mutational effects on the modeling. J Phys Chem B. 2013;117:1269–79.
aggregation of peptides and proteins. Nat Biotechnol. 2004;22 83. Izadi S, Patapoff TW, Walters BT. Multiscale coarse-grained
(10):1302–06. doi:10.1038/nbt1012. approach to investigate self-association of antibodies. Biophys J.
67. Tartaglia GG, Vendruscolo M. The zyggregator method for pre­ 2020;118:2741–54.
dicting protein aggregation propensities. Chem Soc Rev. 2008;37 84. Kamerzell TJ, Middaugh CR. Prediction machines: applied
(7):1395–401. doi:10.1039/b706784b. machine learning for therapeutic protein design and
68. Zambrano R, Jamroz M, Szczasiuk A, Pujols J, Kmiecik S, development. J Pharm Sci. 2021;110:665–81.
Ventura S. AGGRESCAN3D (A3D): server for prediction of 85. Lai P-K, Fernando A, Cloutier TK, Gokarn Y, Zhang J,
aggregation properties of protein structures. Nucleic Acids Res. Schwenger W, Chari R, Calero-Rubio C, Trout BL. Machine learn­
2015;43(W1):W306–13. doi:10.1093/nar/gkv359. ing applied to determine the molecular descriptors responsible for
69. Kuriata A, Iglesias V, Pujols J, Kurcinski M, Kmiecik S, Ventura S. the viscosity behavior of concentrated therapeutic antibodies. Mol
Aggrescan3d (A3D) 2.0: prediction and engineering of protein Pharmaceut. 2021;18:1167–75.
solubility. Nucleic Acids Res. 2019;47(W1):W300–7. doi:10.1093/ 86. Lou H, Hageman MJ. Machine learning attempts for predicting
nar/gkz321. human subcutaneous bioavailability of monoclonal antibodies.
70. Agrawal NJ, Helk B, Kumar S, Mody N, Sathish HA, Samra HS, Pharmaceut Res. 2021;38:451–60.
Buck PM, Li L, Trout BL. Computational tool for the early screen­ 87. Lai P-K, Fernando A, Cloutier TK, Kingsbury JS, Gokarn Y,
ing of monoclonal antibodies for their viscosities. MAbs. 2016;8 Halloran KT, Calero-Rubio C, Trout BL. Machine learning feature
(1):43–48. doi:10.1080/19420862.2015.1099773. selection for predicting high concentration therapeutic antibody
71. Voynov V, Chennamsetty N, Kayser V, Helk B, Trout BL. aggregation. J Pharm Sci. 2021;110:1583–91.
88. Lai P-K, Gallegos A, Mody N, Sathish HA, Trout BL. Machine
Predictive tools for stabilization of therapeutic proteins. MAbs.
learning prediction of antibody aggregation and viscosity for high
2009;1(6):580–82. doi:10.4161/mabs.1.6.9773.
concentration formulation development of protein therapeutics.
72. Sankar K, Krystek SR, Carl SM, Day T, JKX M. AggScore: predic­
MAbs. 2022;14:2026208.
tion of aggregation‐prone regions in proteins based on the dis­
89. Jain T, Boland T, Lilov A, Burnina I, Brown M, Xu Y, Vásquez M.
tribution of surface patches. Proteins Struct Funct Bioinform.
Prediction of delayed retention of antibodies in hydrophobic
2018;86(11):1147–56. doi:10.1002/prot.25594.
interaction chromatography from sequence using machine
73. Lauer TM, Agrawal NJ, Chennamsetty N, Egodage K, Helk B,
learning. Bioinformatics. 2017;33:3758–66.
Trout BL. Developability index: a rapid in silico tool for the
90. Lai P-K, Swan JW, Trout BL. Calculation of therapeutic antibody
screening of antibody aggregation propensity. J Pharm Sci.
viscosity with coarse-grained models, hydrodynamic calculations
2012;101(1):102–15. doi:10.1002/jps.22758.
and machine learning-based parameters. MAbs. 2021;13:1907882.
74. Raybould MIJ, Marks C, Krawczyk K, Taddese B, Nowak J,
91. Cloutier TK, Sudrik C, Mody N, Sathish HA, Trout BL. Machine
Lewis AP, Bujotzek A, Shi J, Deane CM. Five computational
learning models of antibody–excipient preferential interactions for
developability guidelines for therapeutic antibody profiling. Proc
use in computational formulation design. Mol Pharmaceut.
National Acad Sci. 2019;116(10):4025–30. doi:10.1073/pnas.
2020;17:3589–99.
1810576116.
92. Khetan R, Curtis R, Deane CM, Hadsund JT, Kar U, Krawczyk K,
75. Li L, Kumar S, Buck PM, Burns C, Lavoie J, Singh SK,
Kuroda D, Robinson SA, Sormanni P, Tsumoto K, et al. Current
Warne NW, Nichols P, Luksha N, Boardman D. Concentration
advances in biopharmaceutical informatics: guidelines, impact and
dependent viscosity of monoclonal antibody solutions: explain­
challenges in the computational developability assessment of anti­
ing experimental behavior in terms of molecular properties.
body therapeutics. MAbs. 2022;14:2020082.
Pharmaceut Res. 2014;31(11):3161–78. doi:10.1007/s11095-014-
93. Kuroda D, Tsumoto K. Engineering stability, viscosity, and immu­
1409-0. nogenicity of antibodies by computational design. J Pharm Sci.
76. Buck PM, Chaudhri A, Kumar S, Singh SK. Highly viscous antibody
2020;109:1631–51.
solutions are a consequence of network formation caused by domain–
94. Spiess C, Zhai Q, Carter PJ. Alternative molecular formats and
domain electrostatic complementarities: insights from coarse-grained therapeutic applications for bispecific antibodies. Mol Immunol.
simulations. Mol Pharmaceut. 2015;12(1):127–39. doi:10.1021/ 2015;67:95–106.
mp500485w. 95. Guan X, Eris T, Zhang L, Ren D, Ricci MS, Thiel T, Goudar CT. A
77. Chaudhri A, Zarraga IE, Kamerzell TJ, Brandt JP, Patapoff TW, high-resolution multi-attribute method for product characteriza­
Shire SJ, Voth GA. Coarse-grained modeling of the self-association tion, process characterization, and quality control of therapeutic
of therapeutic monoclonal antibodies. J Phys Chem B. proteins. Anal Biochem. 2022;643:114575.
2012;116:8045–57.
MABS 13

96. Carvalho SB, Gomes RA, Pfenninger A, Fischer M, Strotbek M, 99. Hedberg SHM, Heng JYY, Williams DR, Liddell JM. Micro scale
Isidro IA, Tugçu N, Gomes-Alves P. Multi attribute method self-interaction chromatography of proteins: a mAb case-study.
implementation using a high resolution mass spectrometry plat­ J Chromatogr A. 2016;1434:57–63.
form: from sample preparation to batch analysis. PLos One. 100. Kinnunen HM, Sharma V, Contreras-Rojas LR, Yu Y, Alleman C,
2022;17:e0262711. Sreedhara A, Fischer S, Khawli L, Yohe ST, Bumbaca D, et al.
97. Hao Z, Moore B, Ren C, Sadek M, Macchi F, Yang L, Harris J, A novel in vitro method to model the fate of subcutaneously
Yee L, Liu E, Tran V, et al. Multi-attribute method performance administered biopharmaceuticals and associated formulation
profile for quality control of monoclonal antibody therapeutics. components. J Control Release. 2015;214:94–102.
J Pharmaceut Biomed. 2021;205:114330. 101. Bown HK, Bonn C, Yohe S, Yadav DB, Patapoff TW, Daugherty A,
98. Woldeyes MA, Josephson LL, Leiske DL, Galush WJ, Roberts CJ, Mrsny RJ. In vitro model for predicting bioavailability of subcuta­
Furst EM. Viscosities and protein interactions of bispecific anti­ neously injected monoclonal antibodies. J Control Release.
bodies and their monospecific mixtures. Mol Pharmaceut. 2018;273:13–20.
2018;15:4745–55.
TYPE Review
PUBLISHED 18 September 2023
DOI 10.3389/fphar.2023.1274088

A comprehensive overview on
OPEN ACCESS antibody-drug conjugates: from
EDITED BY
Ayaz Shahid,
Western University of Health Sciences,
the conceptualization to cancer
United States

REVIEWED BY
therapy
Jifa Zhang,
Sutro Biopharma, Inc., United States
Marco Cavaco,
Federico Riccardi 1, Michele Dal Bo 1, Paolo Macor 2* and
Universidade de Lisboa, Portugal Giuseppe Toffoli 1
*CORRESPONDENCE 1
Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico (CRO), IRCCS, Aviano,
Paolo Macor,
Italy, 2Department of Life Sciences, University of Trieste, Trieste, Italy
pmacor@units.it

RECEIVED 07 August 2023


ACCEPTED 07 September 2023
PUBLISHED 18 September 2023
Antibody-Drug Conjugates (ADCs) represent an innovative class of potent anti-
CITATION
Riccardi F, Dal Bo M, Macor P and cancer compounds that are widely used in the treatment of hematologic
Toffoli G (2023), A comprehensive malignancies and solid tumors. Unlike conventional chemotherapeutic drug-
overview on antibody-drug conjugates:
based therapies, that are mainly associated with modest specificity and
from the conceptualization to
cancer therapy. therapeutic benefit, the three key components that form an ADC (a
Front. Pharmacol. 14:1274088. monoclonal antibody bound to a cytotoxic drug via a chemical linker moiety)
doi: 10.3389/fphar.2023.1274088
achieve remarkable improvement in terms of targeted killing of cancer cells and,
COPYRIGHT while sparing healthy tissues, a reduction in systemic side effects caused by off-
© 2023 Riccardi, Dal Bo, Macor and
tumor toxicity. Based on their beneficial mechanism of action, 15 ADCs have been
Toffoli. This is an open-access article
distributed under the terms of the approved to date by the market approval by the Food and Drug Administration
Creative Commons Attribution License (FDA), the European Medicines Agency (EMA) and/or other international
(CC BY). The use, distribution or
governmental agencies for use in clinical oncology, and hundreds are
reproduction in other forums is
permitted, provided the original author(s) undergoing evaluation in the preclinical and clinical phases. Here, our aim is to
and the copyright owner(s) are credited provide a comprehensive overview of the key features revolving around ADC
and that the original publication in this
therapeutic strategy including their structural and targeting properties,
journal is cited, in accordance with
accepted academic practice. No use, mechanism of action, the role of the tumor microenvironment and review the
distribution or reproduction is permitted approved ADCs in clinical oncology, providing discussion regarding their toxicity
which does not comply with these terms.
profile, clinical manifestations and use in novel combination therapies. Finally, we
briefly review ADCs in other pathological contexts and provide key information
regarding ADC manufacturing and analytical characterization.

KEYWORDS

antibody-drug conjugate, antibodies, linkers, payloads, target therapy, cancer treatment

1 Introduction
Cancer is a multi-stage progression that transforms normal healthy cells into
malignant lesions (Cooper, 2000). It is the leading cause of death worldwide,
accounting for nearly 10 million deaths in 2020, and one of the most challenging
diseases to treat (Sung et al., 2021). Conventional therapeutic strategies are grouped
into chemotherapy, radiotherapy, immunotherapy and surgery, with the former being the
main approach for treating various cancers (Dan et al., 2018). Although most remarkable
goals have been achieved with small cytotoxic drugs, they had several drawbacks that
limited their efficacy, including a low therapeutic index and a high off-tumor effect
(Senapati et al., 2018). Usually, the low effectiveness of chemotherapy provoked a high
incidence of severe side effects in patients, mainly caused by the non-specific action of

Frontiers in Pharmacology 01 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

chemotherapeutic drugs on rapidly dividing normal cells (Senapati


et al., 2018). Therefore, one of the hot topics in the field concerns
the discovery of new chemical agents with enhanced therapeutic
efficacy and that preferentially ablate tumor-derived cells, without
harming the body itself (Casi and Neri, 2012). In the field of
immunotherapy, several monoclonal antibodies (mAbs) have been
clinically approved as they showed therapeutic benefits in both
hematologic malignancies and solid tumors by selectively targeting
cancer cells and by activating direct and/or indirect killing
mechanisms (Weiner et al., 2009; Kimiz-Gebologlu et al., 2018;
Castelli et al., 2019). However, issues including tissue accessibility,
poor pharmacokinetics and lame interactions with the immune
system have led to the need to exploit newer, safer and more
effective targeted therapies (Chames et al., 2009; Talotta et al.,
2019). In 1907, German nobel laureate Paul Erlich postulated that
there could exist compounds that would selectively target
pathogenic microbes, such as bacteria, while sparing normal
cells, a concept gone down in history as “zauberkugel” (magic FIGURE 1
bullet) (Schwartz, 2004). From his hypothesis, an innovative Schematic representation of the modular components of an
Antibody-Drug Conjugate (ADC). ADC consists of a monoclonal
therapeutic approach, known as Antibody-Drug Conjugate antibody (blue), a linker (blue lines) and the cytotoxic drugs (gray/red).
(ADC), has arisen in the oncology field and was firstly 40 used In this picture, the representative ADC on the left has a Drug-to-
years ago to treat patients with advanced cancer (Ford et al., 1983). Antibody ratio equal to 4. A brief description of the basic function of
each module is indicated on the right.
ADCs are an emerging class of pharmacological compounds that
combine the potency of anti-cancer drugs (often called payloads)
with the specificity of mAbs to the tumor site, thus combining
chemotherapy and immunotherapy. They are composed of three 2 ADCs consist of antibodies linked to
portions, a mAb, an organic spacer and a cytotoxic drug. Ideally, cytotoxic payloads via a linker
they use the mAb targeting ability to take the cytotoxic agent,
which is bound to the mAb via a stable linker, to the cancerous cells 2.1 Antibodies form the scaffold that guides
or to the cellular components of the tumor microenvironment ADCs to target cells
(TME) where it can exert its anti-tumor activity and lead to cell
death (Baah et al., 2021; Mckertish and Kayser, 2021; Fu et al., An ADC is a synthetic molecule with pharmacological
2022). Compared with standard chemotherapy, this strategy offers activity comprising three blocks: a selective mAb, a stable
several advantages including better drug tolerability, cytotoxicity linker, and a potent cytotoxic drug (Figure 1). Together, they
even at low concentrations, drug stability in the bloodstream and ensure tumor-specific targeting and efficient ablation of
in lysosomes, reduced off-target effects, and systemic toxicity, all malignant cells by creating a “new” compound with enhanced
features that contribute to the expansion of its therapeutic therapeutic efficacy.
potential (Khongorzul et al., 2020; Drago et al., 2021; Mckertish
and Kayser, 2021; Fu et al., 2022). Beginning with the first ADC, 2.1.1 Full size antibodies are the most used as ADCs
which was approved by the Food and Drug Administration (FDA) scaffolds
in 2000 (Norsworthy et al., 2018), and given the ever-evolving An antibody (Ab) or immunoglobulin (Ig) is a Y-shaped
technology of mAbs, linkers, and payloads, by April glycoprotein produced by plasma cells that presents an intrinsic
2023 13 different ADCs have been FDA-approved for clinical selective ability to bind to its target (Baah et al., 2021; Pettinato,
use for both solid and hematologic malignancies, setting the 2021). Several Abs used in oncology, upon interaction with their
stage for a new era of targeted cancer therapy (Dumontet et al., target antigens, possess the capacity to influence the biological
2023). In addition, a few studies have already addressed the activity of the tumor mass by modulating survival-related
potential use of ADCs in non-oncologic context, including pathways and/or activating potent immune effector functions
infections (O’Leary et al., 2023; Tvilum et al., 2023) and auto- through three main mechanisms: antibody-dependent cell-
immune disorders (Lee et al., 2017; Yasunaga et al., 2017), with mediated cytotoxicity (ADCC), in which the Fc portion of
promising results showing the possibility of expanding the use of bound Ab is recognized by Natural Killer (NK) cell Fc-
ADCs in various diseases. In this review, we aim to summarize the receptor and activates the release of lytic factors, complement-
current knowledge of ADCs and address some key points about dependent cytotoxicity (CDC), in which the interaction between
their molecular properties, their interaction with tumor mass and the Fc region and C1q triggers the classic pathway of the
TME, their clinical use, toxicities and combinate regimens in complement system leading to cell lysis (Macor and Tedesco,
cancer treatment. Finally, we will provide an overview on 2007; Macor et al., 2018) and antibody-dependent cellular
current research on ADCs in other diseases and address the phagocytosis (ADCP), a mechanism that relies on active
main challenges and limitations in their production and macrophages to engulf tumor cells (Peters and Brown, 2015;
characterization. Chen et al., 2020; Vozella et al., 2021). Similarly, tumor-targeting

Frontiers in Pharmacology 02 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Abs in ADCs shall be monoclonal and ensure high target cellular signatures of the mass (Deonarain et al., 2018;
specificity and binding affinity, long half-life in plasma, Richards, 2018). Other problems related to scaffold size
minimal immunogenicity combined with low cross-reactivity, include systemic accumulation and slowed target-independent
and allow efficient internalization as well as induce direct/ clearance rate (Adams et al., 2001; Jain, 2001; Thurber and Dane
indirect killing effects (Peters and Brown, 2015; Dean et al., Wittrup, 2012). Because of these limitations, researchers are
2021; Liu and Chen, 2022). According to the amino acid seeking new “miniaturized” versions of natural antibodies
sequence of their heavy chain constant regions, human Igs are (also known as antibody-fragments) as a new and smaller
classified into 5 isotypes or classes (IgM, IgG, IgA, IgE, and IgD), drug-conjugatable alternative to expand the ADC therapeutic
with IgG being the most abundant in serum. Based on further benefits. These fragments are produced either by proteolytic
amino acid variations, this isotype can be subdivided into cleavage of full-size antibodies or by recombinant protein
4 subtypes (IgG1, IgG2, IgG3, and IgG4). IgG1 consists of the engineering and primarily retain the binding capacity of full-
variable heavy (VH) and light (VL) domains in the N-terminal size IgG through the VH and VL regions (Brinkmann and
portion of the antibody, the C-domains of the light chains Kontermann, 2017; Kholodenko et al., 2019). They present
constant region (CL) and the heavy chain constant regions engineered scaffolds that lack the CH2 domain and Fc region
(CH1, CH2, CH3) and the hinge region between the CH1 and and include three different formulations: the Fab format, a
CH2 domains (Chiu et al., 2019). To date, its backbone is the ~50 kDa structure in which VL and VH are bound to CL and
most used in ADC preparations because of its serum half-life CH1, respectively, and linked by a disulfide bond between the
(~21 days), high Fcγ receptors avidity, thereby having strong chains, the single-chain variable fragment (scFv), a ~27 kDa
immune activation of Fc-dependent pathways, and more potent structure in which VH is linked to VL by a short peptide
complement activation (Yu et al., 2020). Beside these indirect linker, and the diabody, a non-covalent ~55 kDa dimer scFv
cytotoxic mechanisms, upon the interaction with specific consisting of the VH and VL regions linked by a small peptide
antigens on malignant cells, IgG1-based mAb can also exert linker (Xenaki et al., 2017; Deonarain et al., 2018; Kholodenko
direct killing effects by blocking pathways associated to cancer et al., 2019). In addition, small immunoproteins (SIPs) composed
cell proliferation, metastasis and invasiveness (Natsume et al., of dimerized scFvs through a CHε4 domain are a fragmentated
2009; Drago et al., 2021). As for the other subclasses, IgG2 plays a format developed against fibronectin and other vascular antigens
role in the response against bacterial capsular polysaccharides (Perrino et al., 2014). By preserving the targeting capacity of the
but exhibits low Fcγ receptor avidity, low plasma concentration full-size antibody and combining it with smaller and dynamic
and tends to form covalent dimers that likely lead to aggregation formats, antibody-fragments have the potential to overcome
and ADC inefficacy (Zhang et al., 2018). IgG3 protects the body some of the major drawbacks of full-size Ig moieties and may
from a range of intracellular bacteria, parasites, and viruses. They represent an innovation in the treatment of solid tumors. To date,
are potent mediators of effector functions, including enhanced promising data showed a remarkable improvement in stability,
ADCC and CDC responses, but they are also the subtype with the tumor targeting and penetration and epitope accessibility,
shortest circulating half-life (Stapleton et al., 2011; Hoffmann particularly in cancers that are still difficult to reach via
et al., 2018), limiting their suitability for ADCs. The IgG4 subtype conventional IgG-based ADCs (Dennis et al., 2007; Thurber
has a similar half-life to IgG1 and IgG2 but is less efficient at et al., 2008). In addition, smaller formats should be better
triggering the C1q-related pathway because it has only tolerated by the body and produce fewer adverse effects
intermediate affinity for the Fc receptor on phagocytic cells mainly because they do not show cross-reactivity caused by
(Spiess et al., 2013; Fu et al., 2022). However, the ability to the interaction with Fc receptor and targeted immune cells
induce a poor immune response results in a more favorable (Cilliers et al., 2018; Gogesch et al., 2021). If these benefits
safety profile compared to the IgG1 backbone, making should be assessed to a greater extent, significant weaknesses
IgG4 ADCs suitable in cases where antibody-mediated can be identified. For example, because of their smaller size and
cytotoxicity is not desired (Herbener et al., 2018). the absence of the Fc domain, without which they cannot trigger
the neonatal Fc receptor rescue pathway, antibody-fragments are
2.1.2 Antibody-fragments represent a new and degraded more rapidly and potentially do not remain in the body
innovative approach in ADC strategy long enough to exert sufficient anti-tumor activity. From this
A growing number of studies raises questions regarding the perspective, one solution might be to control the dosing regimen
usage of full-size IgG moiety in the treatment of solid cancers and by administering higher and/or more frequent doses to achieve a
points to some critical limitations, particularly on its size therapeutic effect, but too little is known about the behavior of
(Deonarain et al., 2018; Richards, 2018). As IgG1, which is the these formats in the clinic and extensive efforts are underway to
most used in ADC synthesis, is quite large (150 kDa) it may hinder improve their feasibility (Deonarain et al., 2018; Deonarain and
the distribution of the drug in the tumor mass and consequently Xue, 2020). Moreover, the absence of the Fc domain severely
affect in a negative manner ADC pharmacokinetics and the limits the possibility of activating the immune system, thus losing
therapeutic outcome. Although this issue may find a partial an important partner in the fight against the tumor progression.
solution in the tumor-associated leaky vasculature, which shall Besides these formulations, scaffolds not based on antibodies are
still allow sufficient pharmacological benefit due to the retention currently being explored (Luo et al., 2022; Kaplon et al., 2023).
and permeability effect (EPR) (Ferl et al., 2006), its efficiency and Nevertheless, though the Ab moiety properties must be carefully
the microdistribution of the compound in the tumor depend on considered, the choice of linker and payload are equally
many factors, such as ADC preparations and molecular and important to create the most suitable therapeutic ADC.

Frontiers in Pharmacology 03 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

2.2 Linkers are sequences that connect 2.2.2 Non-cleavable linkers avoid non-specific
antibodies to payloads by a chemical bond payloads release
Non-cleavable linkers include maleimidocaproyl (MC) and 4-
In the development of the ADC strategy, linkers represent the maleimidomethyl cyclohexane-1-carboxylate (MCC) structures and
technology that ensures a bridge between the mAb and the consist of chemical structures that are not fragmented by enzymatic
cytotoxic payload. They are the most modifiable part of an degradation. They are inert to conventional chemicals but allow the
ADC and influence its biophysical and functional properties release of the cytotoxic drug once the mAb has been completely
such as stability, potency, efficacy, and toxicity. The two main catabolized by the lysosome. In this way, they release their toxic
purposes of linkers are to prevent the premature release of the payload into the tumor target cells without harming normal healthy
cytotoxic drug in the blood circulation and to ensure its efficient cells. Due to their chemical synthesis, these linkers offer some
release at the target site (Lu et al., 2016; Bargh et al., 2019). advantages over the cleavable alternative, including lower toxicity
Depending on the release mechanisms of the payloads, linkers and longer half-life in plasma (Baah et al., 2021; Fu et al., 2022). On
can be broadly classified into two classes: cleavable and non- the other hand, their limitations are mainly related to their
cleavable. Recent advances in linker chemistry, including mechanism of action as they are strongly dependent on an
formulations not currently used in clinical ADCs, such as efficient intracellular trafficking and on cellular components with
biorthogonal, photo-responsive and Fe(II)-cleavable linkers, high-expression and internalizing antigens (Lu et al., 2016;
are discussed in Su et al., 2021. Tsuchikama and An, 2018).

2.2.1 Cleavable linkers are versatile and widely


employed in ADCs 2.3 Payloads consist of potent cytotoxic
Cleavable linkers are most commonly used in the synthesis of agents against tumor cells
ADCs and are designed to disengage the cytotoxic drug in response
to changes in environmental conditions (pH, redox potential, GSH As described above, the linker serves as a spacer to connect the
concentration, or enzymatic activity) that occur between the mAb to the payload, a cytotoxic drug that must be released in the
bloodstream, the tumor cells and the TME niche. They are stable tumor site to properly exert its pharmacological effects. To be
under physiological conditions and, following the internalization of suitable as payloads in ADCs, chemicals shall ideally have low
the ADC into the tumor cell, they are rapidly cleaved to ensure molecular weight and immunogenicity, high stability in the blood
selective release of the cytotoxic preparation (Peters and Brown, circulation and endosomal/lysosomal pathways, and high
2015; Tsuchikama and An, 2018). In addition, these linkers are often cytotoxicity (Peters and Brown, 2015; Khongorzul et al., 2020;
cleaved in the TME because of its higher acidity and oxidative stress, Baah et al., 2021; Mckertish and Kayser, 2021). Because
making them the most used preparation to affect large solid masses intravenous administration has shown that only a very small
barely impenetrable to full-size antibodies (Ponziani et al., 2020). fraction of ADC reaches the tumor (0.1%–2%) (Chari, 2008;
Cleavable linkers are commonly divided into chemical (hydrazone Hughes, 2010; Beck et al., 2017), their payloads must be 100- to
and disulfide bonds) and enzyme (peptide bonds and glucuronide) 1000-fold more effective than the drugs used in chemotherapeutics
cleavage linkers (Bargh et al., 2019). Hydrazone linkers are an as free small molecules (Baah et al., 2021). Given that the goal of the
example of acid-labile linkers used mainly in hematologic ADC strategy is to achieve potent cytotoxic activity, an important
malignancies. They are usually stable in the physiological attribute to consider in the design process of these compounds is the
pH range of the blood circulation and undergo hydrolysis within Drug-to-antibody ratio (DAR), a value that indicates the average
the acidic microenvironment of the endosomes and lysosomes number of chemical molecules conjugated to the mAb. For current
(pH 4.8–6.2) of the tumor cell (Jain et al., 2015). Similarly, conjugation methods based on lysine side-chain amidation or
linkers based on disulfide bonds are stable in the bloodstream mainly on the reduction of cysteine intermediate-chain disulfide
alkaline environment, but payload release is sensitive to bonds, the common DAR values range from 0 (lowest value) to 8
glutathione (GSH), a metabolite whose concentration is much (highest value) (Dan et al., 2018; Wagh et al., 2018; Sun Kang et al.,
higher in the cytoplasm of cancer cells (Balendiran et al., 2004; 2021). Nevertheless, in vivo experiments have demonstrated that a
Estrela et al., 2006). However, both linkers raised concerns about high DAR value negatively correlates with Ab pharmacokinetics.
their non-targeted cytotoxicity (Baah et al., 2021; Fu et al., 2022). Although a low DAR implies the loading of poor number of drug
Peptide bonds ensure that ADC remains integral in the circulation molecules and consequently lower therapeutic efficacy, it is worth
and enable the release of the cytotoxic drug upon interaction with noting that an average DAR of 2 to 4 results in an ADC with greater
lysosomal proteases (Tsuchikama and An, 2018), such as cathepsin anti-cancer activity/efficacy compared to an ADC with higher DAR,
B, which is generally overexpressed in several tumor cell types likely because the latter is more rapidly cleared from the body when
(Gondi and Rao, 2013). Peptide linkers are associated with compared to the corresponding average-conjugated counterparts
improved serum stability and anticancer activity compared (Hamblett et al., 2004; Strop et al., 2015). Nowadays, cytotoxic
chemical linkers (Lu et al., 2016; Tang et al., 2019; Khongorzul payloads usually act as DNA-damaging agents or tubulin inhibitors,
et al., 2020). In addition, linkers based on glucuronide bonds, but novel potential drugs under investigation include inhibitors of
another type of enzyme-sensitive chemical bridge, are commonly B-cell lymphoma-extra large (Bcl-xL) anti-apoptotic protein, RNA
used in ADCs design and rely on cleavage by β-glucuronidase, the and Niacinamide phosphate ribose transferase (NAMPT) inhibitors,
level of which is often high in the tumor cellular microenvironment and carmaphycins, inhibitors of proteasome activity (Wang et al.,
(Kostova et al., 2021). 2023).

Frontiers in Pharmacology 04 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

2.3.1 DNA-damaging drugs act as crosslinkers, effects, such as hematotoxicity, and morbidity in patients, mainly
alkylators and topoisomerase inhibitors due to the mAbs backbone, linker chemistry, and target non-
The available agents that induce cell death by damaging DNA specificity rather than the drug itself (Sau et al., 2017). Originally,
can act up to picomolar concentrations (Hartley, 2011) and affect these biologics utilized mouse-derived and then chimeric mAbs
both proliferating and non-proliferating cells, so they can potentially conjugated via unstable linkers to the few weakly potent drugs
contribute to the ablation of the tumor mass by affecting tumor- available in chemotherapy. Unfortunately, after administration,
initiating cells (Cheung-Ong et al., 2013; Bornstein, 2015). From a these mAbs were recognized as non-self from the body and
mechanistic point of view, these chemicals alter the double helix in inevitably triggered an immune response through the formation
different ways, e.g., by inducing single/double strand breaks, of human anti-mouse antibodies (HAMA), often leading to serious
alkylation and cross-linking of the DNA minor groove, or by immunogenicity problems (Kim and Kim, 2015). In addition, the
inhibiting Topoisomerase I/II and thus replication. Some of them chemistry of acid-labile linkers, which are quite unstable at
include amanitins (naturally byciclic octapeptides that inhibit RNA bloodstream pH, led to uncontrolled release of payloads (Beck
Polymerase II action and disrupt RNA and protein synthesis), et al., 2017), such as calicheamicin, duocarmycin, and
calicheamicins (DNA-interactive antitumor antibiotics, that cause doxorubicin, whose potency was in any case too low to cause
DNA double-strand breaks and inhibit replication), duocarmycins cancer cell death. In this context, stochastic conjugation to
(natural DNA minor groove alkylating molecules), and random lysines did not allow to control DAR and resulted in
pyrrolobenzodiazepines (highly potent DNA minor groove heterogeneous mAbs mixtures containing unconjugated, partially
crosslinking agents). Two other compounds that have been used conjugated, and overconjugated mAbs in unknown proportions,
in first-generation ADCs that are worth mentioning are which negatively affected ADC efficacy, limited tumor penetration,
camptothecin (DNA topoisomerase I inhibitor at the replication and resulted in a narrow therapeutic window (Lucas et al., 2018).
bubble) and doxorubicin (antibiotic molecule that damages DNA by Furthermore, antigens were selected even though they lacked tumor-
intercalating into it and generating free radicals) (Sau et al., 2017; Fu specific expression, resulting in severe systemic off-target effects
and Ho, 2018; Baah et al., 2021; Mckertish and Kayser, 2021). (Sau et al., 2017; Fu et al., 2022). As expected, based on the
limitations of the first-generation ADCs, the second-generation
2.3.2 Tubulin-targeting agents block the mitotic ADCs offered some implementations aimed at improving
fuse formation and the cell cycle compound efficacy and largely reducing off-target toxicity. To
Tubulin inhibitors block the rapid proliferation of tumor cells at limit potential side effects associated with the mAb backbone as
the G2/M cell cycle stage by binding tubulin subunits, leading to cell much as possible, humanized mAbs were preferred over mouse-
death by apoptosis. This class includes maytansinoids and derived or chimeric mAbs due to the lower immunological response
auristatins, a family of tubulin-inhibiting cytotoxins that arrest upon administration (Lambert and Chari, 2014). Cleavable linkers
cells in metaphase. The auristatin derivatives monomethyl have been replaced by the non-cleavable alternative to ensure the
auristatin E (MMAE) and the less toxic F (MMAF) (Park et al., stability of ADCs in the blood and to reduce the premature and
2019) are commonly used as payloads in ADC design and exert their dangerous release of drugs (Sau et al., 2017; Tsuchikama and An,
function by blocking tubulin polymerization, thereby perturbing 2018). In addition, far more potent cytotoxic chemicals have been
microtubule growth and causing cell cycle arrest (Sau et al., 2017; discovered and selected to induce cell death by attacking DNA
Baah et al., 2021; Mckertish and Kayser, 2021). Looking at the structure (disrupting its double helix conformation) and tubulin
mechanism in detail, microtubule formation involves either polymerization (disrupting mitotic fusion formation) (Carter and
nucleation or assembly of the αβ-tubulin heterodimer into a Senter, 2008; Senter, 2009). Due to the low amount of ADC in situ,
microtubule seed in the cytoplasm (Francisco et al., 2003; the IgG1 subtype was preferred over IgG4 because it has better
Goodson and Jonasson, 2018). As auristatin, by interfering with targeting abilities and better conjugation capabilities (Lambert and
GTP hydrolysis on the β subunit, causes an excessive and sustained Chari, 2014). However, despite the introduction of improvements in
growth of microtubules, they lose the ability to shorten and separate linker stability and higher drug cytotoxicity, the main weaknesses of
sister chromatids in anaphase, causing cells to freeze in metaphase of this generation of ADCs were still heterogeneous DAR (4–8), rapid
mitosis (Waight et al., 2016). clearance for high DAR drugs, off-target toxicity, and drug
resistance effects (Sau et al., 2017). What makes the third
generation of ADCs the best (so far) is based on fully human
2.4 The development of ADC therapeutic mAbs, avoiding the disadvantage of immunogenicity, and
strategy goes through three generations of optimization in terms of linker stability, payload cytotoxicity, and
compounds site-specific conjugation. This new method consists in an evolution
of previous ones and was introduced to address heterogeneous
In the last years the ADC development path brought into the DARs and consequently improve ADC pharmacokinetics and
marketplace a dozen ADCs against various hematologic and solid utility (Fu et al., 2022). To this end, the synthesis of recombinant
malignancies. Generally, these ADCs have been divided into three Abs bearing engineered cysteine residues enabled the precise
generations (first, second, and third) according to the type of mAb, bioconjugation of various drugs, resulting in the so-called
the chemistry of the linker, the mechanism of action as well as the THIOMAB drug conjugates (TDCs). The THIOMAB antibody
relationship between DAR and the conjugation method (Sau et al., technology platform results in a highly stable and effective ADC,
2017; Fu et al., 2022). Representing the first attempt at a novel whose DAR value is almost uniform (from 2 to 4) and is associated
therapeutic strategy, the first-generation ADCs caused acute adverse with fewer systemic side effects, improved drug activity, toxicity, and

Frontiers in Pharmacology 05 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Ideally, the target proteins should be tumor-specific antigens


(TSAs), which are present only on cancer cell types, and/or
tumor-associated antigens (TAAs), which are proteins that are
highly overexpressed in tumors but rare or sparsely present in
normal tissue. In conjunction with this feature, the epitope of the
antigen should ideally face the extracellular matrix (rather than the
internal site) to ensure easier accessibility and interaction with ADC
after diffusing from blood vessels (Tipton et al., 2015; Zhao et al.,
2020). In addition, to avoid undesirable systemic side effects and
safety issues, the target antigen should be a protein that is anchored
to the membrane and not secreted into the blood circulation. If this
were the case, ADC would promote unwanted binding outside the
tumor and thus reduce the anticancer effect on malignant cells
(Ritchie et al., 2013; Zhao et al., 2020). Nevertheless, after the ADC
interaction, the optimal antigen should ensure proper
internalization of the antigen- ADC complex into the endosomal/
lysosomal pathway, leading to drug release and ultimately
cytotoxicity. It should be noted that the speed and efficiency of
the internalization process strictly depend on the nature of the
target, the type of the epitope, and the payload conjugated to ADC
(Carter and Senter, 2008; Donaghy, 2016; Fu et al., 2022). Finally,
since the tumor mass and its surrounding microenvironment (TME)
are tightly coupled and constantly communicate with each other,
TME components have been targeted as novel potential ADC targets
FIGURE 2 (see below in the text) (Andersen, 2023).
Mechanism of action of ADC strategy. The major steps of the
process are indicated on the figure. Basically, following ADC-target
interaction on the surface of cancer cell (step 1), this complex 2.5.2 The mechanism of action of ADCs requires
undergoes receptor-mediated endocytosis and enters the internalization to exert antitumor cytotoxicity
endosomal/lysosomal pathway until the payload is released in the
cytoplasm (steps 2, 3a and 4). Then, the drug can exert its killing The presence of a mAb targeting an antigen that is either
activity either damaging DNA structure in the nucles or derange specifically present on cancer cell types or highly overexpressed
mitotic fuse polymerization (step 5), leading to cell death by apoptosis. during carcinogenesis is a fundamental requirement for the ADC
A fraction of ADCs binds to FcRn receptors in the early step on
endosomal/lysosomal pathway and get transported out of the cell strategy (Alley et al., 2010; Damelin et al., 2015). With this in mind,
(step 2a and 3b). the mechanism of action of ADCs is quite simple and allows for
therapeutic action against the disease as well as potent on-target
cytotoxicity (Figure 2) (Peters and Brown, 2015; Khongorzul et al.,
2020; Drago et al., 2021; Samantasinghar et al., 2023). After
efficacy (Junutula et al., 2008). In addition, despite the potential administration, usually by intravenous injection to preserve drug
toxicity due to the high potency of the payloads, these ADCs have functionality (Nolting, 2013), the mAb portion of the ADC binds to
lower immunogenicity and hydrophilic linkers, giving patients a the target antigen on cancer cells, and after internalization of the
more chance to counteract cancer progression (Baah et al., 2021; Fu antigen- ADC complex by receptor-mediated endocytosis, the newly
et al., 2022). formed early endosome matures into a late endosome that
ultimately fuses with the lysosome. In this cellular compartment,
acidic or redox conditions combined with the presence of proteases
2.5 Binding to specific tumor-related targets (cathepsin B, plasmin, etc.) allow the detachment of the cytotoxic
triggers internalization and cytotoxicity of payload from its mAb carrier, whereupon the drug diffuses into the
ADCs cell and leads to cell death by attacking DNA structure or
microtubule polymerization (Peters and Brown, 2015;
2.5.1 Choosing the right targeting antigen is critical Khongorzul et al., 2020; Drago et al., 2021; Samantasinghar et al.,
for ADCs killing 2023). Of note, the IgG subtype that is widely employed in ADC
Considering that one of the advantages of ADC is that a potent synthesis can be rescued from the endosomal/lysosomal degradation
cytotoxic drug can be delivered specifically to cancer cells, the choice pathway and recycled outside cells through interaction with the
of target antigen must be the first consideration in developing this neonatal Fc receptor (FcRn), an IgG receptor involved in the
strategy. To take advantage of the maximal therapeutic index of regulation of IgG turnover. Therefore, this FcRn-mediated
ADCs, the ideal antigen should be a cell surface structure (such as transcytosis into the extracellular space, although involving only
proteins, glycoproteins, or aberrant gangliosides) that is highly or a small percentage of the internalized ADC -complex, can
predominantly expressed on tumor cells compared to healthy, potentially enhance the clearance of ADC and thus reduce its
normal cells, or at least abundant on malignant, disease- therapeutic index (Xu, 2015). On the other hand, if the small
associated cells (Damelin et al., 2015; Peters and Brown, 2015). molecule is permeable to the cell membrane, it can partially

Frontiers in Pharmacology 06 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

diffuse back into the extracellular matrix and enter neighboring cells 2022; Ceci et al., 2022; Andersen, 2023). The development of agents
regardless of the expression level of the antigen, causing a “bystander against these antigens not only weakens the tumor mass but also
effect” (Staudacher and Brown, 2017). This phenomenon, by provides the opportunity to modulate the TME itself, making it less
altering components of the TME, such as neovascular endothelial immunotolerant and more susceptible to tumor ablation (Andersen,
cells and/or cancer-associated fibroblasts (CAFs), could further 2023). Preclinical and clinical evidence suggest that cell types/factors
enhance the killing effect of ADC, especially in cancer lesions belonging to the tumor extracellular matrix and neo-blood vessels
with high heterogeneous target expression (Gerber et al., 2009; may be valuable choices for new ADC target antigens (Peters and
Purcell et al., 2018; Szot et al., 2018). Moreover, the therapeutic Brown, 2015; Xiao and Yu, 2021). For instance, an ADC targeting
strategy of ADCs involves other killing mechanisms to ensure stromal cells may cause cell death by altering the concentration of
efficacy against cancer. In several contexts, it has been shown growth factors in the tumor niche or induce hypoxia and nutrient
that the interaction of the mAb with its specific target can deprivation by binding to an antigen on the neo-vasculature (Jain,
directly cause potent inhibition of downstream signaling 2005; Mahadevan and Von Hoff, 2007; Xiao and Yu, 2021). In this
pathways triggered by antigen receptor stimulation (Albanell regard, it is worth mentioning the effect of the “binding site barrier”
et al., 2003; Vu and Claret, 2012; Marei et al., 2022). While the (BSB), a phenomenon that occurs between mAb and cell
Fab fragment of the carrier is bound to the target epitope on the populations near blood vessels and retains part of the ADC near
malignant cell, the Fc portion of the same mAb can interact with the them, reducing the penetration of Ab into the tumor mass (van
FcR on NK cells and macrophages, triggering ADCC and ADCP, Dongen, 2021). However, most TMAs have been identified on cells
respectively, as well as the C1q component of the complement of the immune system and targeting them offers an innovative anti-
system, triggering CDC (Junttila et al., 2011; Tai et al., 2014; Redman cancer therapeutic approach achieved by promoting effector cell
et al., 2015). proliferation, the anti-cancer cytokine/chemokine production, and
overall survival to create a new immune-hostile tumor niche with
reduced neo angiogenesis (Gajewski et al., 2006; Labani-Motlagh
2.6 The TME offers new potential targets to et al., 2020; Andersen, 2023; Del Prete et al., 2023). To date, some
ADCs strategy TMAs targeted by novel ADCs include CD74, an MHC class
chaperone II targeted by the ADC STRO-001, currently in phase
Most of the ADCs in preclinical and clinical development target I in the treatment of B-cell malignancies (NCT03424603) (Le et al.,
TAAs or TSAs localized mainly on the cancer mass (Sau et al., 2017; 2023) and CCR7, a chemokine receptor targeted by the novel ADC
Tong et al., 2021). Compared to hematologic malignancies, solid JBH492 in non-Hodgkin lymhoma and chronic lymphocytic
tumors thrive in a complex and dynamic entity called the TME, leukemia patients (NCT04240704). In addition, Camidanlumab
whose composition can vary widely depending on the tumor type. tesirine, also known as ADCT-301, is an ADC in phase 1/2 for
Key features of the TME generally include abundant extracellular the treatment of classical Hodgkin’s lymphoma (cHL) and non-HL
matrix, stromal cells (e.g., cancer-associated fibroblasts, CAFs), new (NCT02432235) (Hamadani et al., 2021), and CD276, an immune
and abnormal blood vessels, and immune cells, the latter capable of checkpoint overexpressed during pathologic angiogenesis and an
infiltrating the tumor mass and exerting pro- and anti-tumor interesting candidate target of different ADCs in advanced solid
functions (Anderson and Simon, 2020; Baghban et al., 2020). tumors (NCT04145622, NCT03729596 and NCT03595059) (Ceci
Particularly in the early stages of tumor growth, a bidirectional et al., 2022; Ziogas et al., 2023).
and complex interaction exists between cancer cells and the TME
components through the release of soluble factors that promote the
survival of the tumor mass, its local invasion and subsequent 2.7 Cancer creates different ways to escape
metastatic spread (Visser and Joyce, 2023). In this sense, the the effectiveness of ADCs
TME supports the basic needs of the tumor through a neo-
angiogenesis program that removes metabolic waste and, most A well-known feature of cancers is their ability to overcome the
importantly, restores oxygen and nutrient supply to the mass efficacy of therapeutic approaches, making them susceptible to
(Anderson and Simon, 2020; Visser and Joyce, 2023; Wang et al., various mechanisms of resistance. The evasion mechanisms
2023). Given the close relationship between these two entities, TME- developed by malignant cells can be divided into down-/high-
associated antigens (TMAs), i.e., proteins that are dysregulated on regulation of antigen, the presence of drug efflux pumps, defects
non-malignant cells within the TME, offer new potential targets for in lysosomal functions, and deregulation of signaling pathways
the treatment of solid tumors as they differentiate from more involved in cell cycle progression and apoptotic dysregulation
traditional tumor antigens (Andersen, 2023). Major TMAs (Shefet-Carasso and Benhar, 2015; Loganzo et al., 2016; García-
include chemokines and cytokines, transcription factors, Alonso et al., 2018). In this context, an association between antigen
metabolic enzymes, and checkpoint molecules. Some of their levels and the efficacy of ADC treatment with brentuximab vedotin
advantages lie in their overexpression on endothelial, stromal, was observed, whose multiple treatment cycles correlated with
and immune cells, whereas they are rare or very low in healthy CD30 downregulation and consequently stronger tumor
tissues, and in their easier accessibility to ADCs when administered resistance to MMAE (Chen et al., 2015). In another study, the
into the bloodstream, especially to antigens present on neo-vessels cancer cell line JIMT1-TM showed long-term resistance to the drug
or stromal cells. Furthermore, because TME components are distinct after repeated administration of anti-human epidermal growth
from cancer cells, they are less susceptible to resistance mechanisms factor receptor 2 (anti-HER2) transtuzumab maytansinoid, as the
caused by inefficient DNA repair mechanisms (Agrafiotis et al., level of HER2 protein decreased (Loganzo et al., 2015). Nevertheless,

Frontiers in Pharmacology 07 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

upregulation of CD33 antigen in the blood limited gemtuzumab into the marketplace for the treatment of hematologic malignancies
ozogamicin penetration into the bone marrow, suggesting that and solid tumors. Over the past 23 years, the following ADCs have
elevated CD33 levels still negatively affect treatment and likely been developed for the treatment of hematologic tumors:
reduce drug exposure (van der Velden et al., 2004). Another ®
Gemtuzumab ozogamicin (Mylotarg ), Brentuximab vedotin
non-negligible mechanism of resistance relies on a family of ® ®
(Adcetris ), Inotuzumab ozogamicin (Besponsa ), Polatuzumab
transmembrane proteins called ABC transporters (Zheng et al., vedotin ®
(Polivy ), Belantamab mafodotin (Blenrep ), ®
2021). These transmembrane proteins act as drug efflux pumps, ®
Loncastuximab tesirine (Zynlonta ) and Moxetumomab
causing various chemicals, including those used as payloads, to be ®
pasudotox (Lumoxiti ), an ADC, which uses an immunotoxin
excreted from the cancer cell, making it resistant or at least less rather than a chemotherapeutic agent as a payload. The ADCs
susceptible to treatment (Zheng et al., 2021). This mechanism has currently approved for solid tumor therapy are Ado-trastuzumab
been observed in AML cells, in which overexpression of the ABC- ®
emtansine (Kadcyla ), Fam-trastuzumab deruxtecan (Enhertu ), ®
family member MDR1 made them resistant to gemtuzumab ®
Enfortumab vedotin (Padcev ), Sacituzumab govitecan
ozogamicin (Matsumoto et al., 2012; Senter and Sievers, 2012) ® ®
(Trodelvy ), Tisotumab vedotin-tftv (Tivdak ), Mirvetuximab
and in breast carcinoma cells, in which cyclic dosing of TDM-1 ®
soravtansine (ELAHERE ), Disitamab vedotin (Aidixi ), and ®
induced an increase in ABCC1 transporter levels (Loganzo et al., ®
Cetuximab sarotalocan (Akalux ) (Fu et al., 2022; Kaplon et al.,
2015). Another escape mechanism involves lysosomal acidification. 2023; Yao et al., 2023). A brief description of each agent is provided
After administration of ADC and internalization, linkers are cleaved below and key characteristics are listed in Table 1. In addition,
by lysosomal acid hydrolases to subsequently release the cytotoxic Supplementary Table 1 provides the recruiting clinical trials on the
agent into the cytoplasm of cancer cells. However, upon persistent use of novel ADCs being investigated for the treatment of cancer.
treatments malignant cells may acquire the ability to disrupt this
process by altering the pH of the lysosomal compartment to slow the 2.8.1 Hematologic malignancies
catabolic activity of their proteases, a process that has been 2.8.1.1 Gemtuzumab ozogamicin
demonstrated in HER2-positive breast cancer clones resistant to Gemtuzumab ozogamicin (Mylotarg ; Pfizer) was the first ADC
®
long-term T-DM1 (Ríos-Luci et al., 2017; García-Alonso et al., ever developed and clinically approved by the FDA in 2000 and by
2018). Nevertheless, perturbations in signaling pathways involved the EMA in 2018. As a first-generation ADC, it was based on a
in cell cycle regulation and alterations in apoptotic regulation may humanized anti-CD33 IgG4 antibody linked to the DNA-interactive
also modulate tumor cell sensitivity to ADC (Collins et al., 2019). In agent calicheamicin (or ozogamicin) via a hydrazone-cleavable
T-DM1-resistant HER2-positive breast cancer cells, an increase in linker bound to surface lysines (average DAR 2–3). It was
cyclin B levels, a protein required for the G2/M cell cycle transition, indicated for the treatment of relapsed/refractory acute myeloid
has been observed. This upregulation at the protein level could affect leukemia (AML) though it was withdrawn in 2010, but reapproved
cell cycle dynamics by altering sensitivity to treatment with ADC at a lower dose in 2017, because patients suffered severe toxicity
(Sabbaghi et al., 2017). Moreover, in AML cells, activation of a problems likely due to the higher dose (Fu et al., 2022). Following
related pathway (PI3K/Akt) was associated with lower efficacy of administration, Mylotarg binds to the CD33 transmembrane
gemtuzumab ozogamicin and a deletion in the PTEN pathway was glycoprotein on AML cells, and upon internalization, a precursor
associated with trastuzumab low effectiveness (García-Alonso et al., of calicheamicin is released through hydrolysis of its linker. The
2018). Of note, in the same hematologic tumor, overexpression of active form of the drug then exerts a cytotoxic activity by binding to
members of the anti-apoptotic BCL-2 and BCL-X families plays a DNA and breaking its conformation to cause cell cycle arrest and cell
role in sensitivity to gemtuzumab ozogamicin (Godwin et al., 2020). death. Of note, the hydrophobic nature of calicheamicin enables a
bystander killing of cells in the TME that are negative for the
CD33 target antigen (Coats et al., 2019; Kayser and Levis, 2022).
2.8 Clinically approved ADCs for the
treatment of hematologic and solid 2.8.1.2 Brentuximab vedotin
malignancies ®
Brentuximab vedotin (Adcetris ; Seagen, Takeda Pharma) was
approved by the FDA in 2011 and by the EMA in 2012 as
The ADC strategy represents a highly successful therapeutic monotherapy for the treatment of systemic anaplastic large cell
alternative in cancer treatment. The excellent ability to deliver a lymphoma (sALCL) and in 2018 in combination with chemotherapy
pharmacological compound in situ by drawing the mAb of choice for relapsed/refractory Hodgkin lymphoma (HL). It consists of a
along with various linkers and chemical alternatives represented an chimeric IgG1 targeting CD30, a cell membrane protein of the
innovative development compared to mAb-only based therapy and tumor necrosis factor receptor family, on cancer cells and is cysteine
traditional chemotherapy. As mentioned earlier, although the conjugated to MMAE (DAR equals 4) through a protease-cleavable
development of ADC remains challenging in terms of drug linker (Mckertish and Kayser, 2021). After interacting with its target,
safety, efficacy, and targeting, the development of new and more Adcetris enters the endosomal/lysosomal pathway via clathrin-
precise technologies, as well as the identification of new targets and dependent endocytosis, where its linker is cleaved by acid
components, has led to an explosion in the use of ADC in clinical hydrolases to release MMAE in the cytoplasm. The drug
oncology (Drago et al., 2021; Dumontet et al., 2023; Fuentes-Antrás interferes with microtubule polymerization and induces apoptosis
et al., 2023). To date, hundreds of ADCs are in clinical trials, and and cell death. Like calicheamicin, MMAE exerts its cytotoxic effect
15 of them have been approved by the FDA, the European Medicines on neighboring CD30-negative cells using the bystander killing
Agency (EMA), and/or other government agencies and launched effect, suggesting that the efficacy of Adcetris in heterogeneous

Frontiers in Pharmacology 08 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

TABLE 1 FDA/EMA approved ADCs in clinical oncology. e early; m metastatic; AML acute myeloid leukemia; HL Hodgkin lymphoma; sALCL systemic Anaplastic
Large Cell Lymphoma; B-cell prec. L B-cell precursor leukemia; DLBCL diffuse large B-cell lymphoma; MM multiple myeloma; HCL hairy cell leukemia; BC breast
cancer; UC urothelial cancer; NSCLC non-small-cell lung cancer; GC gastric cancer; GOJ gastro-oesophageal junction cancer; TNBC triple negative breast cancer; CC
cervical cancer; OC ovarian cancer; HNSCC Head and neck squamous cell carcinoma. The detailed description of the treatment for each disease is described in the
text. *: withdraw from market in 2022. **: Aidixi® and Akalux® have not received FDA/EMA approval yet but Akalux® is approved by PMDA (Pharmaceuticals and
Medical Devices Agency of Japan) and Aidixi® by NMPA (National Medical Products Administration of China).

ADC Manufacturer Trade name® Target FDA/EMA approval Cancer


Gemtuzumab ozogamicin Pfizer Mylotarg CD33 2000(2017)/2018 AML

Brentuximab vedotin Seagen, Takeda Pharma Adcetris CD30 2011/2012 HL; sALCL

Inotuzumab ozogamicin Pfizer Besponsa CD22 2017 B-cell prec. ALL

Polatuzumab vedotin Genentech Polivy CD79b 2019/2020 DLBCL

Belantamab mafodotin GlaxoSmithKline Blenrep BCMA 2020* MM

Loncastuximab tesirine ADC Therapeutics Zynlonta CD19 2021/2022 large B-cell prec. L

Moxetumomab pasudotox AstraZeneca Lumoxiti PE38 2018 HCL

Ado-Trastuzumab Emtansine Genentech Kadcyla HER2 2013 HER2+ e/m BC

Enfortumab vedotin Astellas Pharma US, Seagen Padcev NECTIN4 2019/2022 mUC

Fam-trastuzumab Deruxtecan Daichii Sankyo Enhertu HER2 2019/2021 HER2+ BC; NSCLC; GC/GOJ

Sacituzumab govitecan Gilead Sciences Trodelvy TROP2 2020/2021 TNBC; mUC

Tisotumab vedotin-tftv Seagen Tivdak TF 2021 (only FDA) CC

Mirvetuximab Soravtansine ImmunoGen ELAHERE FRα 2022 (only FDA) OC

Disitamab Vedotin Remegen Aidixi** HER2 — UC; GC

Cetuximab Sarotalocan Rakuten Medical Akalux** EGFR — HNSCC

lymphomas in vivo may be related to this effect (Katz et al., 2011; 2.8.1.5 Belantamab mafodotin
Scott, 2017). ®
Belantamab mafodotin (Blenrep ; GlaxoSmithKline) was
approved by the FDA and EMA for the treatment of refractory/
2.8.1.3 Inotuzumab ozogamicin relapsed multiple myeloma in 2020 (MM) but was withdrawn in
®
Inotuzumab ozogamicin (Besponsa ; Pfizer) was approved by 2022 because it did not meet FDA standards (https://www.myeloma.
the FDA and EMA in 2017. It targets CD22, an antigen expressed on org/news-events/withdrawal-blenrep-us-market). The mAb portion
relapsed/refractory B-cell precursors in acute lymphoblastic of this ADC is unique in that it consists of a humanized Fc-
leukemia (ALL). It consists of a humanized IgG4 mAb linked to afucosylated IgG1, a modification that enhances binding and
calicheamicin via an acid-cleavable linker attached to lysine residues. cytotoxicity of the ADC. From the mAb backbone, a cysteine-
It has a DAR, ranging from 5 to 7. From a mechanistic perspective, it bound, non-cleavable linker bridges the mAb with the cytotoxic
acts similarly to gemtuzumab ozogamicin in that it is based on the payload MMAF. Its DAR is 4. The target of Blenrep is B cell
same Ab backbone and loaded with the same drug (Dahl et al., 2016; maturation antigen (BCMA), a member of the tumor necrosis
Garrett et al., 2019; Lanza et al., 2020). factor receptor family that is overexpressed in mature B
lymphocytes and plasma cells (Chen et al., 2020; Yu et al., 2020).
2.8.1.4 Polatuzumab vedotin As with other ADCs, BCMA targeting internalizes ADC and degrades
®
Polatuzumab vedotin (Polivy ; Genentech) contains a mAb to release MMAF, a tubulin inhibitor, into the cytoplasm, where
humanized IgG1 anti-CD79b, a component of the B-cell it blocks cancer cell cycle progression and leads them to death
receptor conjugated to MMAE via the same organic bridge through apoptosis (Seckinger et al., 2017; Kaplon et al., 2020).
used in the synthesis of brentuximab vedotin. The conjugation
method was via engineered cysteines utilizing the THIOMAB 2.8.1.6 Loncastuximab tesirine
system and has a DAR of 3.5. The clinical use of Polivy has been ®
Loncastuximab tesirine (Zynlonta ; ADC Therapeutics) targets
approved by the FDA in 2019 and by the EMA in 2020 for the CD19 and received accelerated approval from the FDA in 2021 and
treatment of adult patients with relapsed/refractory diffuse large from the EMA in 2022 for relapsed/refractory B-cell lymphoma after
B-cell lymphoma (DLBCL) in combination with bendamustine two or more lines of systemic therapy, including DLBCL not
and rituximab (anti-CD20 mAb) (Sehn et al., 2022; Varma et al., otherwise specified, DLBCL from low-grade lymphoma, and
2022). Upon administration, this agent is internalized into cancer high-grade B-cell lymphoma. Zynlonta targets CD19, a
cells and proteolytically cleaved to release MMAE, which causes transmembrane protein commonly expressed in all B cell
apoptotic cell death by inhibiting tubulin polymerization (Deeks, lineages, and consists of a humanized IgG1 mAb linked to
2019; Kaplon et al., 2020). SG3199 (a dimeric PBD alkylating agent) via an enzymatically

Frontiers in Pharmacology 09 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

cleavable linker (DAR of 2–3) (Lee, 2021). The payload exerts its ®
(T-Dxd, Enhertu , Daiichi Sankyo, Astrazeneca), which have
pharmacological benefit by irreversibly binding to DNA and improved the OS in the second and third-line settings and are
generating a strong adduct that inhibits DNA synthesis and currently used for the treatment of HER2+ early/metastatic and
causes cell death. Because SG3199 exhibits cytotoxicity in the HER2+ low BC, respectively (Ferraro et al., 2021; Rassy et al., 2022).
picomolar range, it is the most toxic drug currently available on
the market ADC. Currently, Zynlonta is the only anti-CD19 drug 2.8.2.1.1 Ado-trastuzumab emtansine
approved ADC for relapsed/refractory DLBCL as a single agent ®
Ado-trastuzumab emtansine (Kadcyla ; Genentech) is based on
(Zammarchi et al., 2018; Fu et al., 2022). a humanized IgG1 linked to emtansine via a non-cleavable linker
attached to the lysine residues. Its average DAR is 3.5 (Mckertish and
2.8.1.7 Moxetumomab pasudotox Kayser, 2021). After interaction with HER2 antigen, Kadcyla is
®
Moxetumomab pasudotox (Lumoxiti ; AstraZeneca) is not internalized by endocytosis and reaches the lysosome, where
widely considered an ADC because its payload consists of a IgG1 is completely proteolytically degraded. Subsequently, lysine-
fragment of Pseudomonas aeruginosa exotoxin A called PE38. MCC-DM1, a DM1-containing metabolite, is released into the
However, since it uses the same targeting mechanism based on cytosol, where it disrupts the microtubule network and causes
mAb, we would still like to consider it as part of the ADC cell death. Interestingly, lysine-MCC-DM1 has similar toxicity to
biocompound family. It was approved by the FDA in 2018 for DM1 but cannot exert its pharmacological effect via the bystander
patients with refractory/relapsed hairy cell leukemia (HCL) who killing effect due to its charge at neutral pH (Barok et al., 2014;
have not received at least two systemic therapies. It was granted Lambert and Chari, 2014). T-DM1 received FDA approval in
marketing authorization in the EU in 2021. Lumoxiti is based on an 2013 for the treatment of advanced HER2+ BC based on data
anti-CD22 mouse IgG1 mAb carrying a cleavable linker bound to from the EMILIA clinical trial (Phase III). This study evaluated
the immunotoxin PE38. After interaction, internalization, and the efficacy of T-DM1 compared to capecitabine and lapatinib in
cleavage, PE38 is released into the cell cytoplasm and acts by patients with HER2+ BC previously treated with transtuzumab and
blocking translation and inducing cell apoptosis (Kreitman and taxane chemotherapy. Results based on 911 included patients were
Pastan, 2011; Kreitman, 2019; Kang, 2021). favorable for T-DM1, whose administration resulted in an
improvement in objective response rate (ORR) (43.6% vs. 30.8%),
2.8.2 Solid tumors median PFS (9.6 months vs. 6.4 months; p < 0.001), and median OS
2.8.2.1 Trastuzumab-based ADCs (29.9 vs. 25.9 months, p < 0.001) after a median follow-up of
Since FDA approval of rituximab for the treatment of non-HL in 47.8 months (Blackwell et al., 2012; Diéras et al., 2017). A few
1997 (Leget and Czuczman, 1998), several mAbs have been years later, positive results from the KATHERINE clinical trial
investigated and achieved approval in clinical oncology. (phase III) established the novel use of T-DM1 as adjuvant
Considering the mAbs that form the scaffold of approved ADCs, therapy for patients with early-stage HER2+ BC with residual
trastuzumab is an example of an anti-cancer molecule used in disease after neoadjuvant treatment (taxane and trastuzumab).
combination therapy or alone, and also represents the carrier Among the 1,486 patients who met criteria, those who received
motif in two preparations ADC. Trastuzumab (Herceptin ) is a ® T-DM1 showed a significant 50% improvement in invasive disease-
humanized IgG1mAb that binds to the extracellular domain of free survival (IDFS) at 3 years compared with patients treated with
HER2, a tyrosine kinase receptor that is upregulated in 20% of trastuzumab alone in the control arm (88.3% vs. 77%, p < 0.001)
breast cancer (BC) patients, preventing its homodimerization and (von Minckwitz et al., 2019; Wedam et al., 2020; Mamounas et al.,
thereby blocking its intracellular signaling (Greenblatt and 2021). From the data collected in these two studies, T-DM1 exhibits
Khaddour, 2023). Because of its role in cell growth, survival, and stronger therapeutic efficacy compared to chemotherapeutic agents
differentiation, HER2+ breast cancers tend to grow and spread more and trastuzumab alone in HER2+ early or metastatic BC, likely
aggressively than HER2 negative tumors (Iqbal and Iqbal, 2014). because this ADC preserves the antineoplastic functions of
Trastuzumab was approved by the FDA in 1998 for the treatment of trastuzumab and adds a novel cytotoxic effect (Ferraro et al.,
HER2+ BC. It improved overall survival (OS) and progression-free 2021). Remarkably, the superior benefit of T-DM1 was associated
survival (PFS) (Albanell and Baselga, 1999; Goldenberg, 1999), but with manageable side effects, mostly grade 1 or 2, as only a small
its administration was also associated with risk of cardiac toxicities, percentage of included patients reported elevations in liver enzymes
such as left ventricular ejection fraction (LVEF) decline and aspartate transaminase (AST) and alanine transaminase (ALT) and
congestive heart failure (Bouwer et al., 2020). In the U.S. it is thrombocytopenia (Diéras et al., 2017).
approved for HER2+ BC in adjuvant therapy (with
anthracyclines and taxane) and for metastatic HER2+ BC in 2.8.2.1.2 Fam-trastuzumab deruxtecan
monotherapy or in combination with chemotherapeutics, tyrosine ®
Fam-trastuzumab Deruxtecan (Enhertu ; Daiichi Sankyo) is the
kinase inhibitors (TKIs), and immunotherapy. It is also used in a second ADC HER2-targeting drug approved by the FDA. T-Dxd was
combination regimen for HER2+ gastric cancer (Dumontet et al., approved by the FDA in 2019 and by the EMA in 2021 for the
2023). Trastuzumab is administered by intravenous infusion, and treatment of unresectable or metastatic HER2+ breast cancer (after
the dosing regimen can be adjusted depending on the stage of tumor patients have received two or more anti-HER2 therapies), non-small
growth (Greenblatt and Khaddour, 2023). Nowadays, the cell lung cancer, and for locally advanced or metastatic HER2+ gastric
trastuzumab backbone has been used for the synthesis of two or gastroesophageal junction adenocarcinoma, after a trastuzumab-
FDA- and EMA-approved ADCs, ado-trastuzumab emtansine based therapy. It consists of a humanized IgG1-mAb carrying Dxd, a
®
(T-DM1, Kadcyla ; Genentech) and fam-trastuzumab deruxtecan more potent DNA topoisomerase I inhibitor than SN-38, as a

Frontiers in Pharmacology 10 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

cytotoxic payload via an enzymatically cleavable linker. It has a DAR 2.8.2.3 Sacituzumab govitecan
of 7 or 8 (Mckertish and Kayser, 2021; Fu et al., 2022; Dumontet et al., ®
Sacituzumab govitecan (Trodelvy ; Gilead Sciences) is an ADC
2023). After internalization of the HER2-Enhertu complex and consisting of a humanized IgG1 mAb targeting Tumor-associated
cleavage, Dxd blocks DNA topoisomerase I, an enzyme that calcium signal transducer 2 (TROP2), a transmembrane
controls and alters the topological state of DNA during glycoprotein involved in cell self-renewal, proliferation, invasion,
transcription, leading to cell death. Compared to Kadcyla, which and survival, and plays an important role in intracellular calcium
has the same target, Enhertu has several improvements related to the signaling. It is generally overexpressed in most solid tumors,
novel cysteine-conjugated peptide linker, higher DAR and including triple negative breast cancer (TNBC) (Furlanetto et al.,
cytotoxicity of the drug, which is more potent and hydrophobic to 2022). The mAb harnesses to malignant cells SN-38, a DNA
increase the bystander killing effect on neighboring cells (Kaplon topoisomerase I inhibitor that causes DNA breaks and ultimately
et al., 2020; Shitara et al., 2021). This is essential for extending cell death. IgG1 and payload are connected via an acid-cleavable
cytotoxic activity to cells with low or heterogeneous HER2 levels. linker bound to cysteine residues with a DAR between 7 and 8 (Tong
In this sense, the bystander-killing effect achieved by T-Dxd allows for et al., 2021). Trodelvy was approved by the FDA in 2020 and by the
a better therapeutic response and thus greater cytotoxicity in tumors EMA in 2021 for the treatment of locally advanced or metastatic
refractory to its T-DM1 counterpart (Ferraro et al., 2021). In 2019, TNBC in patients who have received at least two prior therapies and
T-Dxd received accelerated FDA approval based on positive results in locally advanced or metastatic urothelial carcinoma following Pt-
from the single-arm DESTINY -Breast01 trial (Phase II). In this study, containing therapy and a PD -1 or PD -L1 inhibitor (Mehanna et al.,
a cohort of 184 female patients with HER2+ metastatic BC who had 2019; Bardia et al., 2021; Tong et al., 2021).
received two or more prior lines of therapy, including T-DM1, was
enrolled to test the efficacy of T-Dxd. Interestingly, 60.9% of patients 2.8.2.4 Tisotumab vedotin-tftv
showed an objective response, and the median duration and median ®
Tisotumab vedotin-tftv (Tivdak ; Seagen) consists of a fully
response were 16.4 months and 14.8 months, respectively, with a human IgG1, an enzymatically cleavable linker, MMAE as a payload,
median time response of 1.6 months (95% CI) (Modi et al., 2020; and a DAR equal to 4. It targets tissue factor (TF), a membrane
2021). In addition, the efficacy of T-Dxd and T-DM1 was evaluated in protein related to cancer metastasis and invasiveness that is highly
the DESTINY -Breast 03 trial (phase III), which enrolled 524 patients expressed in various solid tumors. Being the last ADC on the market,
with HER2+ metastatic BC previously treated with trastuzumab and it was approved by the FDA in 2021 for the treatment of relapsed/
taxane. Randomization data showed superior efficacy of T-Dxd in refractory metastatic cervical cancer with disease progression during
terms of ORR (79.9% vs. 34.2%), PFS (not reached vs. 6.8 months, p < or after chemotherapy (Liu et al., 2011; Alley et al., 2019; Heitz et al.,
0.001), and OS (94.1% vs. 85.9%) (Cortés et al., 2021; 2022). In 2023).
general, hematotoxicity, nausea, and fatigue were the most common
grade ≥3 adverse effects observed in patients treated with T-Dxd. Of 2.8.2.5 Mirvetuximab soravtansine
note, treatment with T-Dxd was also associated with pulmonary ®
Mirvetuximab soravtansine (ELAHERE ; ImmunoGen) was
toxicity and, in particular, interstitial lung disease (ILD), a group of approved by the FDA in 2022 for the treatment of adult patients
respiratory diseases that require careful management and may lead to with Folate Receptor α (FRα)-positive, platinum-resistant epithelial
treatment discontinuation (Diéras et al., 2017; Cardoso et al., 2020). ovarian cancer who have previously received 1–3 systemic therapies.
Finally, the DESTINY-Breast04 trial evaluated the use of T-Dxd It targets FRα, a member of the folate receptor family that is
versus physician’s choice chemotherapy in 577 patients with low overexpressed on several epithelial-derived cancer cells (Macor
HER2+ metastatic BC who had received prior chemotherapy in the et al., 2006). It consists of a chimeric mAb bound via a cleavable
metastatic setting or developed a recurrence within 6 months of linker to DM4, a potent tubulin targeting agent that belongs to the
completing adjuvant chemotherapy. The results of this study showed maytansinoids. Like other ADC, the drug exerts its cytotoxic effect
that administration of T-Dxd resulted in significantly longer median after internalization into cancer cells, leading to their death by
progression-free (9.9 months versus 5.1 months) and overall survival blocking their mitotic fuse formation (Dilawari et al., 2023; Heo,
(23.4 months versus 16.8 months) than pharmacologic therapy in 2023; Kaplon et al., 2023).
enrolled patients (Modi et al., 2022).
2.8.2.6 Disitamab vedotin
2.8.2.2 Enfortumab vedotin ®
Disitamab vedotin (Aidixi ; RemeGen) was approved by the
®
Enfortumab vedotin (Padcev ; Astellas Pharma US, NMPA (National Medical Products Administration of China) in
Seagen) is a fully human IgG1 conjugated to the 2021 as a second-line treatment for patients with HER2-expressing,
microtubule inhibitor MMAE via a protease-cleavable linker locally advanced or metastatic urothelial carcinoma (mUC) who
on cysteine residues and has a DAR of 3.8 (Joubert et al., 2020). have previously received Pt-containing chemotherapy (Fu et al.,
It targets nectin-4, a transmembrane protein involved in 2022), and approved for patients with HER2-overexpressing locally
multiple cellular signaling pathways, including cell adhesion, advanced or metastatic gastric cancer who have received at least two
proliferation, and migration, and is overexpressed in several systemic chemotherapy regimens (Deeks, 2021). It delivers HER2+
malignancies. in 2019, Padcev was approved by the FDA for cancer cell MMAE (DAR equals 4) via a cleavable linker bound to a
locally advanced or metastatic urothelial carcinoma following humanized mAb. Interestingly, Aidixi showed high specific
Pt-containing therapy and a PD -1 or PD -L1 inhibitor (Alt antigenic activity and stronger tumor activity compared to other
et al., 2020; Chang et al., 2021) and received EU-wide HER2+-targeted ADCs in preclinical experiments and in animal
marketing approval in 2022. models (Shi et al., 2022).

Frontiers in Pharmacology 11 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

2.8.2.7 Cetuximab sarotalocan This out-of-cell toxicity has two sides. On the one hand, it may
®
Cetuximab sarotalocan (Akalux ; Rakuten Medical) received extend the efficacy of treatment to antigen-negative cells found in
PMDA (Pharmaceuticals and Medical Devices Agency of Japan) the tumor niche, to cells that form the core of the tumor mass being
approval in 2020 (Gomes-da-Silva et al., 2020). It is comprised of a less accessible to the ADC (Jain, 2005; Mahadevan and Von Hoff,
chimeric IgG1 mAb specifically targeting epidermal growth factor 2007; Xiao and Yu, 2021), and to cells that have low/heterogeneous
receptor (EGFR), the triggering of which is involved in cell expression of the antigen. On the other hand, due to the non-specific
proliferation, angiogenesis, and invasion/metastasis. The effect of the conventional small drugs, normal cells can also suffer
conjugation of Akalux is not with a small molecule, but with a severe damages with potential unpredictable consequences
light-activatable near-infrared dye called 700DX (DAR 1.3–3.8) (Fu (Donaghy, 2016; Staudacher and Brown, 2017). Other limitations
et al., 2022). In this case, we would also like to consider it as ADC relate to the pharmacokinetic properties of ADCs. Rapid clearance
given its mechanism of action against cancer cells. After interaction and aggregation represent two important aspects that may
with ADC-EGFR, this ADC inhibits EGFR signaling pathway and negatively impact the therapeutic activity of these compounds
achieves high anticancer effect by laser activation of 700DX dye. In (Lucas et al., 2018; Mahmood, 2021; Pettinato, 2021). To solve
this way, malignant cells are targeted and rapidly eliminated while complications and improve the body compatibility of ADC, the
healthy cells surrounding the tumor mass are spared. It has been mAb component and linker can be chemically modified by
approved for the treatment of unresectable locally advanced or glycosylation or PEGylation (Mahmood, 2021; Edwards et al.,
recurrent head and neck squamous cell carcinoma (HNSCC) 2022). While the former has not been studied in detail within
(Kitamura et al., 2020; Omura et al., 2023). this platform, the latter allows overcoming some drawbacks
(Pettinato, 2021). PEGylation involves the addition of
polyethylene glycol (PEG) to specific amino acid residues. In
2.9 The therapeutic strategy of ADC reveals general, it has been shown that the use of PEG as a linker can
challenges and limitations and can be improve the solubility of ADCs and reduce aggregation, improving
combined in combinatorial regimens their pharmacokinetic by enhancing stability and distribution in the
body (Verhoef and Anchordoquy, 2013; Mahmood, 2021).
2.9.1 Payloads are the main cause of ADCs toxicity
In 2 decades, ADCs have achieved remarkable results in the 2.9.2 Clinical manifestations of ADCs include major
treatment of hematologic malignancies and solid tumors and toxicities
represent a valid alternative in the field of clinical oncology. As ADCs are developed to limit their exposure to healthy tissues,
Although they have been designed to release cytotoxic agents by they are associated with quite manageable toxicities, with nausea,
targeting selective cell populations, a striking number of clinical vomiting, diarrhea and fatigue being among the most frequent ones.
trials have shown that ADCs are not free of adverse effects, Unfortunately, the large amount of data provided by several clinical
sometimes leading to toxicities commonly observed with trials also highlights the presence of severe toxicities (grade 3 or
conventional chemotherapy (Dumontet et al., 2023; Tarantino higher) that include peripheral neuropathy and hematotoxicity,
et al., 2023). In general, a significant proportion of patients often dose-limiting (Masters et al., 2018; Professional Committee
suffered various toxicities, sometimes so severe (or fatal) to on Clinical Research of Oncology Drugs et al., 2022). Peripheral
require dose reduction or interruption, treatment delays, and neuropathy includes tingling, pain in the extremities, numbness, and
supportive medications (Dumontet et al., 2023; Tarantino et al., rarely muscle weakness and is commonly associated with ADCs
2023). Each of the blocks that form an ADC can result in significant carrying cleavable linkers bound to tubulin inhibitors (i.e., all ADCs
side effects when these compounds are administered to the human loaded with MMAE) and Mirvetuximab soravtansine, carrying DM4
body. Even if the nature of mAbs is responsible for moderate/severe (Nguyen et al., 2023). As cleavable linkers are associated to the
immunogenic side effects, especially in ADCs preparations using premature drug release and that these compounds block tubulin
murine and chimeric mAb scaffolds (Kim and Kim, 2015), the polymerization, this common side effect is not unexpected as
primary manifestation of a toxicity profile is highly dependent on microtubules are deeply involved in the axonal transport, an
the type of payload (Zahavi and Weiner, 2020). Most ADCs used in essential process to the growth and maturation of neurons
the clinic are loaded with tubulin inhibitors or DNA-interacting (Yogev et al., 2016). Hematologic side effects include anemia,
agents that exert their cytotoxic effects in the range of nano- or neutropenia, thrombocytopenia, leukopenia and are mainly due
picomolar concentrations and are highly toxic when used in to the off-target Fc receptor-mediated uptake of ADCs into
unconjugated form (Mecklenburg, 2018). Unfortunately, because immune cells, with neutropenia being the most prominent
only a very small percentage of ADCs reach their targets and a part toxicity in ADC-based monotherapy (Mahalingaiah et al., 2019).
of the payload is released prematurely, a significant portion of the Besides, major toxicities responsible for dose limitations are related
dose is virtually free to interact with numerous non-target healthy to drug classes and mainly include hepatotoxicity (for MMAF, DM1,
cells and cause unconventional systemic or local side effects and calicheamicin), skin toxicity (for MMAE and PBD), and ocular
(Dahlgren and Lennernäs, 2020). The out-of-target toxicities are toxicity (for MMAF and DM4) (Zhao et al., 2020; Hurwitz et al.,
closely related to the linker. Non-cleavable linkers exhibit greater 2023). Of note, the mechanism underlying corneal toxicity has not
stability in plasma and the ADCs are better tolerated by the body. been solved yet but occurred in a relevant percentage of patients
However, in the majority of ADCs used in the clinic and foremost to treated with Belantamab mafotodin, Trastuzumab emtansine and
the ones use in solid tumors, cleavable linkers are preferred as they Mirvetuximab soravtansine, all loaded with tubulin inhibitors
have shown more benefits, probably due to the bystander side effect. (Aschauer et al., 2022; Domínguez-Llamas et al., 2023). Other

Frontiers in Pharmacology 12 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

relevant clinical manifestations include gastrointestinal side effects have demonstrated robust clinical activity in several malignancies
upon administration of Sacituzumab govitecan and Trastuzumab but only a fraction of patients experience long-term benefits with
deruxtecan, two ADC used in the treatment of breast cancer and monotherapy (Wojtukiewicz et al., 2021). From this perspective, an
loaded with Topoisomerase inhibitors and serosal effusion for effort could come from ADCs, capable to enhance antitumor immune
duocarmycin and PBD, the latter responsible of nephrotic responses by inducing tumor-specific adaptive immunity through
toxicity in Loncastuzimab tesirine preparation (Nguyen et al., increasing T cell infiltration into the TME, while ICIs revitalize
2023). In addition, Trastuzumab emtansine and Trastuzumab exhausted T cells (Nicolò et al., 2022). Accordingly, encouraging
deruxtecan, both targeting HER2+ breast cancer, are associated results come from the combination of ICIs and HER2-targeted
with an increased risk of interstitial lung disease (ILD)/ trastuzumab emtansine in the treatment of PD-L1+, HER2+
pneumonitis, which must be carefully managed with dose advanced breast cancer (Emens et al., 2020).
adjustment and supportive care recommendations to avoid fatal
outcomes (Ma et al., 2018; Hackshaw et al., 2020). Like other
strategies based on small molecules and immunotherapy, ADCs 2.10 ADC strategy shows application in non-
show some challenges that need to be carefully addressed to improve oncology indications
their efficacy and reduce systemic side effects. A variety of
approaches is being taken into consideration in clinics to deeply In recent years, research groups have investigated the use of ADCs
counteract the manifestation of side effects, and many of them focus in non-oncologic contexts, mainly focusing on the treatment of bacterial
on an individual basis. Extensive efforts are being made to identify infections and autoimmune diseases. Tvilum et al. achieved
patients who have potentially life-threatening toxicities at an early antimicrobial efficacy by developing an Antibody-Antibiotic
stage and offer them supportive measures, such as dose and schedule Conjugate (AAC), an antibody directed against bacteria conjugated
adjustments. The investigation of patients’ history and data on to the antimicrobial molecule mitomycin C, for the treatment of
previous treatments, comorbidities, or genetic profiles, including implant-associated biofilm infections caused by Staphylococcus
pharmacogenetic analysis to identify SNPs or potential mutations in aureus, the most common causative agent in prosthetic joint
key genes, will undoubtedly play a critical role in the determination infections (Tvilum et al., 2023). In another study, O-Leary et al.
of the best strategy to improve the tolerability and efficacy of ADCs proposed the development of an Antibody-Bactericide Conjugate
(Lambert and Morris, 2017; Dumontet et al., 2023; Tarantino et al., (ABC), an antibody conjugated to an antimicrobial peptide that
2023). exerts its effect by binding to the cell surface of P. aeruginosa,
providing another interesting example of ADC activity against
2.9.1 ADCs can be used in combination therapies bacterial infections (O’Leary et al., 2023). Regarding inflammatory
In addition to the use of ADCs as monotherapy, recent preclinical diseases, Yasunaga et al. developed an ADC targeting IL-7 receptor
and clinical studies have also focused on ADCs combinations with (IL-7R) conjugated with MMAE and showed that ADC-mediated
chemo-immunotherapics. Ideally, concomitant administration of immunoregulation of the IL-7R, the upregulation of which is a
ADCs with antiangiogenic agents, i.e., agents that damage neo-blood common mechanism in the pathogenesis of autoimmunity,
vessels to facilitate ADC penetration into the tumor niche, or specifically depleted IL-7R-positive cells in the inflammation site of a
immunomodulatory drugs that promote immune surveillance should mouse model of autoimmune arthritis and abrogated disease
amplify the body’s anti-neoplastic response to the tumor mass and its progression (Yasunaga et al., 2017). In the same year, Lee et al.
TME, without or with limited severe toxicities and safety issues demonstrated in a model of rheumatoid arthritis (RA) the
(Dumontet et al., 2023; Fuentes-Antrás et al., 2023). Regarding ADC immunosuppressive efficacy of TCZ-ALD, an ADC that targets the
combination with chemotherapy, promising data include the IL-6 receptor (IL-6R) and is conjugated to the small molecule
combination of Brentuximab vedotin and CHP (cyclophosphamide, alendronate. TCZ-ALD blocks the IL-6R activity and macrophages
doxorubicin, and prednisone) and Polatuzumab vedotin and activity in the manifestation of RA symptoms and joint inflammation
Rituximab-CHP in CD30+ peripheral T cell lymphoma (Herrera (Lee et al., 2017). A few years later, Gillard et al. achieved immune reset
et al., 2021) and in DLBCL (Tilly et al., 2022), respectively. of disease-causing reactive T cells by a single administration of CD45-
Similarly, the combination therapy based on Trastuzumab emtansine ADC and bone marrow transplantation, resulting in significant disease-
and the selective anti-HER2 tyrosine kinase inhibitor (TKI) tucatinib modifying effect in mouse models of autoimmune disease (Gillard et al.,
achieved remarkable benefits in metastatic breast cancer (Nader-Marta 2020). In addition, a few studies also addressed the use of ADCs in
et al., 2022) and the administration of bevacizumab, the mAb targeting cardiovascular (Lim et al., 2015) and renal diseases (Kvirkvelia et al.,
the Vascular-Endothelial Growth Factor (VEGF), with Mirvetuximab 2015), highlighting the potential application of this therapeutic strategy
soravtansine obtained similar benefits in preclinal models of ovarian in other pathological conditions.
cancer (Ponte et al., 2016). Combinatorial approaches using ADCs and
immunotherapeutic agents have been recently explored in several types
of cancers (Fuentes-Antrás et al., 2023). Immune checkpoint inhibitors 2.11 Challenges and solutions in the
(ICIs) are immunotherapy that enhance antineoplastic immune manufacturing of ADCs
responses by converting exhausted T-cells into activated ones and
bypassing the pathways that cause the tumor escape from the 2.11.1 Process development consists of several
immune system (Shiravand et al., 2022; von Arx et al., 2023). ICIs steps
targeting Cytotoxic T-Lymphocyte Antigen 4 (CTLA-4) and the The main goal in the manufacture of ADCs by companies is to
Programmed Cell Death Protein 1/its ligand (PD-1/PD-L1) axis produce a pure and bioburden-free compound that is safe for the

Frontiers in Pharmacology 13 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

human body for clinical use. Since the preparations of ADC consist improvement in technology and investments by major
of mixtures of mAbs with multiple conjugation sites and small biopharmaceutical companies in extensive research programs will
organic molecules, several analytical steps are required in the drive the growth of the ADC market (valued at approximately
development of these compounds, and several challenges must be $7 billion in June 2022 but expected to reach $22.4 billion in
overcome. Overall, the production of ADCs can be divided into 2030 (www.researchandmarkets.com) and increase the number of
three steps: production of the antibody, synthesis of the drug-linker available ADC-based therapies in new medical areas.
complex, and conjugation to form the final ADC (Hutchinson et al.,
2018; Bulger et al., 2023). The conjugation step is of fundamental 2.11.2 Analytical characterization startegies
importance as it determines the therapeutic efficacy of the Another challenge in the production of ADCs is the evaluation
biomolecule. Conventional conjugation methods based on lysine of their biochemical attributes to obtain a safe and effective product.
side-chains or reduced cysteines result in heterogeneous ADC Critical quality attributes of ADCs that must be carefully controlled
preparations whose safety and therapeutic efficacy are difficult to during the manufacturing process include determination of DAR
predict. To overcome this crucial limitation, site-selective and distribution of drug, residual non-conjugated species, especially
conjugation methods have recently been developed. In this way, a in terms of mAb and payload, and evaluation of size and charge
known number of drug molecules are constantly bound to selected variants in the final preparations. Nowadays, various in vitro
sites on mAbs, resulting in a more homogeneous mixture with instruments and techniques are used either alone or in
improved batch-to-batch consistency and therapeutic efficacy (Cao combination to perform comprehensive analytical
et al., 2019; Nejadmoghaddam et al., 2019). After the conjugation characterization of ADCs, including spectroscopic,
step, ADC proceeds to purification and filling into aseptic vials, all chromatographic, and mass spectrometric methods (Wagh et al.,
following the sterile pipeline of current Good Manufacturing 2018; Liu et al., 2022).
Practices (cGMP). Quality is controlled throughout the
development process. Production requires a biological 2.11.2.1 Determination of purity
manufacturing environment that allows for the safe handling of Purity of compound is a fundamental goal in any
sensitive structures, such as various powders and chemicals, purified biopharmaceutical manufacturing process. One of the techniques
mAbs, and high potency drugs, to minimize potential pollution and used to evaluate the purity of ADC preparations is size exclusion
losses that may occur at various stages of production. To ensure chromatography followed by ultraviolet detection (SEC-UV), a
product purity and sterility, synthesis and bioconjugation reactions method that separates molecules by size and, in some cases,
are performed in aseptic rooms and conditions, with regular molecular weight, and is used to fractionate large
documentation of instrument accuracy in accordance with cGMP macromolecules such as proteins (e.g., mAbs) or protein
standards. To reduce potential contamination and exposure from complexes (de Mel et al., 2019). In ADC synthesis, separation of
the use of hazardous substances, each step of the experimental macromolecules by size allows purification of the mAb from
process is achieved by providing in-depth expertise and specialized potential fragments, aggregates, and particles, three examples of
equipment to personnel and operators (Hutchinson et al., 2018; undesirable species that affect the efficacy of the final ADC as well as
Schmidhalter et al., 2019). In addition, the production of ADC its safety when administered to patients (Jiang et al., 2019). In
usually involves several departments/services and, in this sense, an general, coupling SEC with multi-angle laser light scattering (SEC-
extremely low temperature supply chain and secure packaging must MALS) offers some advantages for biopharmaceutical applications.
be ensured to reduce possible variations, damage and leakage during In MALS detection, a laser beam passes through a sample solution
long-term storage and transportation (Ducry, 2012). Considering containing the target molecule, and depending on the size of the
the complexity and number of processes involved, the production of molecules, the intensity of the scattered light is measured at specific
ADCs is quite laborious, time-consuming and economically angles (Some et al., 2019). Compared to SEC itself, SEC-MALS
expensive. Large capital investments are required to cover a requires high-purity columns but provides additional information
multitude of steps ranging from the design of the experiment, the by increasing the sensitivity for detecting impurities in the
invention and development of new drugs to product innovation, preparation (Beck et al., 2019). Other techniques for determining
differentiation, and safety monitoring, all at a cost that must make the presence of aggregation or fragmentation include dynamic light
the finished drug marketable (www.cbo.gov). To overcome these scattering (DLS), sedimentation velocity analytical
barriers, contract development and manufacturing organizations ultracentrifugation (SV-AUC) (Ducry, 2012), and capillary
(CDMOs), companies that provide drug development and electrophoresis followed by sodium dodecyl sulfate analysis (CE-
manufacturing services to the pharmaceutical industry, are SDS), a valuable method commonly used in the biopharmaceutical
turning to single-use technologies (SUTs) and automated end-to- industry for mAbs and ADC preparations to determine batch
end systems (Schmidhalter et al., 2019). SUTs are sterile, single-use consistency and overall protein purity (Wagner et al., 2020). In
items made of various plastics, most of which can be used in the addition, various residual species in ADC preparations may pose a
same way as their stainless-steel counterparts without the need for potential safety risk to patients and are a critical quality attribute that
sterilization and recycling after use. As a result, these technologies must be carefully evaluated. The levels of these unconjugated forms
offer significant advantages over traditional reusable systems by can be monitored by measuring the charge on the molecules
reducing the risk of cross-contamination, better ensuring sterility, (Wakanar, 2011). Since the bioconjugation reaction between the
allowing more flexibility throughout the process and, most mAb and the payload can significantly alter the electrostatic profile
importantly, improving cost efficiency and reducing time to of the newly formed ADC, charge-based separation techniques such
market (Schmidhalter et al., 2019). In these terms, continuous as ion exchange chromatography (IEC), isoelectric focusing gel

Frontiers in Pharmacology 14 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

electrophoresis (IEF), and capillary isoelectric focusing (cIEF) can


provide information on charge heterogeneity, drug distribution
pattern, and overall preparation quality (Ducry, 2012). To date,
imaged capillary isoelectric focusing (iCIEF) is considered a robust
method in biopharmaceutical quality control because it can
quantitatively separate samples based on the isoelectric point (pI)
of individual variants (Wagh et al., 2018; Abbood, 2023). Therefore,
iCIEF can be used to rapidly measure the content of free mAb in an
ADC mixture according to the different pI of the mAb and its
conjugated form. However, the drawbacks of this assay are that
conjugates cannot be distinguished from reaction intermediates and
other impurities, and it can only be used for conjugation chemistries
that result in significant changes in charge and net pI (Wagh et al.,
2018).

2.11.2.2 Determination of DAR and drug distribution


Probably the hottest topic in the ADC manufacturing
process is the ability to achieve bioconjugation reactions that
lead to the synthesis of a homogeneous mixture and thus a
controlled DAR and payload. Various analytical methods have
been used to determine these parameters, including ultraviolet/
FIGURE 3
visible spectroscopy (UV/Vis) (Andris et al., 2018), hydrophobic
Overview on the tumor-targeting ADCs. Taking advantage of
interaction chromatography (HIC) (Andris and Hubbuch, blood circulation, ADCs reach the tumor microenvironment (TME),
2020), reversed-phase liquid chromatography (RPLC) (Chen which is composed by cancer-associated fibroblasts (CAFs) and other
cell types and interact with malignant cells that exposed the
et al., 2019), and mass spectrometry (MS). As for MS, its
tumor associated/specific antigen on their surface. In addition to their
extended use in characterization of ADCs is discussed in canonical mechanism of action, ADCs can potentiate their positive
other works (Debaene et al., 2014; Zmolek et al., 2016; Liu response against the tumor mass. To this aim, ADCs hydrophobic
payloads may diffuse through the cell membrane inducing the killing
and Chen, 2022; Barbero et al., 2023). UV/Vis spectrometry is
of neighborhood antigen-negative cells via bystander killing effect
relatively easy to measure DAR compared to the other (black arrows) and/or the ADCs antibody Fc fragment may elicit anti-
techniques, although it requires sufficiently different tumor immunity (ADCC, CDC, and ADCP) by engaging immune
effector mechanisms, such as complement system, macrophages and
absorption profiles between the mAb and the payload and a
NK cells. All together, these mechanisms aim to induce the death of
UV/Vis chromophore on the payload (Wagh et al., 2018). the cellular component of the tumor mass via apoptosis.
Among liquid chromatographic methods, HIC and RPLC are
routinely used to measure average DAR and drug distribution
(Ouyang, 2013; D’atri et al., 2019). HIC is a method that allows
determination of species distribution based on differences in 2.12 Conclusion and perspectives
their hydrophobic properties and uniquely preserves the native
structures and activity of ADCs. Because the analysis is Nowadays, ADC represents a solid strategy to treat different
performed under mild, non-denaturing conditions, ADCs can types of malignancies. The design of ADCs provides an exceptional
be studied in their native conformation, which is an advantage opportunity to selectively deliver an effective anti-tumor chemical to
because isolation of chromatographically pure species allows the target cell and eliminate it without severe toxic off-target effects.
their further characterization in subsequent analysis. This The main advantage of ADC lies in its mechanism of action, as it
method is commonly used to analyze cysteine-conjugated offers the potential to overcome some major limitations of
ADCs and other site-specific conjugations but cannot be conventional small molecule-based therapies, such as low
applied to ADCs obtained by lysine conjugation because the therapeutic index and high off-tumor toxicity The possibility of
greater heterogeneity of these preparations complicates killing neighboring antigen-negative cells, through the bystander
chromatographic separation (D’atri et al., 2019; Liu and effect, in the mass and in the TME and the potential activation of
Chen, 2022). Nevertheless, HIC requires a large amount of direct and indirect anti-cancer mechanisms via mAb-antigen
starting material, shows low efficiency for randomly interaction and immune cell activation, respectively, argue for
conjugated ADCs, and cannot separate positional isomers their use in the clinic (Figure 3). In this context, it is worth
from cysteine-conjugated ADCs or determine the chain, H or noting that the benefits associated with the activity of ADCs
L, to which the drug was conjugated (Becker et al., 2020; when administered as monotherapy may encounter consistent
Fleming, 2020; Liu and Chen, 2022). The RPLC method also clinical limitations due to the presence of tumor-derived
separates the components of a mixture according to their resistance mechanisms and several manageable and few severe
hydrophobicity, but this approach requires denaturation of adverse effects mainly caused by the payload of the ADC
the proteins, resulting in the loss of some information about (Fuentes-Antrás et al., 2023). Despite the promising results
the distribution of some ADCs and the loss of certain DAR obtained so far, the ADC technology is still under investigation
species (Liu and Chen, 2022). and has some limitations in terms of its pharmacokinetic properties

Frontiers in Pharmacology 15 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

and biological efficacy in different tumor contexts. To develop a of University and Research under PNRR—M4C2-I1.3 Project PE_
more suitable generation of ADCs, future prospects in this field are 00000019 “HEAL ITALIA” to MDB (Spoke 5) and GT (Spoke 6)
based on the optimization of available technologies and especially on (CUP J33C22002930006 of Centro di Riferimento Oncologico di
the discovery of new methodologies. Accordingly, profound Aviano, IRCCS). This research was supported in part by the Italian
improvements in the validation of newly developed mAbs, in the Ministry of Health (Ricerca Corrente).
synthesis of less immunogenic and more stable linkers, and in the
discovery of more effective payloads are being actively explored by
scientists in this field, and similar advance are also focused on the Conflict of interest
development of more appropriate formulations as well as on the
identification of new target antigens. Given the central role of the The authors declare that the research was conducted in the
TME in solid tumor progression and spread, targeting novel absence of any commercial or financial relationships that could be
candidates upregulated in stromal cells, blood vessels and most construed as a potential conflict of interest.
importantly immune cells within the TME could lead to greater
inhibition of tumor escape mechanisms and metabolic dysfunctions
to achieve long-lasting therapeutic effects. Moreover, combinatorial Publisher’s note
therapies with different drug classes have already shown synergistic
and promising results in the treatment of hematologic and solid All claims expressed in this article are solely those of the authors
tumors by enhancing the anticancer efficacy and therapeutic index and do not necessarily represent those of their affiliated
of ADCs. Based on these observations, all these efforts are aimed at organizations, or those of the publisher, the editors and the
developing a new generation of ADCs that will undoubtedly show reviewers. Any product that may be evaluated in this article, or
significant improvements in terms of pharmacological properties, claim that may be made by its manufacturer, is not guaranteed or
therapeutic efficacy and safety in the field of oncology and in other endorsed by the publisher.
pathological conditions.

Author disclaimer
Author contributions
The views and opinions expressed are those of the authors only
FR: Writing–original draft. MDB: Writing–review and editing. and do not necessarily reflect those of the European Union or the
PM: Writing–review and editing. GT: Writing–review and editing. European Commission. Neither the European Union nor the
European Commission can be held responsible for them.

Funding
Supplementary material
The authors declare financial support was received for the
research, authorship, and/or publication of this article. The The Supplementary Material for this article can be found online
research leading to these results has received funding from the at: https://www.frontiersin.org/articles/10.3389/fphar.2023.1274088/
European Union—NextGenerationEU through the Italian Ministry full#supplementary-material

References
Abbood, A. (2023). Optimization of the imaged cIEF method for monitoring the Alley, S. C., Okeley, N. M., and Senter, P. D. (2010). Antibody-drug conjugates:
charge heterogeneity of antibody-maytansine conjugate. J. Anal. Methods Chem. 2023, targeted drug delivery for cancer. Curr. Opin. Chem. Biol. 14, 529–537. doi:10.1016/j.
8150143. doi:10.1155/2023/8150143 cbpa.2010.06.170
Abrahams, C. L., Li, X., Embry, M., Yu, A., Krimm, S., Krueger, S., et al. (2018). Alt, M., Stecca, C., Tobin, S., Jiang, D. M., and Sridhar, S. S. (2020). Enfortumab
Targeting CD74 in multiple myeloma with the novel, site-specific antibody-drug Vedotin in urothelial cancer. Ther. Adv. Urol. 12, 1756287220980192. doi:10.1177/
conjugate STRO-001. Oncotarget 9, 37700–37714. doi:10.18632/oncotarget.26491 1756287220980192
Adams, G. P., Schier, R., McCall, A. M., Simmons, H. H., Horak, E. M., Alpaugh, R. K., Andersen, M. H. (2023). Tumor microenvironment antigens. Semin. Immunopathol.
et al. (2001). High affinity restricts the localization and tumor penetration of single- 45, 253–264. doi:10.1007/s00281-022-00966-0
chain fv antibody molecules. Cancer Res. 61, 4750–4755.
Anderson, N. M., and Simon, M. C. (2020). The tumor microenvironment. Curr. Biol.
Agrafiotis, A. C., Siozopoulou, V., Hendriks, J. M. H., Pauwels, P., Koljenovic, S., and 30, R921–R925. doi:10.1016/j.cub.2020.06.081
Van Schil, P. E. (2022). Tumor microenvironment in thymic epithelial tumors: A
Andris, S., and Hubbuch, J. (2020). Modeling of hydrophobic interaction
narrative review. Cancers (Basel) 14, 6082. doi:10.3390/cancers14246082
chromatography for the separation of antibody-drug conjugates and its application
Albanell, J., and Baselga, J. (1999). Trastuzumab, a humanized anti-HER2 towards quality by design. J. Biotechnol. 317, 48–58. doi:10.1016/j.jbiotec.2020.04.018
monoclonal antibody, for the treatment of breast cancer. Drugs Today (Barc) 35,
Andris, S., Rüdt, M., Rogalla, J., Wendeler, M., and Hubbuch, J. (2018). Monitoring of
931–946. doi:10.1358/dot.1999.35.12.564040
antibody-drug conjugation reactions with UV/Vis spectroscopy. J. Biotechnol. 288,
Albanell, J., Codony, J., Rovira, A., Mellado, B., and Gascón, P. (2003). Mechanism of 15–22. doi:10.1016/j.jbiotec.2018.10.003
action of anti-HER2 monoclonal antibodies: scientific update on trastuzumab and 2C4. Aschauer, J., Donner, R., Lammer, J., Roberts, P., Funk, M., Agis, H., et al. (2022).
Adv. Exp. Med. Biol. 532, 253–268. doi:10.1007/978-1-4615-0081-0_21 Corneal toxicity associated with Belantamab mafodotin is not restricted to the
epithelium: Neuropathy studied with confocal microscopy. Am. J. Ophthalmol. 242,
Alley, S. C., Harris, J. R., Cao, A., Heuvel, E. G., Velayudhan, J., Satijn, D., et al. (2019).
116–124. doi:10.1016/j.ajo.2022.06.009
Abstract 221: Tisotumab vedotin induces anti-tumor activity through MMAE-
mediated, Fc-mediated, and Fab-mediated effector functions in vitro. Cancer Res. Baah, S., Laws, M., and Rahman, K. M. (2021). Antibody–drug conjugates—a tutorial
79, 221. doi:10.1158/1538-7445.AM2019-221 review. Molecules 26, 2943. doi:10.3390/molecules26102943

Frontiers in Pharmacology 16 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Baghban, R., Roshangar, L., Jahanban-Esfahlan, R., Seidi, K., Ebrahimi-Kalan, A., Chen, T., Chen, Y., Stella, C., Medley, C. D., Gruenhagen, J. A., and Zhang, K. (2016).
Jaymand, M., et al. (2020). Tumor microenvironment complexity and therapeutic Antibody-drug conjugate characterization by chromatographic and electrophoretic
implications at a glance. Cell Commun. Signal. 18, 59. doi:10.1186/s12964-020-0530-4 techniques. J. Chromatogr. B 1032, 39–50. doi:10.1016/j.jchromb.2016.07.023
Balendiran, G. K., Dabur, R., and Fraser, D. (2004). The role of glutathione in cancer. Chen, W., Yuan, Y., and Jiang, X. (2020). Antibody and antibody fragments for cancer
Cell Biochem. Funct. 22, 343–352. doi:10.1002/cbf.1149 immunotherapy. J. Control Release 328, 395–406. doi:10.1016/j.jconrel.2020.08.021
Barbero, L. M., Ianni, A. D., Molinaro, F., Cowan, K. J., and Sirtori, F. R. (2023). Cheung-Ong, K., Giaever, G., and Nislow, C. (2013). DNA-Damaging agents in
Hybrid liquid chromatography high resolution and accuracy mass spectrometry cancer chemotherapy: serendipity and chemical biology. Chem. Biol. 20, 648–659.
approach for quantification of antibody-drug conjugates at the intact protein level doi:10.1016/j.chembiol.2013.04.007
in biological samples. Eur. J. Pharm. Sci. 188, 106502. doi:10.1016/j.ejps.2023.106502
Cilliers, C., Menezes, B., Nessler, I., Linderman, J., and Thurber, G. M. (2018).
Bardia, A., Hurvitz, S. A., Tolaney, S. M., Loirat, D., Punie, K., Oliveira, M., et al. Improved tumor penetration and single-cell targeting of antibody drug conjugates
(2021). Sacituzumab govitecan in metastatic triple-negative breast cancer. N. Engl. increases anticancer efficacy and host survival. Cancer Res. 78, 758–768. doi:10.1158/
J. Med. 384, 1529–1541. doi:10.1056/NEJMoa2028485 0008-5472.CAN-17-1638
Bargh, J. D., Isidro-Llobet, A., Parker, J. S., and Spring, D. R. (2019). Cleavable linkers Coats, S., Williams, M., Kebble, B., Dixit, R., Tseng, L., Yao, N.-S., et al. (2019).
in antibody-drug conjugates. Chem. Soc. Rev. 48, 4361–4374. doi:10.1039/c8cs00676h Antibody-drug conjugates: Future directions in clinical and translational strategies to
improve the therapeutic index. Clin. Cancer Res. 25, 5441–5448. doi:10.1158/1078-0432.
Barok, M., Joensuu, H., and Isola, J. (2014). Trastuzumab emtansine: mechanisms of
CCR-19-0272
action and drug resistance. Breast Cancer Res. 16, 209. doi:10.1186/bcr3621
Collins, D. M., Bossenmaier, B., Kollmorgen, G., and Niederfellner, G. (2019).
Beck, A., D’Atri, V., Ehkirch, A., Fekete, S., Hernandez-Alba, O., Gahoual, R., et al.
Acquired resistance to antibody-drug conjugates. Cancers (Basel) 11, 394. doi:10.
(2019). Cutting-edge multi-level analytical and structural characterization of antibody-
3390/cancers11030394
drug conjugates: present and future. Expert Rev. Proteomics 16, 337–362. doi:10.1080/
14789450.2019.1578215 Cooper, G. M. (2000). “The development and causes of cancer,” in The cell: A
molecular approach. 2nd edition (Sunderland: Sinauer Associates). Available at: https://
Beck, A., Goetsch, L., Dumontet, C., and Corvaïa, N. (2017). Strategies and challenges
www.ncbi.nlm.nih.gov/books/NBK9963/ (Accessed July 12, 2023).
for the next generation of antibody-drug conjugates. Nat. Rev. Drug Discov. 16,
315–337. doi:10.1038/nrd.2016.268 Cortés, J., Kim, S.-B., Chung, W.-P., Im, S.-A., Park, Y. H., Hegg, R., et al. (2021).
LBA1 Trastuzumab deruxtecan (T-DXd) vs trastuzumab emtansine (T-DM1) in
Becker, C. L., Duffy, R. J., Gandarilla, J., and Richter, S. M. (2020). Purification of
patients (Pts) with HER2+ metastatic breast cancer (mBC): Results of the
ADCs by hydrophobic interaction chromatography. Methods Mol. Biol. 2078, 273–290.
randomized phase III DESTINY-Breast03 study. Ann. Oncol. 32, S1287–S1288.
doi:10.1007/978-1-4939-9929-3_19
doi:10.1016/j.annonc.2021.08.2087
Blackwell, K. L., Miles, D., Gianni, L., Krop, I. E., Welslau, M., Baselga, J., et al. (2012).
Cortés, J., Kim, S.-B., Chung, W.-P., Im, S.-A., Park, Y. H., Hegg, R., et al. (2022).
Primary results from EMILIA, a phase III study of trastuzumab emtansine (T-DM1)
Trastuzumab deruxtecan versus trastuzumab emtansine for breast cancer. N. Engl.
versus capecitabine (X) and lapatinib (L) in HER2-positive locally advanced or
J. Med. 386, 1143–1154. doi:10.1056/NEJMoa2115022
metastatic breast cancer (MBC) previously treated with trastuzumab (T) and a
taxane. JCO 30, LBA1. doi:10.1200/jco.2012.30.18_suppl.lba1 Dahl, J., Marx, K., and Jabbour, E. (2016). Inotuzumab ozogamicin in the treatment of
acute lymphoblastic leukemia. Expert Rev. Hematol. 9, 329–334. doi:10.1586/17474086.
Bornstein, G. G. (2015). Antibody drug conjugates: Preclinical considerations. AAPS
2016.1143771
J. 17, 525–534. doi:10.1208/s12248-015-9738-4
Dahlgren, D., and Lennernäs, H. (2020). Antibody-drug conjugates and targeted
Bouwer, N. I., Jager, A., Liesting, C., Kofflard, M. J. M., Brugts, J. J., Kitzen, J. J. E. M.,
treatment strategies for hepatocellular carcinoma: A drug-delivery perspective.
et al. (2020). Cardiac monitoring in HER2-positive patients on trastuzumab treatment:
Molecules 25, 2861. doi:10.3390/molecules25122861
A review and implications for clinical practice. Breast 52, 33–44. doi:10.1016/j.breast.
2020.04.005 Damelin, M., Zhong, W., Myers, J., and Sapra, P. (2015). Evolving strategies for target
selection for antibody-drug conjugates. Pharm. Res. 32, 3494–3507. doi:10.1007/
Brinkmann, U., and Kontermann, R. E. (2017). The making of bispecific antibodies.
s11095-015-1624-3
MAbs 9, 182–212. doi:10.1080/19420862.2016.1268307
Dan, N., Setua, S., Kashyap, V. K., Khan, S., Jaggi, M., Yallapu, M. M., et al. (2018).
Bulger, P. G., Conlon, D. A., Cink, R. D., Fernandez-Cerezo, L., Zhang, Q.,
Antibody-drug conjugates for cancer therapy: Chemistry to clinical implications.
Thirumalairajan, S., et al. (2023). Drug-linkers in antibody–drug conjugates:
Pharm. (Basel) 11, 32. doi:10.3390/ph11020032
Perspective on current industry Practices. Org. Process Res. Dev. 27, 1248–1257.
doi:10.1021/acs.oprd.3c00136 D’Atri, V., Pell, R., Clarke, A., Guillarme, D., and Fekete, S. (2019). Is hydrophobic
interaction chromatography the most suitable technique to characterize site-specific
Cao, M., De Mel, N., Jiao, Y., Howard, J., Parthemore, C., Korman, S., et al. (2019).
antibody-drug conjugates? J. Chromatogr. A 1586, 149–153. doi:10.1016/j.chroma.2018.
Site-specific antibody-drug conjugate heterogeneity characterization and heterogeneity
12.020
root cause analysis. MAbs 11, 1064–1076. doi:10.1080/19420862.2019.1624127
de Mel, N., Mulagapati, S. H. R., Cao, M., and Liu, D. (2019). A method to directly
Cardoso, F., Paluch-Shimon, S., Senkus, E., Curigliano, G., Aapro, M. S., André, F.,
analyze free-drug–related species in antibody-drug conjugates without sample
et al. (2020). 5th ESO-ESMO international consensus guidelines for advanced breast
preparation. J. Chromatogr. B 1116, 51–59. doi:10.1016/j.jchromb.2019.04.012
cancer (ABC 5). Ann. Oncol. 31, 1623–1649. doi:10.1016/j.annonc.2020.09.010
Dean, A. Q., Luo, S., Twomey, J. D., and Zhang, B. (2021). Targeting cancer with
Carter, P. J., and Senter, P. D. (2008). Antibody-drug conjugates for cancer therapy.
antibody-drug conjugates: Promises and challenges. MAbs 13, 1951427. doi:10.1080/
Cancer J. 14, 154–169. doi:10.1097/PPO.0b013e318172d704
19420862.2021.1951427
Casi, G., and Neri, D. (2012). Antibody-drug conjugates: basic concepts, examples and
Debaene, F., Boeuf, A., Wagner-Rousset, E., Colas, O., Ayoub, D., Corvaïa, N., et al.
future perspectives. J. Control Release 161, 422–428. doi:10.1016/j.jconrel.2012.01.026
(2014). Innovative native MS methodologies for antibody drug conjugate
Castelli, M. S., McGonigle, P., and Hornby, P. J. (2019). The pharmacology and characterization: High resolution native MS and IM-MS for average DAR and DAR
therapeutic applications of monoclonal antibodies. Pharmacol. Res. Perspect. 7, e00535. distribution assessment. Anal. Chem. 86, 10674–10683. doi:10.1021/ac502593n
doi:10.1002/prp2.535
Deeks, E. D. (2021). Disitamab vedotin: First approval. Drugs 81, 1929–1935. doi:10.
Ceci, C., Lacal, P. M., and Graziani, G. (2022). Antibody-drug conjugates: Resurgent 1007/s40265-021-01614-x
anticancer agents with multi-targeted therapeutic potential. Pharmacol. Ther. 236,
Deeks, E. D. (2019). Polatuzumab vedotin: First global approval. Drugs 79,
108106. doi:10.1016/j.pharmthera.2021.108106
1467–1475. doi:10.1007/s40265-019-01175-0
Chames, P., Van Regenmortel, M., Weiss, E., and Baty, D. (2009). Therapeutic
Del Prete, A., Salvi, V., Soriani, A., Laffranchi, M., Sozio, F., Bosisio, D., et al. (2023).
antibodies: successes, limitations and hopes for the future. Br. J. Pharmacol. 157,
Dendritic cell subsets in cancer immunity and tumor antigen sensing. Cell Mol.
220–233. doi:10.1111/j.1476-5381.2009.00190.x
Immunol. 20, 432–447. doi:10.1038/s41423-023-00990-6
Chang, E., Weinstock, C., Zhang, L., Charlab, R., Dorff, S. E., Gong, Y., et al. (2021). FDA
Dennis, M. S., Jin, H., Dugger, D., Yang, R., McFarland, L., Ogasawara, A., et al.
approval summary: Enfortumab vedotin for locally advanced or metastatic urothelial
(2007). Imaging tumors with an albumin-binding Fab, a novel tumor-targeting agent.
carcinoma. Clin. Cancer Res. 27, 922–927. doi:10.1158/1078-0432.CCR-20-2275
Cancer Res. 67, 254–261. doi:10.1158/0008-5472.CAN-06-2531
Chari, R. V. J. (2008). Targeted cancer therapy: conferring specificity to cytotoxic
drugs. Acc. Chem. Res. 41, 98–107. doi:10.1021/ar700108g Deonarain, M. P., and Xue, Q. (2020). Tackling solid tumour therapy with small-
format drug conjugates. Antib. Ther. 3, 237–245. doi:10.1093/abt/tbaa024
Chen, R., Hou, J., Newman, E., Kim, Y., Donohue, C., Liu, X., et al. (2015).
CD30 downregulation, MMAE resistance, and MDR1 upregulation are all associated Deonarain, M. P., Yahioglu, G., Stamati, I., Pomowski, A., Clarke, J., Edwards, B. M.,
with resistance to brentuximab vedotin. Mol. Cancer Ther. 14, 1376–1384. doi:10.1158/ et al. (2018). Small-format drug conjugates: A viable alternative to ADCs for solid
1535-7163.MCT-15-0036 tumours? Antibodies (Basel) 7, 16. doi:10.3390/antib7020016
Chen, T.-H., Yang, Y., Zhang, Z., Fu, C., Zhang, Q., Williams, J. D., et al. (2019). Diéras, V., Miles, D., Verma, S., Pegram, M., Welslau, M., Baselga, J., et al. (2017).
Native reversed-phase liquid chromatography: A technique for lcms of intact antibody- Trastuzumab emtansine versus capecitabine plus lapatinib in patients with previously
drug conjugates. Anal. Chem. 91, 2805–2812. doi:10.1021/acs.analchem.8b04699 treated HER2-positive advanced breast cancer (EMILIA): a descriptive analysis of final

Frontiers in Pharmacology 17 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

overall survival results from a randomised, open-label, phase 3 trial. Lancet Oncol. 18, Gerber, H.-P., Senter, P. D., and Grewal, I. S. (2009). Antibody drug-conjugates
732–742. doi:10.1016/S1470-2045(17)30312-1 targeting the tumor vasculature: Current and future developments. MAbs 1, 247–253.
doi:10.4161/mabs.1.3.8515
Dilawari, A., Shah, M., Ison, G., Gittleman, H., Fiero, M. H., Shah, A., et al. (2023).
FDA approval summary: Mirvetuximab soravtansine-gynx for FRα-positive, platinum- Gillard, G. O., Proctor, J. L., Brooks, M. L., Lamothe, T. L., Hyzy, S. L., McDonough, S.
resistant ovarian cancer. Clin. Cancer Res. 22, OF1–OF6. CCR-23-0991. doi:10.1158/ M., et al. (2020). A novel targeted approach to achieve immune system reset: CD45-
1078-0432.CCR-23-0991 Targeted antibody drug conjugates enable autologous HSCT and ameliorate disease in
preclinical autoimmune disease models. Biol. Blood Marrow Transplant. 26, S307–S308.
Domínguez-Llamas, S., Caro-Magdaleno, M., Mataix-Albert, B., Avilés-Prieto, J.,
doi:10.1016/j.bbmt.2019.12.407
Romero-Barranca, I., and Rodríguez-de-la-Rúa, E. (2023). Adverse events of
antibody–drug conjugates on the ocular surface in cancer therapy. Clin. Transl. Godwin, C. D., Bates, O. M., Jean, S. R., Laszlo, G. S., Garling, E. E., Beddoe, M. E.,
Oncol. 2023, 03261. doi:10.1007/s12094-023-03261-y et al. (2020). Anti-apoptotic BCL-2 family proteins confer resistance to calicheamicin-
based antibody-drug conjugate therapy of acute leukemia. Leuk. Lymphoma 61,
Donaghy, H. (2016). Effects of antibody, drug and linker on the preclinical and
2990–2994. doi:10.1080/10428194.2020.1786553
clinical toxicities of antibody-drug conjugates. MAbs 8, 659–671. doi:10.1080/19420862.
2016.1156829 Gogesch, P., Dudek, S., van Zandbergen, G., Waibler, Z., and Anzaghe, M. (2021). The
role of Fc receptors on the effectiveness of therapeutic monoclonal antibodies. Int.
Drago, J. Z., Modi, S., and Chandarlapaty, S. (2021). Unlocking the potential of
J. Mol. Sci. 22, 8947. doi:10.3390/ijms22168947
antibody–drug conjugates for cancer therapy. Nat. Rev. Clin. Oncol. 18, 327–344. doi:10.
1038/s41571-021-00470-8 Goldenberg, M. M. (1999). Trastuzumab, a recombinant DNA-derived humanized
monoclonal antibody, a novel agent for the treatment of metastatic breast cancer. Clin.
Ducry, L. (2012). Challenges in the development and manufacturing of antibody-drug
Ther. 21, 309–318. doi:10.1016/S0149-2918(00)88288-0
conjugates. Methods Mol. Biol. 899, 489–497. doi:10.1007/978-1-61779-921-1_29
Gomes-da-Silva, L. C., Kepp, O., and Kroemer, G. (2020). Regulatory approval of
Dumontet, C., Reichert, J. M., Senter, P. D., Lambert, J. M., and Beck, A. (2023).
photoimmunotherapy: photodynamic therapy that induces immunogenic cell death.
Antibody–drug conjugates come of age in oncology. Nat. Rev. Drug Discov. 22, 641–661.
Oncoimmunology 9, 1841393. doi:10.1080/2162402X.2020.1841393
doi:10.1038/s41573-023-00709-2
Gondi, C. S., and Rao, J. S. (2013). Cathepsin B as a cancer target. Expert Opin. Ther.
Edwards, E., Livanos, M., Krueger, A., Dell, A., Haslam, S. M., Mark Smales, C., et al.
Targets 17, 281–291. doi:10.1517/14728222.2013.740461
(2022). Strategies to control therapeutic antibody glycosylation during bioprocessing:
Synthesis and separation. Biotechnol. Bioeng. 119, 1343–1358. doi:10.1002/bit.28066 Goodson, H. V., and Jonasson, E. M. (2018). Microtubules and microtubule-
associated proteins. Cold Spring Harb. Perspect. Biol. 10, a022608. doi:10.1101/
Emens, L. A., Esteva, F. J., Beresford, M., Saura, C., Laurentiis, M. D., Kim, S.-B., et al.
cshperspect.a022608
(2020). Trastuzumab emtansine plus atezolizumab versus trastuzumab emtansine plus
placebo in previously treated, HER2-positive advanced breast cancer (KATE2): a phase Greenblatt, K., and Khaddour, K. (2023). Trastuzumab, in StatPearls (treasure island
2, multicentre, randomised, double-blind trial. Lancet Oncol. 21, 1283–1295. doi:10. (FL): StatPearls publishing). Available at: http://www.ncbi.nlm.nih.gov/books/
1016/S1470-2045(20)30465-4 NBK532246/ [Accessed August 29, 2023].
Estrela, J. M., Ortega, A., and Obrador, E. (2006). Glutathione in cancer biology and Hackshaw, M. D., Danysh, H. E., Singh, J., Ritchey, M. E., Ladner, A., Taitt, C., et al.
therapy. Crit. Rev. Clin. Lab. Sci. 43, 143–181. doi:10.1080/10408360500523878 (2020). Incidence of pneumonitis/interstitial lung disease induced by HER2-targeting
therapy for HER2-positive metastatic breast cancer. Breast Cancer Res. Treat. 183,
Fleming, R. (2020). ADC analysis by hydrophobic interaction chromatography.
23–39. doi:10.1007/s10549-020-05754-8
Methods Mol. Biol. 2078, 147–161. doi:10.1007/978-1-4939-9929-3_10
Hamadani, M., Collins, G. P., Caimi, P. F., Samaniego, F., Spira, A., Davies, A., et al.
Ferl, G. Z., Kenanova, V., Wu, A. M., and DiStefano, J. J. (2006). A two-tiered
(2021). Camidanlumab tesirine in patients with relapsed or refractory lymphoma: a
physiologically based model for dually labeled single-chain Fv-Fc antibody fragments.
phase 1, open-label, multicentre, dose-escalation, dose-expansion study. Lancet
Mol. Cancer Ther. 5, 1550–1558. doi:10.1158/1535-7163.MCT-06-0072
Haematol. 8, e433–e445. doi:10.1016/S2352-3026(21)00103-4
Ferraro, E., Drago, J. Z., and Modi, S. (2021). Implementing antibody-drug conjugates
Hamblett, K. J., Senter, P. D., Chace, D. F., Sun, M. M. C., Lenox, J., Cerveny, C. G.,
(ADCs) in HER2-positive breast cancer: state of the art and future directions. Breast
et al. (2004). Effects of drug loading on the antitumor activity of a monoclonal antibody
Cancer Res. 23, 84. doi:10.1186/s13058-021-01459-y
drug conjugate. Clin. Cancer Res. 10, 7063–7070. doi:10.1158/1078-0432.CCR-04-0789
Flynn, M. J., Zammarchi, F., Tyrer, P. C., Akarca, A. U., Janghra, N., Britten, C. E.,
Hartley, J. A. (2011). The development of pyrrolobenzodiazepines as antitumour
et al. (2016). ADCT-301, a pyrrolobenzodiazepine (PBD) dimer-containing antibody-
agents. Expert Opin. Investig. Drugs 20, 733–744. doi:10.1517/13543784.2011.573477
drug conjugate (ADC) targeting CD25-expressing hematological malignancies. Mol.
Cancer Ther. 15, 2709–2721. doi:10.1158/1535-7163.MCT-16-0233 Herbener, P., Schönfeld, K., König, M., Germer, M., Przyborski, J. M., Bernöster, K.,
et al. (2018). Functional relevance of in vivo half antibody exchange of an
Ford, C. H., Newman, C. E., Johnson, J. R., Woodhouse, C. S., Reeder, T. A., Rowland,
IgG4 therapeutic antibody-drug conjugate. PLoS One 13, e0195823. doi:10.1371/
G. F., et al. (1983). Localisation and toxicity study of a vindesine-anti-CEA conjugate in
journal.pone.0195823
patients with advanced cancer. Br. J. Cancer 47, 35–42. doi:10.1038/bjc.1983.4
Heitz, N., Greer, S. C., and Halford, Z. (2023). A review of Tisotumab vedotin-tftv in
Francisco, J. A., Cerveny, C. G., Meyer, D. L., Mixan, B. J., Klussman, K., Chace, D. F.,
recurrent or metastatic cervical cancer. Ann. Pharmacother. 57, 585–596. doi:10.1177/
et al. (2003). cAC10-vcMMAE, an anti-CD30-monomethyl auristatin E conjugate with
10600280221118370
potent and selective antitumor activity. Blood 102, 1458–1465. doi:10.1182/blood-2003-
01-0039 Heo, Y.-A. (2023). Mirvetuximab soravtansine: First approval. Drugs 83, 265–273.
doi:10.1007/s40265-023-01834-3
Fu, Y., and Ho, M. (2018). DNA damaging agent-based antibody-drug conjugates for
cancer therapy. Antib. Ther. 1, 33–43. doi:10.1093/abt/tby007 Herrera, A. F., Zain, J., Savage, K. J., Feldman, T. A., Brammer, J. E., Chen, L., et al.
(2021). Brentuximab vedotin plus cyclophosphamide, doxorubicin, etoposide, and
Fu, Z., Li, S., Han, S., Shi, C., and Zhang, Y. (2022). Antibody drug conjugate: the
prednisone (CHEP-BV) followed by BV consolidation in patients with CD30-
“biological missile” for targeted cancer therapy. Sig Transduct. Target Ther. 7, 93. doi:10.
expressing peripheral T-cell lymphomas. Blood 138, 133. doi:10.1182/blood-2021-
1038/s41392-022-00947-7
151105
Fuentes-Antrás, J., Genta, S., Vijenthira, A., and Siu, L. L. (2023). Antibody–drug
Hoffmann, R. M., Coumbe, B. G. T., Josephs, D. H., Mele, S., Ilieva, K. M., Cheung, A.,
conjugates: in search of partners of choice. Trends Cancer 9, 339–354. doi:10.1016/j.
et al. (2018). Antibody structure and engineering considerations for the design and
trecan.2023.01.003
function of Antibody Drug Conjugates (ADCs). Oncoimmunology 7, e1395127. doi:10.
Fukuda, Y., Bustos, M. A., Cho, S.-N., Roszik, J., Ryu, S., Lopez, V. M., et al. (2022). 1080/2162402X.2017.1395127
Interplay between soluble CD74 and macrophage-migration inhibitory factor drives
Hughes, B. (2010). Antibody-drug conjugates for cancer: poised to deliver? Nat. Rev.
tumor growth and influences patient survival in melanoma. Cell Death Dis. 13, 117.
Drug Discov. 9, 665–667. doi:10.1038/nrd3270
doi:10.1038/s41419-022-04552-y
Hurwitz, J., Haggstrom, L. R., and Lim, E. (2023). Antibody–drug conjugates:
Furlanetto, J., Marmé, F., and Loibl, S. (2022). Sacituzumab govitecan: past, present
Ushering in a new era of cancer therapy. Pharmaceutics 15, 2017. doi:10.3390/
and future of a new antibody-drug conjugate and future horizon. Future Oncol. 18,
pharmaceutics15082017
3199–3215. doi:10.2217/fon-2022-0407
Hutchinson, M. H., Hendricks, R. S., Lin, X. X., and Olson, D. A. (2018). “Chapter 40 -
Gajewski, T. F., Meng, Y., and Harlin, H. (2006). Immune suppression in the tumor
process development and manufacturing of antibody-drug conjugates,” in
microenvironment. J. Immunother. 29, 233–240. doi:10.1097/01.cji.0000199193.
Biopharmaceutical processing. Editors G. Jagschies, E. Lindskog, K. Łącki, and
29048.56
P. Galliher (Amsterdam: Elsevier), 813–836. doi:10.1016/B978-0-08-100623-8.00041-4
García-Alonso, S., Ocaña, A., and Pandiella, A. (2018). Resistance to antibody–drug
conjugates. Cancer Res. 78, 2159–2165. doi:10.1158/0008-5472.CAN-17-3671 Iqbal, N., and Iqbal, N. (2014). Human epidermal growth factor receptor 2 (HER2) in
cancers: Overexpression and therapeutic implications. Mol. Biol. Int. 2014, 852748.
Garrett, M., Ruiz-Garcia, A., Parivar, K., Hee, B., and Boni, J. (2019). Population doi:10.1155/2014/852748
pharmacokinetics of inotuzumab ozogamicin in relapsed/refractory acute
lymphoblastic leukemia and non-Hodgkin lymphoma. J. Pharmacokinet. Jain, N., Smith, S. W., Ghone, S., and Tomczuk, B. (2015). Current ADC linker
Pharmacodyn. 46, 211–222. doi:10.1007/s10928-018-9614-9 chemistry. Pharm. Res. 32, 3526–3540. doi:10.1007/s11095-015-1657-7

Frontiers in Pharmacology 18 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Jain, R. K. (2001). Delivery of molecular and cellular medicine to solid tumors. Adv. Leget, G. A., and Czuczman, M. S. (1998). Use of rituximab, the new FDA-approved
Drug Deliv. Rev. 46, 149–168. doi:10.1016/s0169-409x(00)00131-9 antibody. Curr. Opin. Oncol. 10, 548–551. doi:10.1097/00001622-199811000-00012
Jain, R. K. (2005). Normalization of tumor vasculature: an emerging concept in Lim, R. K. V., Yu, S., Cheng, B., Li, S., Kim, N.-J., Cao, Y., et al. (2015). Targeted
antiangiogenic therapy. Science 307, 58–62. doi:10.1126/science.1104819 delivery of LXR agonist using a site-specific antibody-drug conjugate. Bioconjug Chem.
26, 2216–2222. doi:10.1021/acs.bioconjchem.5b00203
Jiang, Q., Patel, B., Jin, X., Di Grandi, D., Bortell, E., Czapkowski, B., et al. (2019).
Structural characterization of the aggregates of gemtuzumab ozogamicin. ACS Omega 4, Lin, J., and Lazar, A. C. (2013). Determination of charge heterogeneity and level of
6468–6475. doi:10.1021/acsomega.8b03627 unconjugated antibody by imaged cIEF. Methods Mol. Biol. 1045, 295–302. doi:10.1007/
978-1-62703-541-5_19
Joubert, N., Beck, A., Dumontet, C., and Denevault-Sabourin, C. (2020).
Antibody–drug conjugates: The last decade. Pharmaceuticals 13, 245. doi:10.3390/ Lindley, C., McCune, J. S., Thomason, T. E., Lauder, D., Sauls, A., Adkins, S., et al.
ph13090245 (1999). Perception of chemotherapy side effects cancer versus noncancer patients.
Cancer Pract. 7, 59–65. doi:10.1046/j.1523-5394.1999.07205.x
Junttila, T. T., Li, G., Parsons, K., Phillips, G. L., and Sliwkowski, M. X. (2011).
Trastuzumab-DM1 (T-DM1) retains all the mechanisms of action of trastuzumab and Liu, L., and Chen, J. (2022). Therapeutic antibodies for precise cancer
efficiently inhibits growth of lapatinib insensitive breast cancer. Breast Cancer Res. immunotherapy: current and future perspectives. Med. Rev. 2, 555–569. doi:10.1515/
Treat. 128, 347–356. doi:10.1007/s10549-010-1090-x mr-2022-0033
Junutula, J. R., Raab, H., Clark, S., Bhakta, S., Leipold, D. D., Weir, S., et al. (2008). Liu, Y., Jiang, P., Capkova, K., Xue, D., Ye, L., Sinha, S. C., et al. (2011). Tissue factor-
Site-specific conjugation of a cytotoxic drug to an antibody improves the therapeutic activated coagulation cascade in the tumor microenvironment is critical for tumor
index. Nat. Biotechnol. 26, 925–932. doi:10.1038/nbt.1480 progression and an effective target for therapy. Cancer Res. 71, 6492–6502. doi:10.1158/
0008-5472.CAN-11-1145
Kang, C. (2021). Moxetumomab pasudotox in hairy cell leukaemia: A profile of its
use. Clin. Drug Investig. 41, 829–834. doi:10.1007/s40261-021-01069-8 Loganzo, F., Sung, M., and Gerber, H.-P. (2016). Mechanisms of resistance to
antibody–drug conjugates. Mol. Cancer Ther. 15, 2825–2834. doi:10.1158/1535-7163.
Kaplon, H., Crescioli, S., Chenoweth, A., Visweswaraiah, J., and Reichert, J. M. (2023).
MCT-16-0408
Antibodies to watch in 2023. MAbs 15, 2153410. doi:10.1080/19420862.2022.2153410
Loganzo, F., Tan, X., Sung, M., Jin, G., Myers, J. S., Melamud, E., et al. (2015). Tumor
Kaplon, H., Muralidharan, M., Schneider, Z., and Reichert, J. M. (2020). Antibodies to
cells chronically treated with a trastuzumab-maytansinoid antibody-drug conjugate
watch in 2020. MAbs 12, 1703531. doi:10.1080/19420862.2019.1703531
develop varied resistance mechanisms but respond to alternate treatments. Mol. Cancer
Katz, J., Janik, J. E., and Younes, A. (2011). Brentuximab vedotin (SGN-35). Clin. Ther. 14, 952–963. doi:10.1158/1535-7163.MCT-14-0862
Cancer Res. 17, 6428–6436. doi:10.1158/1078-0432.CCR-11-0488
Lu, J., Jiang, F., Lu, A., and Zhang, G. (2016). Linkers having a crucial role in antibody-
Kayser, S., and Levis, M. J. (2022). Updates on targeted therapies for acute myeloid drug conjugates. Int. J. Mol. Sci. 17, 561. doi:10.3390/ijms17040561
leukaemia. Br. J. Haematol. 196, 316–328. doi:10.1111/bjh.17746
Lucas, A. T., Price, L. S. L., Schorzman, A. N., Storrie, M., Piscitelli, J. A., Razo, J., et al.
Kholodenko, R. V., Kalinovsky, D. V., Doronin, I. I., Ponomarev, E. D., and (2018). Factors affecting the pharmacology of antibody-drug conjugates. Antibodies
Kholodenko, I. V. (2019). Antibody fragments as potential biopharmaceuticals for (Basel) 7, 10. doi:10.3390/antib7010010
cancer therapy: Success and limitations. CMC 26, 396–426. doi:10.2174/
Luo, R., Liu, H., and Cheng, Z. (2022). Protein scaffolds: antibody alternatives for
0929867324666170817152554
cancer diagnosis and therapy. RSC Chem. Biol. 3, 830–847. doi:10.1039/D2CB00094F
Khongorzul, P., Ling, C. J., Khan, F. U., Ihsan, A. U., and Zhang, J. (2020).
Ma, Z., Wang, H., Cai, Y., Wang, H., Niu, K., Wu, X., et al. (2018). Epigenetic drift of
Antibody–drug conjugates: A comprehensive review. Mol. Cancer Res. 18, 3–19.
H3K27me3 in aging links glycolysis to healthy longevity in Drosophila. eLife 7, e35368.
doi:10.1158/1541-7786.MCR-19-0582
doi:10.7554/eLife.35368
Kim, E. G., and Kim, K. M. (2015). Strategies and advancement in antibody-drug
Macor, P., Capolla, S., and Tedesco, F. (2018). Complement as a biological tool to
conjugate optimization for targeted cancer therapeutics. Biomol. Ther. Seoul. 23,
control tumor growth. Front. Immunol. 9, 2203. doi:10.3389/fimmu.2018.02203
493–509. doi:10.4062/biomolther.2015.116
Macor, P., Mezzanzanica, D., Cossetti, C., Alberti, P., Figini, M., Canevari, S., et al.
Kimiz-Gebologlu, I., Gulce-Iz, S., and Biray-Avci, C. (2018). Monoclonal antibodies in
(2006). Complement activated by chimeric anti-folate receptor antibodies is an efficient
cancer immunotherapy. Mol. Biol. Rep. 45, 2935–2940. doi:10.1007/s11033-018-4427-x
effector system to control ovarian carcinoma. Cancer Res. 66, 3876–3883. doi:10.1158/
Kitamura, N., Sento, S., Yoshizawa, Y., Sasabe, E., Kudo, Y., and Yamamoto, T. (2020). 0008-5472.CAN-05-3434
Current trends and future prospects of molecular targeted therapy in head and neck
Macor, P., and Tedesco, F. (2007). Complement as effector system in cancer
squamous cell carcinoma. Int. J. Mol. Sci. 22, 240. doi:10.3390/ijms22010240
immunotherapy. Immunol. Lett. 111, 6–13. doi:10.1016/j.imlet.2007.04.014
Kostova, V., Désos, P., Starck, J.-B., and Kotschy, A. (2021). The chemistry behind
Mahadevan, D., and Von Hoff, D. D. (2007). Tumor-stroma interactions in
ADCs. Pharm. (Basel) 14, 442. doi:10.3390/ph14050442
pancreatic ductal adenocarcinoma. Mol. Cancer Ther. 6, 1186–1197. doi:10.1158/
Kreitman, R. J. (2019). Hairy cell leukemia: present and future directions. Leuk. 1535-7163.MCT-06-0686
Lymphoma 60, 2869–2879. doi:10.1080/10428194.2019.1608536
Mahalingaiah, P. K., Ciurlionis, R., Durbin, K. R., Yeager, R. L., Philip, B. K., Bawa, B., et al.
Kreitman, R. J., and Pastan, I. (2011). Antibody fusion proteins: anti-CD22 (2019). Potential mechanisms of target-independent uptake and toxicity of antibody-drug
recombinant immunotoxin moxetumomab pasudotox. Clin. Cancer Res. 17, conjugates. Pharmacol. Ther. 200, 110–125. doi:10.1016/j.pharmthera.2019.04.008
6398–6405. doi:10.1158/1078-0432.CCR-11-0487
Mahmood, I. (2021). Clinical pharmacology of antibody-drug conjugates. Antibodies
Kvirkvelia, N., McMenamin, M., Gutierrez, V. I., Lasareishvili, B., and Madaio, M. P. (Basel) 10, 20. doi:10.3390/antib10020020
(2015). Human anti-α3(IV)NC1 antibody drug conjugates target glomeruli to resolve
Malhotra, V., and Perry, M. C. (2003). Classical chemotherapy: mechanisms,
nephritis. Am. J. Physiol. Ren. Physiol. 309, F680–F684. doi:10.1152/ajprenal.00289.2015
toxicities and the therapeutic window. Cancer Biol. Ther. 2, S2–S4.
Labani-Motlagh, A., Ashja-Mahdavi, M., and Loskog, A. (2020). The tumor
Mamounas, E. P., Untch, M., Mano, M. S., Huang, C.-S., Geyer, C. E., von Minckwitz,
microenvironment: A milieu hindering and obstructing antitumor immune
G., et al. (2021). Adjuvant T-DM1 versus trastuzumab in patients with residual invasive
responses. Front. Immunol. 11, 940. doi:10.3389/fimmu.2020.00940
disease after neoadjuvant therapy for HER2-positive breast cancer: subgroup analyses
Lambert, J. M., and Chari, R. V. J. (2014). Ado-trastuzumab emtansine (T-DM1): an from KATHERINE. Ann. Oncol. 32, 1005–1014. doi:10.1016/j.annonc.2021.04.011
antibody-drug conjugate (ADC) for HER2-positive breast cancer. J. Med. Chem. 57,
Marei, H. E., Cenciarelli, C., and Hasan, A. (2022). Potential of antibody–drug
6949–6964. doi:10.1021/jm500766w
conjugates (ADCs) for cancer therapy. Cancer Cell Int. 22, 255. doi:10.1186/s12935-
Lambert, J. M., and Morris, C. Q. (2017). Antibody–drug conjugates (ADCs) for 022-02679-8
personalized treatment of solid tumors: A review. Adv. Ther. 34, 1015–1035. doi:10.
Masters, J. C., Nickens, D. J., Xuan, D., Shazer, R. L., and Amantea, M. (2018). Clinical
1007/s12325-017-0519-6
toxicity of antibody drug conjugates: a meta-analysis of payloads. Invest. New Drugs 36,
Lanza, F., Maffini, E., Rondoni, M., Massari, E., Faini, A. C., and Malavasi, F. (2020). 121–135. doi:10.1007/s10637-017-0520-6
CD22 expression in B-cell acute lymphoblastic leukemia: Biological significance and
Matsumoto, T., Jimi, S., Hara, S., Takamatsu, Y., Suzumiya, J., and Tamura, K. (2012).
implications for inotuzumab therapy in adults. Cancers (Basel) 12, 303. doi:10.3390/
Importance of inducible multidrug resistance 1 expression in HL-60 cells resistant to
cancers12020303
gemtuzumab ozogamicin. Leuk. Lymphoma 53, 1399–1405. doi:10.3109/10428194.
Le, Q., Tang, T., Leonti, A., Castro, S., McKay, C. N., Perkins, L., et al. (2023). 2012.656102
Preclinical studies targeting CD74 with STRO-001 antibody-drug conjugate in acute
Mckertish, C. M., and Kayser, V. (2021). Advances and limitations of antibody drug
leukemia. Blood Adv. 7, 1666–1670. doi:10.1182/bloodadvances.2022008303
conjugates for cancer. Biomedicines 9, 872. doi:10.3390/biomedicines9080872
Lee, A. (2021). Loncastuximab tesirine: First approval. Drugs 81, 1229–1233. doi:10. Mecklenburg, L. (2018). A brief introduction to antibody-drug conjugates for
1007/s40265-021-01550-w toxicologic pathologists. Toxicol. Pathol. 46, 746–752. doi:10.1177/0192623318803059
Lee, H., Bhang, S. H., Lee, J. H., Kim, H., and Hahn, S. K. (2017). Tocilizumab- Mehanna, J., Haddad, F. G., Eid, R., Lambertini, M., and Kourie, H. R. (2019). Triple-
alendronate conjugate for treatment of rheumatoid arthritis. Bioconjug Chem. 28, negative breast cancer: current perspective on the evolving therapeutic landscape. Int.
1084–1092. doi:10.1021/acs.bioconjchem.7b00008 J. Womens Health 11, 431–437. doi:10.2147/IJWH.S178349

Frontiers in Pharmacology 19 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Modi, S., Jacot, W., Yamashita, T., Sohn, J., Vidal, M., Tokunaga, E., et al. (2022). Redman, J. M., Hill, E. M., AlDeghaither, D., and Weiner, L. M. (2015). Mechanisms
Trastuzumab deruxtecan in previously treated HER2-low advanced breast cancer. N. of action of therapeutic antibodies for cancer. Mol. Immunol. 67, 28–45. doi:10.1016/j.
Engl. J. Med. 387, 9–20. doi:10.1056/NEJMoa2203690 molimm.2015.04.002
Modi, S., Saura, C., Yamashita, T., Park, Y. H., Kim, S.-B., Tamura, K., et al. (2021). Richards, D. A. (2018). Exploring alternative antibody scaffolds: Antibody fragments
Abstract PD3-06: Updated results from DESTINY-breast01, a phase 2 trial of and antibody mimics for targeted drug delivery. Drug Discov. Today Technol. 30, 35–46.
trastuzumab deruxtecan (T-DXd) in HER2 positive metastatic breast cancer. Cancer doi:10.1016/j.ddtec.2018.10.005
Res. 81, PD3–06. doi:10.1158/1538-7445.SABCS20-PD3-06
Ríos-Luci, C., García-Alonso, S., Díaz-Rodríguez, E., Nadal-Serrano, M., Arribas, J.,
Modi, S., Saura, C., Yamashita, T., Park, Y. H., Kim, S.-B., Tamura, K., et al. (2020). Ocaña, A., et al. (2017). Resistance to the antibody-drug conjugate T-DM1 is based in a
Trastuzumab deruxtecan in previously treated HER2-positive breast cancer. N. Engl. reduction in lysosomal proteolytic activity. Cancer Res. 77, 4639–4651. doi:10.1158/
J. Med. 382, 610–621. doi:10.1056/NEJMoa1914510 0008-5472.CAN-16-3127
Nader-Marta, G., Martins-Branco, D., Agostinetto, E., Bruzzone, M., Ceppi, M., Sabbaghi, M., Gil-Gómez, G., Guardia, C., Servitja, S., Arpí, O., García-Alonso, S.,
Danielli, L., et al. (2022). Efficacy of tyrosine kinase inhibitors for the treatment of et al. (2017). Defective cyclin B1 induction in trastuzumab-emtansine (T-DM1)
patients with HER2-positive breast cancer with brain metastases: a systematic review acquired resistance in HER2-positive breast cancer. Clin. Cancer Res. 23, 7006–7019.
and meta-analysis. ESMO Open 7, 100501. doi:10.1016/j.esmoop.2022.100501 doi:10.1158/1078-0432.CCR-17-0696
Natsume, A., Niwa, R., and Satoh, M. (2009). Improving effector functions of Salem, A., Alotaibi, M., Mroueh, R., Basheer, H. A., and Afarinkia, K. (2021). CCR7 as
antibodies for cancer treatment: Enhancing ADCC and CDC. Drug Des. Devel Ther. a therapeutic target in Cancer. Biochim. Biophys. Acta Rev. Cancer 1875, 188499. doi:10.
3, 7–16. doi:10.2147/dddt.s4378 1016/j.bbcan.2020.188499
Nejadmoghaddam, M.-R., Minai-Tehrani, A., Ghahremanzadeh, R., Mahmoudi, M., Samantasinghar, A., Sunildutt, N. P., Ahmed, F., Soomro, A. M., Salih, A. R. C.,
Dinarvand, R., and Zarnani, A.-H. (2019). Antibody-drug conjugates: Possibilities and Parihar, P., et al. (2023). A comprehensive review of key factors affecting the efficacy of
challenges. Avicenna J. Med. Biotechnol. 11, 3–23. antibody drug conjugate. Biomed. Pharmacother. 161, 114408. doi:10.1016/j.biopha.
2023.114408
Nguyen, T. D., Bordeau, B. M., and Balthasar, J. P. (2023). Mechanisms of ADC
toxicity and strategies to increase ADC tolerability. Cancers (Basel) 15, 713. doi:10.3390/ Sau, S., Alsaab, H. O., Kashaw, S. K., Tatiparti, K., and Iyer, A. K. (2017). Advances in
cancers15030713 antibody-drug conjugates: A new era of targeted cancer therapy. Drug Discov. Today 22,
1547–1556. doi:10.1016/j.drudis.2017.05.011
Nicolò, E., Giugliano, F., Ascione, L., Tarantino, P., Corti, C., Tolaney, S. M., et al.
(2022). Combining antibody-drug conjugates with immunotherapy in solid tumors: Schmidhalter, D. R., Elzner, S., and Schmid, R. (2019). “Progress in the development
current landscape and future perspectives. Cancer Treat. Rev. 106, 102395. doi:10.1016/ of single-use solutions in antibody–drug conjugate (ADC) manufacturing,” in
j.ctrv.2022.102395 Single-use technology in biopharmaceutical manufacture (New Jersey: John Wiley &
Sons, Ltd), 303–310. doi:10.1002/9781119477891.ch27
Nolting, B. (2013). Linker technologies for antibody-drug conjugates. Methods Mol.
Biol. 1045, 71–100. doi:10.1007/978-1-62703-541-5_5 Schwartz, R. S. (2004). Paul Ehrlich’s magic bullets. N. Engl. J. Med. 350, 1079–1080.
doi:10.1056/NEJMp048021
Norsworthy, K. J., Ko, C.-W., Lee, J. E., Liu, J., John, C. S., Przepiorka, D., et al. (2018). FDA
approval summary: Mylotarg for treatment of patients with relapsed or refractory CD33-positive Scott, L. J. (2017). Brentuximab vedotin: A review in CD30-positive Hodgkin
acute myeloid leukemia. Oncologist 23, 1103–1108. doi:10.1634/theoncologist.2017-0604 lymphoma. Drugs 77, 435–445. doi:10.1007/s40265-017-0705-5
Ouyang, J. (2013). Drug-to-antibody ratio (DAR) and drug load distribution by Seaman, S., Zhu, Z., Saha, S., Zhang, X. M., Yang, M. Y., Hilton, M. B., et al.
hydrophobic interaction chromatography and reversed phase high-performance liquid (2017). Eradication of tumors through simultaneous ablation of CD276/B7-H3-
chromatography. Methods Mol. Biol. 1045, 275–283. doi:10.1007/978-1-62703-541-5_17 positive tumor cells and tumor vasculature. Cancer Cell 31, 501–515. doi:10.1016/j.
ccell.2017.03.005
O’Leary, M. K., Ahmed, A., and Alabi, C. A. (2023). Development of host-cleavable
antibody-bactericide conjugates against extracellular pathogens. ACS Infect. Dis. 9, Seckinger, A., Delgado, J. A., Moser, S., Moreno, L., Neuber, B., Grab, A., et al. (2017).
322–329. doi:10.1021/acsinfecdis.2c00492 Target expression, generation, preclinical activity, and pharmacokinetics of the BCMA-
T cell bispecific antibody EM801 for multiple myeloma treatment. Cancer Cell 31,
Omura, G., Honma, Y., Matsumoto, Y., Shinozaki, T., Itoyama, M., Eguchi, K., et al.
396–410. doi:10.1016/j.ccell.2017.02.002
(2023). Transnasal photoimmunotherapy with cetuximab sarotalocan sodium:
Outcomes on the local recurrence of nasopharyngeal squamous cell carcinoma. Sehn, L. H., Hertzberg, M., Opat, S., Herrera, A. F., Assouline, S., Flowers, C. R., et al.
Auris Nasus Larynx 50, 641–645. doi:10.1016/j.anl.2022.06.004 (2022). Polatuzumab vedotin plus bendamustine and rituximab in relapsed/refractory
DLBCL: survival update and new extension cohort data. Blood Adv. 6, 533–543. doi:10.
Park, M.-H., Lee, B. I., Byeon, J.-J., Shin, S.-H., Choi, J., Park, Y., et al. (2019).
1182/bloodadvances.2021005794
Pharmacokinetic and metabolism studies of monomethyl auristatin F via liquid
chromatography-quadrupole-time-of-flight mass spectrometry. Molecules 24, 2754. Senapati, S., Mahanta, A. K., Kumar, S., and Maiti, P. (2018). Controlled drug delivery
doi:10.3390/molecules24152754 vehicles for cancer treatment and their performance. Signal Transduct. Target Ther. 3, 7.
doi:10.1038/s41392-017-0004-3
Perrino, E., Steiner, M., Krall, N., Bernardes, G. J. L., Pretto, F., Casi, G., et al. (2014).
Curative properties of noninternalizing antibody-drug conjugates based on Senter, P. D. (2009). Potent antibody drug conjugates for cancer therapy. Curr. Opin.
maytansinoids. Cancer Res. 74, 2569–2578. doi:10.1158/0008-5472.CAN-13-2990 Chem. Biol. 13, 235–244. doi:10.1016/j.cbpa.2009.03.023
Peters, C., and Brown, S. (2015). Antibody–drug conjugates as novel anti-cancer Senter, P. D., and Sievers, E. L. (2012). The discovery and development of
chemotherapeutics. Biosci. Rep. 35, e00225. doi:10.1042/BSR20150089 brentuximab vedotin for use in relapsed Hodgkin lymphoma and systemic
anaplastic large cell lymphoma. Nat. Biotechnol. 30, 631–637. doi:10.1038/nbt.2289
Pettinato, M. C. (2021). Introduction to antibody-drug conjugates. Antibodies 10, 42.
doi:10.3390/antib10040042 Shefet-Carasso, L., and Benhar, I. (2015). Antibody-targeted drugs and drug
resistance--challenges and solutions. Drug Resist Updat 18, 36–46. doi:10.1016/j.
Ponte, J. F., Ab, O., Lanieri, L., Lee, J., Coccia, J., Bartle, L. M., et al. (2016).
drup.2014.11.001
Mirvetuximab soravtansine (IMGN853), a folate receptor alpha-targeting antibody-
drug conjugate, potentiates the activity of standard of care therapeutics in ovarian Shi, F., Liu, Y., Zhou, X., Shen, P., Xue, R., and Zhang, M. (2022). Disitamab vedotin: a
cancer models. Neoplasia 18, 775–784. doi:10.1016/j.neo.2016.11.002 novel antibody-drug conjugates for cancer therapy. Drug Deliv. 29, 1335–1344. doi:10.
1080/10717544.2022.2069883
Ponziani, S., Di Vittorio, G., Pitari, G., Cimini, A. M., Ardini, M., Gentile, R., et al.
(2020). Antibody-drug conjugates: The new frontier of chemotherapy. Int. J. Mol. Sci. Shiravand, Y., Khodadadi, F., Kashani, S. M. A., Hosseini-Fard, S. R., Hosseini, S.,
21, 5510. doi:10.3390/ijms21155510 Sadeghirad, H., et al. (2022). Immune checkpoint inhibitors in cancer therapy. Curr.
Oncol. 29, 3044–3060. doi:10.3390/curroncol29050247
Professional Committee on Clinical Research of Oncology Drugs, Expert Committee
for Monitoring the Clinical Application of Antitumor Drugs, Chinese Anti-Cancer Shitara, K., Baba, E., Fujitani, K., Oki, E., Fujii, S., and Yamaguchi, K. (2021).
Association, and Cancer Chemotherapy Quality Control Expert Committee of Beijing Discovery and development of trastuzumab deruxtecan and safety management for
Cancer Treatment Quality Control and Improvement Center (2022). Expert consensus patients with HER2-positive gastric cancer. Gastric Cancer 24, 780–789. doi:10.1007/
on the clinical application of antibody-drug conjugates in the treatment of malignant s10120-021-01196-3
tumors (2021 edition). Cancer Innov. 1, 3–24. doi:10.1002/cai2.8
Some, D., Amartely, H., Tsadok, A., and Lebendiker, M. (2019). Characterization of
Purcell, J. W., Tanlimco, S. G., Hickson, J., Fox, M., Sho, M., Durkin, L., et al. (2018). proteins by size-exclusion chromatography coupled to multi-angle light scattering
LRRC15 is a novel mesenchymal protein and stromal target for antibody–drug (SEC-MALS). J. Vis. Exp. 2019, 59615. doi:10.3791/59615
conjugates. Cancer Res. 78, 4059–4072. doi:10.1158/0008-5472.CAN-18-0327
Spiess, C., Bevers, J., Jackman, J., Chiang, N., Nakamura, G., Dillon, M., et al. (2013).
Ritchie, M., Tchistiakova, L., and Scott, N. (2013). Implications of receptor-mediated Development of a human IgG4 bispecific antibody for dual targeting of interleukin-4
endocytosis and intracellular trafficking dynamics in the development of antibody drug (IL-4) and interleukin-13 (IL-13) cytokines. J. Biol. Chem. 288, 26583–26593. doi:10.
conjugates. MAbs 5, 13–21. doi:10.4161/mabs.22854 1074/jbc.M113.480483
Rassy, E., Rached, L., and Pistilli, B. (2022). Antibody drug conjugates targeting Staudacher, A. H., and Brown, M. P. (2017). Antibody drug conjugates and bystander
HER2: Clinical development in metastatic breast cancer. Breast 66, 217–226. doi:10. killing: is antigen-dependent internalisation required? Br. J. Cancer 117, 1736–1742.
1016/j.breast.2022.10.016 doi:10.1038/bjc.2017.367

Frontiers in Pharmacology 20 frontiersin.org


Riccardi et al. 10.3389/fphar.2023.1274088

Stapleton, N. M., Andersen, J. T., Stemerding, A. M., Bjarnarson, S. P., Verheul, R. C., trastuzumab-drug conjugates: Future perspectives beyond HER2-positive breast
Gerritsen, J., et al. (2011). Competition for FcRn-mediated transport gives rise to short cancer. Cancer Treat. Rev. 113, 102500. doi:10.1016/j.ctrv.2022.102500
half-life of human IgG3 and offers therapeutic potential. Nat. Commun. 2, 599. doi:10.
von Minckwitz, G., Huang, C.-S., Mano, M. S., Loibl, S., Mamounas, E. P., Untch, M.,
1038/ncomms1608
et al. (2019). Trastuzumab emtansine for residual invasive HER2-positive breast cancer.
Strop, P., Delaria, K., Foletti, D., Witt, J. M., Hasa-Moreno, A., Poulsen, K., et al. N. Engl. J. Med. 380, 617–628. doi:10.1056/NEJMoa1814017
(2015). Site-specific conjugation improves therapeutic index of antibody drug
Vozella, F., Fazio, F., Lapietra, G., Petrucci, M. T., Martinelli, G., and Cerchione, C.
conjugates with high drug loading. Nat. Biotechnol. 33, 694–696. doi:10.1038/nbt.3274
(2021). Monoclonal antibodies in multiple myeloma. Panminerva Med. 63, 21–27.
Su, Z., Xiao, D., Xie, F., Liu, L., Wang, Y., Fan, S., et al. (2021). Antibody–drug doi:10.23736/S0031-0808.20.04149-X
conjugates: Recent advances in linker chemistry. Acta Pharm. Sin. B 11, 3889–3907.
Vu, T., and Claret, F. X. (2012). Trastuzumab: updated mechanisms of action and
doi:10.1016/j.apsb.2021.03.042
resistance in breast cancer. Front. Oncol. 2, 62. doi:10.3389/fonc.2012.00062
Sun Kang, M., See Kong, T. W., Xin Khoo, J. Y., and Loh, T.-P. (2021). Recent
Wagh, A., Song, H., Zeng, M., Tao, L., and Das, T. K. (2018). Challenges and new
developments in chemical conjugation strategies targeting native amino acids in
frontiers in analytical characterization of antibody-drug conjugates. MAbs 10, 222–243.
proteins and their applications in antibody–drug conjugates. Chem. Sci. 12,
doi:10.1080/19420862.2017.1412025
13613–13647. doi:10.1039/D1SC02973H
Wagner, E., Colas, O., Chenu, S., Goyon, A., Murisier, A., Cianferani, S., et al. (2020).
Sung, H., Ferlay, J., Siegel, R. L., Laversanne, M., Soerjomataram, I., Jemal, A., et al.
Determination of size variants by CE-SDS for approved therapeutic antibodies: Key
(2021). Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality
implications of subclasses and light chain specificities. J. Pharm. Biomed. Anal. 184,
worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249. doi:10.3322/
113166. doi:10.1016/j.jpba.2020.113166
caac.21660
Waight, A. B., Bargsten, K., Doronina, S., Steinmetz, M. O., Sussman, D., and Prota, A.
Szot, C., Saha, S., Zhang, X. M., Zhu, Z., Hilton, M. B., Morris, K., et al. (2018). Tumor
E. (2016). Structural basis of microtubule destabilization by potent auristatin anti-
stroma–targeted antibody-drug conjugate triggers localized anticancer drug release.
mitotics. PLoS One 11, e0160890. doi:10.1371/journal.pone.0160890
J. Clin. Invest. 128, 2927–2943. doi:10.1172/JCI120481
Wang, Q., Shao, X., Zhang, Y., Zhu, M., Wang, F. X. C., Mu, J., et al. (2023). Role of
Tai, Y.-T., Mayes, P. A., Acharya, C., Zhong, M. Y., Cea, M., Cagnetta, A., et al. (2014).
tumor microenvironment in cancer progression and therapeutic strategy. Cancer Med.
Novel anti-B-cell maturation antigen antibody-drug conjugate (GSK2857916)
12, 11149–11165. doi:10.1002/cam4.5698
selectively induces killing of multiple myeloma. Blood 123, 3128–3138. doi:10.1182/
blood-2013-10-535088 Wang, Z., Li, H., Gou, L., Li, W., and Wang, Y. (2023). Antibody–drug conjugates: Recent
advances in payloads. Acta Pharm. Sin. B 2023, 15. doi:10.1016/j.apsb.2023.06.015
Talotta, R., Rucci, F., Canti, G., and Scaglione, F. (2019). Pros and cons of the
immunogenicity of monoclonal antibodies in cancer treatment: a lesson from Wedam, S., Fashoyin-Aje, L., Gao, X., Bloomquist, E., Tang, S., Sridhara, R., et al.
autoimmune diseases. Immunotherapy 11, 241–254. doi:10.2217/imt-2018-0081 (2020). FDA approval summary: Ado-trastuzumab emtansine for the adjuvant
treatment of HER2-positive early breast cancer. Clin. Cancer Res. 26, 4180–4185.
Tang, H., Liu, Y., Yu, Z., Sun, M., Lin, L., Liu, W., et al. (2019). The analysis of key
doi:10.1158/1078-0432.CCR-19-3980
factors related to ADCs structural design. Front. Pharmacol. 10, 373. doi:10.3389/fphar.
2019.00373 Weiner, L. M., Dhodapkar, M. V., and Ferrone, S. (2009). Monoclonal antibodies for
cancer immunotherapy. Lancet 373, 1033–1040. doi:10.1016/S0140-6736(09)60251-8
Tarantino, P., Ricciuti, B., Pradhan, S. M., and Tolaney, S. M. (2023). Optimizing the
safety of antibody–drug conjugates for patients with solid tumours. Nat. Rev. Clin. Wojtukiewicz, M. Z., Rek, M. M., Karpowicz, K., Górska, M., Polityńska, B.,
Oncol. 20, 558–576. doi:10.1038/s41571-023-00783-w Wojtukiewicz, A. M., et al. (2021). Inhibitors of immune checkpoints—PD-1, PD-
L1, CTLA-4—New opportunities for cancer patients and a new challenge for internists
Thurber, G. M., and Dane Wittrup, K. (2012). A mechanistic compartmental model
and general practitioners. Cancer Metastasis Rev. 40, 949–982. doi:10.1007/s10555-021-
for total antibody uptake in tumors. J. Theor. Biol. 314, 57–68. doi:10.1016/j.jtbi.2012.
09976-0
08.034
Xiao, Y., and Yu, D. (2021). Tumor microenvironment as a therapeutic target in
Thurber, G. M., Schmidt, M. M., and Wittrup, K. D. (2008). Antibody tumor
cancer. Pharmacol. Ther. 221, 107753. doi:10.1016/j.pharmthera.2020.107753
penetration: transport opposed by systemic and antigen-mediated clearance. Adv.
Drug Deliv. Rev. 60, 1421–1434. doi:10.1016/j.addr.2008.04.012 Xu, S. (2015). Internalization, trafficking, intracellular processing and actions of
antibody-drug conjugates. Pharm. Res. 32, 3577–3583. doi:10.1007/s11095-015-1729-8
Tilly, H., Morschhauser, F., Sehn, L. H., Friedberg, J. W., Trněný, M., Sharman, J. P.,
et al. (2022). Polatuzumab vedotin in previously untreated diffuse large B-cell Yao, P., Zhang, Y., Zhang, S., Wei, X., Liu, Y., Du, C., et al. (2023). Knowledge atlas of
lymphoma. N. Engl. J. Med. 386, 351–363. doi:10.1056/NEJMoa2115304 antibody-drug conjugates on CiteSpace and clinical trial visualization analysis. Front.
Oncol. 12, 1039882. doi:10.3389/fonc.2022.1039882
Tipton, T. R. W., Roghanian, A., Oldham, R. J., Carter, M. J., Cox, K. L., Mockridge, C.
I., et al. (2015). Antigenic modulation limits the effector cell mechanisms employed by Yasunaga, M., Manabe, S., and Matsumura, Y. (2017). Immunoregulation by IL-7R-
type I anti-CD20 monoclonal antibodies. Blood 125, 1901–1909. doi:10.1182/blood- targeting antibody-drug conjugates: overcoming steroid-resistance in cancer and
2014-07-588376 autoimmune disease. Sci. Rep. 7, 10735. doi:10.1038/s41598-017-11255-4
Tong, J. T. W., Harris, P. W. R., Brimble, M. A., and Kavianinia, I. (2021). An insight Yogev, S., Cooper, R., Fetter, R., Horowitz, M., and Shen, K. (2016). Microtubule
into FDA approved antibody-drug conjugates for cancer therapy. Molecules 26, 5847. organization determines axonal transport dynamics. Neuron 92, 449–460. doi:10.1016/j.
doi:10.3390/molecules26195847 neuron.2016.09.036
Tsuchikama, K., and An, Z. (2018). Antibody-drug conjugates: recent advances in Yu, J., Song, Y., and Tian, W. (2020). How to select IgG subclasses in developing anti-tumor
conjugation and linker chemistries. Protein Cell 9, 33–46. doi:10.1007/s13238-016- therapeutic antibodies. J. Hematol. Oncol. 13, 45. doi:10.1186/s13045-020-00876-4
0323-0
Zahavi, D., and Weiner, L. (2020). Monoclonal antibodies in cancer therapy.
Tvilum, A., Johansen, M. I., Glud, L. N., Ivarsen, D. M., Khamas, A. B., Carmali, S., Antibodies (Basel) 9, 34. doi:10.3390/antib9030034
et al. (2023). Antibody-drug conjugates to treat bacterial biofilms via targeting and
Zammarchi, F., Corbett, S., Adams, L., Tyrer, P. C., Kiakos, K., Janghra, N., et al.
extracellular drug release. Adv. Sci. (Weinh) 10, e2301340. doi:10.1002/advs.202301340
(2018). ADCT-402, a PBD dimer-containing antibody drug conjugate targeting CD19-
van der Velden, V. H. J., Boeckx, N., Jedema, I., te Marvelde, J. G., Hoogeveen, P. G., expressing malignancies. Blood 131, 1094–1105. doi:10.1182/blood-2017-10-813493
Boogaerts, M., et al. (2004). High CD33-antigen loads in peripheral blood limit the
efficacy of gemtuzumab ozogamicin (Mylotarg) treatment in acute myeloid leukemia Zhang, J., Woods, C., He, F., Han, M., Treuheit, M. J., and Volkin, D. B. (2018).
patients. Leukemia 18, 983–988. doi:10.1038/sj.leu.2403350 Structural changes and aggregation mechanisms of two different dimers of an
IgG2 monoclonal antibody. Biochemistry 57, 5466–5479. doi:10.1021/acs.biochem.
van Dongen, G. A. M. S. (2021). Improving tumor penetration of antibodies and 8b00575
antibody-drug conjugates: Taking away the barriers for trojan horses. Cancer Res. 81,
3956–3957. doi:10.1158/0008-5472.CAN-21-0952 Zhao, P., Zhang, Y., Li, W., Jeanty, C., Xiang, G., and Dong, Y. (2020). Recent
advances of antibody drug conjugates for clinical applications. Acta Pharm. Sin. B 10,
Varma, G., Wang, J., and Diefenbach, C. (2022). Polatuzumab vedotin in relapsed/ 1589–1600. doi:10.1016/j.apsb.2020.04.012
refractory aggressive B-cell lymphoma. Expert Rev. Anticancer Ther. 22, 795–803.
doi:10.1080/14737140.2022.2093191 Zheng, Y., Ma, L., Sun, Q., and Tian, L. (2021). Clinically-relevant ABC transporter
for anti-cancer drug resistance. Front. Pharmacol. 12, 648407. doi:10.3389/fphar.2021.
Verhoef, J. J. F., and Anchordoquy, T. J. (2013). Questioning the use of PEGylation for 648407
drug delivery. Drug Deliv. Transl. Res. 3, 499–503. doi:10.1007/s13346-013-0176-5
Ziogas, D. C., Theocharopoulos, C., Lialios, P.-P., Foteinou, D., Koumprentziotis, I.-
Vidarsson, G., Dekkers, G., and Rispens, T. (2014). IgG subclasses and allotypes: from A., Xynos, G., et al. (2023). Beyond CTLA-4 and PD-1 inhibition: Novel immune
structure to effector functions. Front. Immunol. 5, 520. doi:10.3389/fimmu.2014.00520 checkpoint molecules for melanoma treatment. Cancers (Basel) 15, 2718. doi:10.3390/
Visser, K. E. de, and Joyce, J. A. (2023). The evolving tumor microenvironment: From cancers15102718
cancer initiation to metastatic outgrowth. Cancer Cell 41, 374–403. doi:10.1016/j.ccell.
Zmolek, W., Bañas, S., Barfield, R. M., Rabuka, D., and Drake, P. M. (2016). A simple
2023.02.016
LC/MRM-MS-based method to quantify free linker-payload in antibody-drug
von Arx, C., De Placido, P., Caltavituro, A., Di Rienzo, R., Buonaiuto, R., De conjugate preparations. J. Chromatogr. B Anal. Technol. Biomed. Life Sci. 1032,
Laurentiis, M., et al. (2023). The evolving therapeutic landscape of 144–148. doi:10.1016/j.jchromb.2016.05.055

Frontiers in Pharmacology 21 frontiersin.org


The AAPS Journal, Vol. 16, No. 4, July 2014 ( # 2014)
DOI: 10.1208/s12248-014-9598-3

Review Article

Understanding Pharmaceutical Quality by Design

Lawrence X. Yu,1,6 Gregory Amidon,2 Mansoor A. Khan,1 Stephen W. Hoag,3 James Polli,3
G. K. Raju,4,5 and Janet Woodcock1

Received 17 November 2013; accepted 24 March 2014; published online 23 May 2014

Abstract. This review further clarifies the concept of pharmaceutical quality by design (QbD) and describes
its objectives. QbD elements include the following: (1) a quality target product profile (QTPP) that identifies
the critical quality attributes (CQAs) of the drug product; (2) product design and understanding including
identification of critical material attributes (CMAs); (3) process design and understanding including
identification of critical process parameters (CPPs), linking CMAs and CPPs to CQAs; (4) a control strategy
that includes specifications for the drug substance(s), excipient(s), and drug product as well as controls for
each step of the manufacturing process; and (5) process capability and continual improvement. QbD tools and
studies include prior knowledge, risk assessment, mechanistic models, design of experiments (DoE) and data
analysis, and process analytical technology (PAT). As the pharmaceutical industry moves toward the
implementation of pharmaceutical QbD, a common terminology, understanding of concepts and expectations
are necessary. This understanding will facilitate better communication between those involved in risk-based
drug development and drug application review.
KEY WORDS: control strategy; critical quality attributes; pharmaceutical quality by design; process
understanding; product understanding.

INTRODUCTION Consider document; and ICH Q11 (Development and Manu-


facture of Drug Substance) have been issued, as have the
Quality by design (QbD) is a concept first developed by the conclusions of FDA-EMA’s parallel assessment of Quality-By-
quality pioneer Dr. Joseph M. Juran (1). Dr. Juran believed that Design elements of marketing applications (6–9). These docu-
quality should be designed into a product, and that most quality ments provide high level directions with respect to the scope and
crises and problems relate to the way in which a product was definition of QbD as it applies to the pharmaceutical industry.
designed in the first place. Woodcock (2) defined a high-quality Nonetheless, many implementation details are not
drug product as a product free of contamination and reliably discussed in these guidances or documents. There is confusion
delivering the therapeutic benefit promised in the label to the among industry scientists, academicians, and regulators despite
consumer. The US Food and Drug Administration (FDA) recent publications (10–13). This paper is intended to describe
encourages risk-based approaches and the adoption of QbD the objectives of pharmaceutical QbD, detail its concept and
principles in drug product development, manufacturing, and elements, and explain implementation tools and studies.
regulation. FDA’s emphasis on QbD began with the recognition
that increased testing does not necessarily improve product PHARMACEUTICAL QUALITY BY DESIGN
quality. Quality must be built into the product. OBJECTIVES
Over the years, pharmaceutical QbD has evolved with the
issuance of ICH Q8 (R2) (Pharmaceutical Development), ICH Pharmaceutical QbD is a systematic approach to devel-
Q9 (Quality Risk Management), and ICH Q10 (Pharmaceutical opment that begins with predefined objectives and empha-
Quality System) (3–5). In addition, the ICH Q1WG on Q8, Q9, sizes product and process understanding and control based on
and Q10 Questions and Answers; the ICH Q8/Q9/Q10 Points to sound science and quality risk management (3). The goals of
pharmaceutical QbD may include the following:

1. To achieve meaningful product quality specifications


1
Center for Drug Evaluation and Research, Food and Drug that are based on clinical performance
Administration, Silver Spring, Maryland 20993, USA. 2. To increase process capability and reduce product
2
University of Michigan, Ann Arbor, Michigan 48109, USA.
3 variability and defects by enhancing product and
University of Maryland, Baltimore, Maryland 21201, USA.
4 process design, understanding, and control
Massachusetts Institute of Technology, Cambridge, Massachusetts
02139, USA. 3. To increase product development and manufacturing
5
Light Pharm Inc., Cambridge, Massachusetts 02142, USA. efficiencies
6
To whom correspondence should be addressed. (e-mail: 4. To enhance root cause analysis and postapproval
lawrence.yu@fda.hhs.gov) change management

771 1550-7416/14/0400-0771/0 # 2014 American Association of Pharmaceutical Scientists


772 Yu et al.

Under QbD, these goals can often be achieved by 1. A quality target product profile (QTPP) that identifies
linking product quality to the desired clinical performance the critical quality attributes (CQAs) of the drug
and then designing a robust formulation and manufactur- product
ing process to consistently deliver the desired product 2. Product design and understanding including the
quality. identification of critical material attributes (CMAs)
Since the initiation of pharmaceutical QbD, the FDA 3. Process design and understanding including the iden-
has made significant progress in achieving the first tification of critical process parameters (CPPs) and a
objective: performance-based quality specifications. Some thorough understanding of scale-up principles, linking
examples of FDA policies include tablet scoring and bead CMAs and CPPs to CQAs
sizes in capsules labeled for sprinkle (14,15). The recent 4. A control strategy that includes specifications for the
FDA discussions on the assayed potency limits for narrow drug substance(s), excipient(s), and drug product as
well as controls for each step of the manufacturing
therapeutic index drugs and physical attributes of generic
process
drug products reflect this trend (16). Nonetheless, it
5. Process capability and continual improvement
should be recognized that ICH documents (3–9) did not
explicitly acknowledge clinical performance-based specifi-
cations as a QbD goal, although this was recognized in a Quality Target Product Profile that Identifies the Critical
recent scientific paper (10). Quality Attributes of the Drug Product
The second objective of pharmaceutical QbD is to
increase process capability and reduce product variability QTPP is a prospective summary of the quality charac-
teristics of a drug product that ideally will be achieved to
that often leads to product defects, rejections, and recalls.
ensure the desired quality, taking into account safety and
Achieving this objective requires robustly designed prod-
efficacy of the drug product. QTPP forms the basis of design
uct and process. In addition, an improved product and for the development of the product. Considerations for
process understanding can facilitate the identification and inclusion in the QTPP could include the following (3):
control of factors influencing the drug product quality.
After regulatory approval, effort should continue to & Intended use in a clinical setting, route of adminis-
improve the process to reduce product variability, defects, tration, dosage form, and delivery system(s)
rejections, and recalls. & Dosage strength(s)
QbD uses a systematic approach to product design and & Container closure system
development. As such, it enhances development capability, & Therapeutic moiety release or delivery and attributes
speed, and formulation design. Furthermore, it transfers affecting pharmacokinetic characteristics (e.g., disso-
resources from a downstream corrective mode to an lution and aerodynamic performance) appropriate to
upstream proactive mode. It enhances the manufacturer’s the drug product dosage form being developed
ability to identify the root causes of manufacturing & Drug product quality criteria (e.g., sterility, purity,
failures. Hence, increasing product development and stability, and drug release) appropriate for the
intended marketed product
manufacturing efficiencies is the third objective of phar-
maceutical QbD.
The final objective of QbD is to enhance root cause
Identification of the CQAs of the drug product is the
analysis and postapproval change management. Without good
next step in drug product development. A CQA is a
product and process understanding, the ability to efficiently
physical, chemical, biological, or microbiological property
scale-up and conduct root cause analysis is limited and
or characteristic of an output material including finished
requires the generation of additional data sets on the
drug product that should be within an appropriate limit,
proposed larger scale. FDA’s change guidances (17,18)
range, or distribution to ensure the desired product
provide a framework for postapproval changes. Recently,
quality (3). The quality attributes of a drug product may
the FDA issued a guidance intended to reduce the regulatory include identity, assay, content uniformity, degradation
filing requirements for specific low-risk chemistry, products, residual solvents, drug release or dissolution,
manufacturing, and control (CMC) postapproval manufactur- moisture content, microbial limits, and physical attributes
ing changes (19). such as color, shape, size, odor, score configuration, and
friability. These attributes can be critical or not critical.
ELEMENTS OF PHARMACEUTICAL QUALITY Criticality of an attribute is primarily based upon the
BY DESIGN severity of harm to the patient should the product fall
outside the acceptable range for that attribute. Probability
In a pharmaceutical QbD approach to product develop- of occurrence, detectability, or controllability does not
ment, an applicant identifies characteristics that are critical to impact criticality of an attribute.
quality from the patient’s perspective, translates them into the It seems obvious that a new product should be ade-
drug product critical quality attributes (CQAs), and estab- quately defined before any development work commences.
lishes the relationship between formulation/manufacturing However, over the years, the value of predefining the target
variables and CQAs to consistently deliver a drug product characteristics of the drug product is often underestimated.
with such CQAs to the patient. QbD consists of the following Consequently, the lack of a well-defined QTPP has resulted in
elements: wasted time and valuable resources. A recent paper by Raw
Understanding Pharmaceutical Quality by Design 773

et al. (12) illustrates the significance of defining the correct database (25) lists the safety limits of excipients based on
QTPP before conducting any development. Also, QbD exam- prior use in FDA-approved drug products.
ples exemplify the identification and use of QTPPs (20–22). It is well recognized that excipients can be a major
source of variability. Despite the fact that excipients can alter
Product Design and Understanding the stability, manufacturability, and bioavailability of drug
products, the general principles of excipient selection are not
Over the years, QbD’s focus has been on the process well-defined, and excipients are often selected ad hoc without
design, understanding, and control, as discussed in the ICH systematic drug-excipient compatibility testing. To avoid
Q8 (R2) guidance (3). It should be emphasized that product costly material wastage and time delays, ICH Q8 (R2)
design, understanding, and control are equally important. recommends drug-excipient compatibility studies to facilitate
Product design determines whether the product is able to the early prediction of compatibility (3). Systematic drug-
meet patients’ needs, which is confirmed with clinical studies. excipient compatibility studies offer several advantages as
Product design also determines whether the product is able to follows: minimizing unexpected stability failures which usual-
maintain its performance through its shelf life, which is ly lead to increased development time and cost, maximizing
confirmed with stability studies. This type of product under- the stability of a formulation and hence the shelf life of the
standing could have prevented some historical stability drug product, and enhancing the understanding of drug-
failures. excipient interactions that can help with root cause analysis
The key objective of product design and understanding is should stability problems occur.
to develop a robust product that can deliver the desired Formulation optimization studies are essential in developing a
QTPP over the product shelf life. Product design is open- robust formulation that is not on the edge of failure. Without
ended and may allow for many design pathways. Key optimization studies, a formulation is more likely to be high risk
elements of product design and understanding include the because it is unknown whether any changes in the formulation itself
following: or in the raw material properties would significantly impact the
quality and performance of the drug product, as shown in recent
examples (26,27). Formulation optimization studies provide impor-
& Physical, chemical, and biological characterization of tant information on the following:
the drug substance(s)
& Identification and selection of excipient type and & Robustness of the formulation including establishing
grade, and knowledge of intrinsic excipient variability functional relationships between CQAs and CMAs
& Interactions of drug and excipients & Identification of CMAs of drug substance, excipients,
& Optimization of formulation and identification of and in-process materials
CMAs of both excipients and drug substance & Development of control strategies for drug substance
and excipients
To design and develop a robust drug product that has the
intended CQAs, a product development scientist must give In a QbD approach, it is not the number of optimization
serious consideration to the physical, chemical, and biological studies conducted but rather the relevance of the studies and
properties of the drug substance. Physical properties include the utility of the knowledge gained for designing a quality
physical description (particle size distribution and particle drug product that is paramount. As such, the QbD does not
morphology), polymorphism and form transformation, aqueous equal design of experiments (DoE), but the latter could be an
solubility as a function of pH, intrinsic dissolution rate, important component of QbD.
hygroscopicity, and melting point(s). Pharmaceutical solid Drug substance, excipients, and in-process materials may
polymorphism, for example, has received much attention have many CMAs. A CMA is a physical, chemical, biological,
recently since it can impact solubility, dissolution, stability, and or microbiological property or characteristic of an input
manufacturability. Chemical properties include pKa, chemical material that should be within an appropriate limit, range,
stability in solid state and in solution, as well as photolytic and or distribution to ensure the desired quality of that drug
oxidative stability. Biological properties include partition coef- substance, excipient, or in-process material. For the purpose
ficient, membrane permeability, and bioavailability. of this paper, CMAs are considered different from CQAs in
Pharmaceutical excipients are components of a drug that CQAs are for output materials including product
product other than the active pharmaceutical ingredient. intermediates and finished drug product while CMAs are for
Excipients can (1) aid in the processing of the dosage input materials including drug substance and excipients. The
form during its manufacture; (2) protect, support, or CQA of an intermediate may become a CMA of that same
enhance stability, bioavailability, or patient acceptability; intermediate for a downstream manufacturing step.
(3) assist in product identification; or (4) enhance any Since there are many attributes of the drug substance
other attribute of the overall safety, effectiveness, or and excipients that could potentially impact the CQAs of the
delivery of the drug during storage or use (23). They intermediates and finished drug product, it is unrealistic that a
are classified by the functions they perform in a pharma- formulation scientist investigate all the identified material
ceutical dosage form. Among 42 functional excipient
attributes during the formulation optimization studies. There-
categories listed in USP/NF (24), commonly used excipi-
ents include binders, disintegrants, fillers (diluents), lubri- fore, a risk assessment would be valuable in prioritizing which
cants, glidants (flow enhancers), compression aids, colors, material attributes warrant further study. The assessment
sweeteners, preservatives, suspending/dispersing agents, should leverage common scientific knowledge and the
pH modifiers/buffers, tonicity agents, film formers/coatings, formulator’s expertise. A material attribute is critical when a
flavors, and printing inks. The FDA’s inactive ingredients realistic change in that material attribute can have a
774 Yu et al.

Table I. Typical Input Material Attributes, Process Parameters, and Quality Attributes of Pharmaceutical Unit Operations

Pharmaceutical unit operation

Input material attributes Process parameters Quality attributes


Blending/mixing
• Particle size • Type and geometry of mixer • Blend uniformity
• Particle size distribution • Mixer load level • Potency
• Fines/oversize • Order of addition • Particle size
• Particle shape • Number of revolutions (time and speed) • Particle size distribution
• Bulk/tapped/true density • Agitating bar (on/off pattern) • Bulk/tapped/true density
• Cohesive/adhesive properties • Discharge method • Moisture content
• Electrostatic properties • Holding time • Flow properties
• Moisture content • Environment temperature and RH • Cohesive/adhesive properties
• Powder segregation
• Electrostatic properties
Size reduction/comminution
• Particle/granule size Ribbon milling
• Particle/granule size • Ribbon dimensions
distribution • Ribbon density
• Fines • Ribbon porosity/solid fraction
• Particle/granule shape
• Bulk/tapped/true density Impact/cutting/screening mills • Particle/granule size
• Adhesive properties • Mill type • Particle/granule size distribution
• Electrostatic properties • Speed • Particle/granule shape
• Hardness/plasticity • Blade configuration, type, orientation • Particle/granule shape factor
• Viscoelasticity • Screen size and type (e.g., aspect ratio)
• Brittleness • Feeding rate • Particle/granule density/Porosity
• Elasticity • Bulk/tapped/true density
• Solid form/polymorph Fluid energy mill • Flow properties
• Moisture content • Number of grinding nozzles • API polymorphic form
• Granule porosity/density • Feed rate • API crystalline morphology
• Nozzle pressure • Cohesive/adhesive properties
• Classifier • Electrostatic properties
• Hardness/Plasticity
Granule/ribbon milling • Viscoelasticity
• Mill type • Brittleness
• Speed • Elasticity
• Blade configuration, type, orientation
• Screen size and type
• Feeding rate
Wet granulation
• Particle size distribution High/low shear granulation • Endpoint measurement
• Fines/Oversize • Type of granulator (High/low shear, top/bottom drive) (e.g., power consumption, torque,
• Particle shape • Fill level etc.)
• Bulk/tapped/true density • Pregranulation mix time • Blend uniformity
• Cohesive/adhesive properties • Granulating liquid or solvent quantity • Potency
• Electrostatic properties • Impeller speed, tip speed, configuration, location, power • Flow
• Hardness/plasticity consumption/torque • Moisture content
• Viscoelasticity • Chopper speed, configuration, location, power consumption • Particle size and distribution
• Brittleness • Spray nozzle type and location • Granule size and distribution
• Elasticity • Method of binder excipient addition (dry/wet) • Granule strength and uniformity
• Solid form/polymorph • Method of granulating liquid addition (spray or pump) • Bulk/tapped/true density
• Moisture content • granulating liquid temperature • API polymorphic form
• granulating liquid addition rate and time • Cohesive/adhesive properties
• Wet massing time (post-granulation mix time) • Electrostatic properties
• Bowl temperature(jacket temperature) • Granule brittleness
• Product temperature • Granule elasticity
• Post mixing time • Solid form/polymorph
• Pump Type: Peristaltic, Gear type
• Granulating liquid vessel (e.g., pressurized, heated)

Fluid bed granulation


• Type of fluid bed
• Inlet air distribution plate
• Spray nozzle (tip size, type/quantity/ pattern/configuration/position)
• Filter type and orifice size
Understanding Pharmaceutical Quality by Design 775

Table I. (continued)

Pharmaceutical unit operation

Input material attributes Process parameters Quality attributes

• Fill level
• Bottom screen size and type
• Preheating temperature/time
• Method of binder excipient addition (dry/wet)
• Granulating liquid temperature
• Granulating liquid quantity
• Granulating liquid concentration/viscosity
• Granulating liquid holding time
• Granulating liquid delivery method
• Granulating liquid spray rate
• Inlet air, volume, temperature, dew point
• Atomization air pressure
• Product and filter pressure differentials
• Product temperature
• Exhaust air temperature, flow
• Filter shaking interval and duration

Drying
• Particle size, distribution Fluidized bed • Granule size and distribution
• Fines/oversize • Inlet air volume, temperature, dew point • Granule strength, uniformity
• Particle shape • Product temperature • Flow
• Cohesive/adhesive properties • Exhaust air temperature, flow • Bulk/tapped/true density
• Electrostatic properties • Filter type and orifice size • Moisture content
• Hardness/plasticity • Shaking interval and duration • Residual solvents
• Viscoelasticity • Total drying time • API polymorphic form or transition
• Brittleness • Purity profile
• Elasticity Tray • Moisture profile (e.g. product
• Solid form/polymorph • Type of tray dryer temperature vs. LOD)
• Moisture content • Bed thickness/tray depth (depth of product per tray) • Potency
• Type of drying tray liner (e.g., paper, plastic, • Cohesive/adhesive properties
synthetic fiber, etc.) • Electrostatic properties
• Quantity carts and trays per chamber
• Quantity of product per tray
• Drying time and temperature
• Air flow
• Inlet dew point

Vacuum/microwave
• Jacket temperature
• Condenser temperature
• Impeller speed
• Bleed air volume
• Vacuum pressure
• Microwave power
• Electric field
• Energy supplied
• Product temperature
• Bowl and lid temperature
• Total drying time
Roller compaction/chilsonation
• Particle size, distribution • Type of roller compactor • Ribbon appearance (edge attrition,
• Fines/oversize • Auger (feed screw) type/design (horizontal, splitting, lamination, color, etc.)
• Particle shape vertical or angular) • Ribbon thickness
• Cohesive/adhesiveproperties • Deaeration (e.g., vacuum) • Ribbon density (e.g., envelop
• Electrostatic properties • Auger (feed screw) speed density)
• Hardness/plasticity • Roll shape (cylindrical or interlocking). • Ribbon porosity/solid fraction
• Bulk/tapped/true density • Roll surface design (smooth, knurled, serrated, • Ribbon tensile strength/breaking
• Viscoelasticity or pocketed) force
• Brittleness • Roll gap width (e.g., flexible or fixed) • Throughput rate
• Elasticity • Roll speed • API polymorphic form and transition
• Roll pressure
776 Yu et al.

Table I. (continued)

Pharmaceutical unit operation

Input material attributes Process parameters Quality attributes

• Solid form/polymorph • Roller temperature


• Fines recycled (yes or no, # of cycles)

Extrusion–Spheronization
• Particle size, distribution • Type of extruder (screw or basket) • Extrudate
• Fines/oversize • Screw length, pitch, and diameter • Density
• Particle shape • Screw channel depth • Length/thickness/diameter
• Cohesive/adhesiveproperties • Screw blade configuration • Moisture content
• Electrostatic properties • Number of screws (single/dual) • API polymorphic form and transition
• Hardness/plasticity • Die or screen configuration (e.g., radial or axial) • Content uniformity
• Bulk/tapped/true density • Die length/diameter ratio • Throughput
• Viscoelasticity • Roll diameter (mm)
• Brittleness • Screen opening diameter (mm) • Pellets after spheronization
• Elasticity • Screw speed (rpm) • Pellets size and distribution
• Solid form/polymorph • Feeding rate (g/min) • Pellets shape factor (e.g. aspect
• Type and scale of spheronizer ratio)
• Spheronizer load level • Bulk/Tapped density
• Plate geometry and speed • Flow properties
• Plate groove design (spacing and pattern) • Brittleness
• Air flow • Elasticity
• Residence time • Mechanical strength
• Friability

Hot melt extrusion


• Particle size, distribution • Screw design (twin/single) • Extrudate density
• Fines/oversize • Screw speed • Length/thickness/diameter
• Particle shape • Screw opening diameter (mm) • Polymorphic form and transition
• Melting point • Solid and liquid feed rates • Content uniformity
• Density • Feeder type/design • Throughput
• Solid form/polymorph • Feed rate
• Moisture content • No. of zones
• Zone temperatures
• Chilling rate

Tabletting
• Particle/granule size • Type of press (model, geometry, number of stations) • Tablet appearance
and distribution • Hopper design, height, angle, vibration • Tablet weight
• Fines/oversize • Feeder mechanism (gravity/forced feed, shape of wheels, • Weight uniformity
• Particle/granule shape direction of rotation, number of bars) • Content uniformity
• Cohesive/adhesive • Feed frame type and speed • Hardness/tablet breaking force/
properties • Feeder fill depth tensile strength
• Electrostatic properties • Tooling design (e.g., dimension, score configuration, • Thickness/dimensions
• Hardness/plasticity quality of the metal) • Tablet porosity/density/solid fraction
• Bulk/tapped/true density • Maximum punch load • Friability
• Viscoelasticity • Press speed/dwell time • Tablet defects
• Brittleness • Precompression force • Moisture content
• Elasticity • Main compression force • Disintegration
• Solid form/polymorph • Punch penetration depth • Dissolution
• Moisture • Ejection force
• Dwell Time
Encapsulation
• Particle/granule size and • Machine type • Capsule appearance
distribution • Machine fill speed • Weight
• Fines/oversize • Tamping Force • Weight uniformity
• Particle/granule shape • No. of tamps • Content uniformity
• Cohesive/adhesive properties • Auger screw design/speed • Moisture content
• Electrostatic properties • Powder bed height • Slug tensile strength
• Hardness/plasticity • Disintegration
• Bulk/tapped/true density • Dissolution
• Viscoelasticity
• Brittleness
Understanding Pharmaceutical Quality by Design 777

Table I. (continued)

Pharmaceutical unit operation

Input material attributes Process parameters Quality attributes

• Elasticity
• Solid form/polymorph
• Moisture

Pan coating
• Tablet dimensions • Type of pan coater (conventional or side-vented) • Coating efficiency
• Tablet defects • Pan (fully perforated or partial perforated) • Core tablet weight before and after
• Hardness/plasticity • Baffle (design, number, location) preheating
• Density • Pan load level • Moisture (gain/loss) during
• Porosity • Pan rotation speed preheating
• Moisture content • Spray nozzle (type, quantity, pattern, configuration, • Environmental equivalency factor
spray pattern) • Coated drug product (e.g., tablet or
• Nozzle to bed distance capsule) appearance
• Distance between nozzles • % weight gain
• Nozzle orientation • Film thickness
• Total preheating time • Coating (polymer and /or color)
• Inlet air flow rate, volume, temperature, dew point uniformity
• Product temperature • Hardness/breaking force/Tensile
• Individual nozzle spray rate strength
• Total spray rate • Friability
• Atomization air pressure • Moisture (gain/loss) during overall
• Pattern air pressure process
• Exhaust air temperature, air flow • Residual solvent(s)
• Total coating, curing time and drying time • Disintegration
• Dissolution
• Tablet defects
• Visual attributes
Fluid bed coating
• Tablet dimensions • Type of fluid bed coater • Coating efficiency
• Tablet defects • Fluid bed load level • Core tablet weight before and after
• Hardness/plasticity • Partition column diameter preheating
• Density/porosity • Partition column height • Moisture (gain/loss) during
moisture content • Number of partition columns preheating
• Air distribution plate type and size • Environmental equivalency factor
• Filter type and orifice size • Coated drug product (e.g., tablet or
• Filter differential pressure capsule) appearance
• Filter shaking interval and duration • % weight gain
• Spray nozzle (type, quantity, pattern, configuration) • Film thickness
• Nozzle port size • Coating (polymer and /or color)
• Total preheating time uniformity
• Spray rate per nozzle • Hardness/breaking force/tensile
• Total spray rate strength
• Atomization air pressure • Friability
• Inlet air flow rate, volume, temperature, dew point • Moisture (gain/loss) during overall
• Product temperature process
• Exhaust air temperature, air flow • Residual solvent(s)
• Total coating, curing and drying time • Disintegration
• Dissolution
• Tablet defects
• Visual attributes

Laser drilling
• Size/dimensions • Conveyor type • Opening diameter (internal and
• Polymer type • Conveyor speed external)
membrane thickness • Laser power • Depth
• Number of pulses • Shape of the opening
• Type(s) of lens(es)
• One or two sided
• Number of holes
778 Yu et al.

significant impact on the quality of the output material. 3. Establish levels or ranges of these potentially high-risk
Product understanding includes the ability to link input parameters
CMAs to output CQAs. The steps taken to gain product 4. Design and conduct experiments, using DoE when
understanding may include the following: appropriate
5. Analyze the experimental data and, when possible,
1. Identify all possible known input material attributes determine scalability and apply first principle models
that could impact the performance of the product to determine if a process parameter is critical. Link
2. Use risk assessment and scientific knowledge to CMAs and CPPs to CQAs when possible.
identify potentially high risk attributes 6. Develop a control strategy. For critical parameters,
3. Establish levels or ranges of these potentially high-risk define acceptable ranges. For noncritical parameters,
material attributes the acceptable range is the range investigated. When
4. Design and conduct experiments, using DoE when more than one process parameter or material attribute
appropriate is involved, these defined acceptable ranges may be
5. Analyze the experimental data and, when possible, apply termed process design space
first principle models to determine if an attribute is critical
6. Develop a control strategy. For critical material While developing a strategy for investigating both
attributes, define acceptable ranges. For noncritical product design and understanding and process design and
material attributes, the acceptable range is the range understanding, studies can be designed in such a way that
investigated. When more than one excipient is in- both the objectives of product and process understanding are
volved, these defined acceptable ranges may be achieved simultaneously. In addition, an interactive (or
termed formulation design space interdependent) relationship among material attributes, pro-
cess parameters, and product attributes can be more easily
developed when such analyses are performed in carefully
Process Design and Understanding planned and designed experimental studies.
ICH Q8 (R2) defines design space as the multidimen-
A pharmaceutical manufacturing process usually consists sional combination and interaction of input variables (e.g.,
of a series of unit operations to produce the desired quality material attributes) and process parameters that have been
product. Unit operations may be executed in batch mode or in a demonstrated to provide assurance of quality (3). Parameter
continuous manufacturing process. A unit operation is a discrete movements that occur within the design space are not
activity that involves physical or chemical changes, such as subjected to regulatory notification. However, movement
mixing, milling, granulation, drying, compression, and coating. out of the design space is considered to be a change and
A process is generally considered well-understood when (1) all would normally initiate a regulatory postapproval change
critical sources of variability are identified and explained, (2) process. Design space is proposed by the applicant and is
variability is managed by the process, and (3) product quality subject to regulatory assessment and approval. Thus, design
attributes can be accurately and reliably predicted (28). space is the direct outcome of analysis of the DoE data or
Process parameters are referred to as the input operating validated models such as first-principle models.
parameters (e.g., speed and flow rate) or process state variables Design space may be scale and equipment dependent.
(e.g., temperature and pressure) of a process step or unit operation. Therefore, the design space determined at laboratory scale
A process parameter is critical when its variability has an impact on may need to be justified for use at commercial scale.
a critical quality attribute and therefore should be monitored or Approaches for justification may include geometric consider-
controlled to ensure the process produces the desired quality. ations, kinematic considerations, heat and mass transfer, or
Under this definition, the state of a process depends on its CPPs dimensionless numbers as well as continual verification
and the CMAs of the input materials. Table I lists the typical during commercial manufacturing. Justification is needed
manufacturing unit operations, material attributes, process param- because the mechanistic understanding of pharmaceutical
eters, and quality attributes for solid oral dosage forms. unit operations may be limited and scale-up is largely based
Process robustness is the ability of a process to deliver on general rule of thumb and trial-and-error approaches;
acceptable drug product quality and performance while tolerating however, when mechanistic understanding or reliable
variability in the process and material inputs (29). The effects of
variations in process parameters and material attributes are CPPs
investigated in process robustness studies. The analysis of these
experiments identifies CPPs that could affect drug product quality
and establishes limits for these CPPs (and CMAs) within which Pharmaceutical
the quality of drug product is assured. The relationship between CMAs CQAs
Unit
input CMAs and CPPs and output CQAs is shown in Fig. 1. Input Operation Output
Steps to establish process understanding are very similar Materials Materials or
to those of product understanding and include the following: Product
1. Identify all possible known process parameters that CQAs = f (CPP1, CPP2 , CPP3 …CMA1, CMA2, CMA3…)
could impact the performance of the process
Fig. 1. Link input critical material attributes (CMAs) and critical
2. Use risk assessment and scientific knowledge to process parameters (CPPs) to output critical quality attributes
identify potentially high-risk parameters (CQAs) for a unit operation
Understanding Pharmaceutical Quality by Design 779

empirical models (i.e., extensive process understanding) release testing (e.g., the use of predictive models as a
exists, then the design space can be translated across scale. surrogate for traditional release test, where the model may
Pharmaceutical products are frequently manufactured by be defined in terms of traditional in-process measurements).
a combination of unit operations. For example, tablets Level 2 consists of pharmaceutical control with reduced
prepared by direct compression may simply involve blending end-product testing and flexible material attributes and process
and compression. However, when tablets are prepared by wet parameters within the established design space. QbD fosters
granulation, unit operations may involve blending, granula- product and process understanding and facilitates identification
tion, wet milling, drying, dry milling, blending for lubrication, of the sources of variability that impact product quality.
compression, coating, and packaging. In such cases, the Understanding the impact that variability has on in-process
output of the first unit operation becomes an input of materials, downstream processing, and drug product quality
subsequent unit operations. Process understanding could be provides an opportunity to shift controls upstream and to reduce
conducted on each unit operation or a combination of unit
the reliance on end-product testing (3).
operations to determine CMAs, CPPs, and CQAs. Figure 2
Level 3 is the level of control traditionally used in the
shows an example how the CMAs and CPPs were deter-
pharmaceutical industry. This control strategy relies on extensive
mined, using an example of an immediate release dosage
end-product testing and tightly constrained material attributes
form (20).
and process parameters. Due to limited characterization of the
Control Strategy sources of variability and inadequate understanding of the
impact that CMAs and CPPs have on the drug product CQAs,
The knowledge gained through appropriately designed any significant change in these requires regulatory oversight.
development studies culminates in the establishment of a Significant industry and regulatory resources are spent debating
control strategy. As shown in Fig. 3, control strategy could issues related to acceptable variability, the need for additional
include three levels of controls as follows: controls, and the establishment of acceptance criteria.
In reality, a hybrid approach combining levels 1 and 2
Level 1 utilizes automatic engineering control to can be used. ICH Q8 (R2) (3) defines a control strategy as a
monitor the CQAs of the output materials in real time. This planned set of controls, derived from current product and
level of control is the most adaptive. Input material attributes process understanding that ensures process performance and
are monitored and process parameters are automatically product quality. The controls can include parameters and
adjusted to assure that CQAs consistently conform to the attributes related to drug substance and drug product
established acceptance criteria. Level 1 control can enable materials and components, facility and equipment operating
real-time release testing and provides an increased level of conditions, in-process controls, finished product specifica-
quality assurance compared to traditional end-product test- tions, and the associated methods and frequency of monitor-
ing. It should be noted that adoption of process analytical ing and control. A control strategy can include, but is not
technology (PAT) is not the only way to implement real-time limited to, the following (3):

Material Attributes Process Parameters


Acetriptan [solid state form, solubility, morphology, particle size distribution (PSD), bulk, Pre-roller compaction blending and lubrication [blender type, order of addition, fill
density, flowability, cohesiveness, moisture content, hygroscopicity, chemical stability, level, rotation speed, time, number of revolutions, intensifier bar (on/off),
process impurities, residual solvents, etc…] environment (temp and RH), etc…]
Lactose [type, grade, source, amount, polymorphism, PSD, morphology, aspect ratio, bulk, Roller compaction [compactor type, feed screw speed, deaeration, roller surface
density, moisture content, flowability, compressibility, lot-to-lot variability, etc…] design, roller pressure, roller speed, roller gap, environment (temp and RH), etc…]
Microcrystalline Cellulose (MCC) [type, grade, source, amount, PSD, bulk, density, Milling [Mill type, blade configuration, speed, screen type, mesh size, # of recycles,
morphology, flowability, moisture, content, compressibility, lot-to-lot variability, etc…] environment (temp and RH), etc…]
Croscarmellose [type, grade, source, amount, degree of substitution, PSD, moisture Final blending and Lubrication [blender type, order of addition, fill level, rotation
content, lot-to-lot variability, etc…] speed, time, number of revolutions, intensifier bar (on/off), environment (temp and
Talc [type, grade, amount, PSD, density, specific surface area, moisture content, lot-to-lot RH), etc…]
variability, etc…] Compression [Press type, number of stations, tooling design, feed frame paddle speed,
Mg St [type, grade, source, amount, PSD, specific surface area, moisture content, lot-to-lot feeder fill depth, pre-compression force, main compression force, press speed
variability, etc…] (dwell time), hopper design, hopper fill level, drop height of finished tablets, run
time, environment (temp and RH), etc…]

Risk Assessment
Prior Knowledge
First Principle
Critical Material Attributes* DoEs Critical Process Parameters*
Acetriptan: PSD, polymorphic form
Pre-roller compaction blending and lubrication:
Lactose and MCC: amount and ratio
number of revolutions
Croscarmellose: amount
Roller compaction: roller pressure and roller gap
Talc: amount
Milling: mill screen orifice size
Mg St: amount
Final blending and Lubrication: none within the ranges studied
(Note: Excipients type, grade, and source are fixed and its quality is
Compression: main compression force
controlled per compendial/in-house specifications.)
Drug Product
Critical Quality Attributes
(Part of QTPP)

DoEs & Scale-up Physicochemical DoEs & Scale-up


-With interactions and Assay -With interactions
quadratic responses Content Uniformity and quadratic
Dissolution
-Univariate OK if responses
proven that there are Safety -Univariate OK if
Stability
no interactions proven that there are
Degradation Products
no interactions
Efficacy
Bioavailability

*Conclusion is drawn based upon the ranges studied and the control strategy for other variables (fixed or controlled within the ranges studied)

Fig. 2. Product and process understanding: an example for immediate release dosage forms
780 Yu et al.

based on sample standard deviation of all individual


(observed) samples taken over a longer period of time; the
result is a process performance index (Pp and Ppk). A state
of statistical control is achieved when the process exhibits
no detectable patterns or trends, such that the variation
seen in the data is believed to be random and inherent to
the process (31).
When a process is not in a state of statistical control, it is
because the process is subject to special cause (source of
intermittent variation in a process). Special causes can give rise
to short-term variability of the process or can cause long-term
shifts or drifts of the process mean. Special causes can also create
transient shifts or spikes in the process mean. On the other hand,
common cause is a source of inherent variation that is random,
always present, and affects every outcome of the process. In a
QbD development process, the product and process under-
standing gained during pharmaceutical development should
result in early identification and mitigation of potential sources
of common cause variation via the control strategy. The
Fig. 3. Control strategy implementation options
manufacturing process will move toward a state of statistical
control, and, once there, the manufacturer will continue to
& Control of input material attributes (e.g., drug substance,
improve process capability by reducing or removing some of the
excipient, in process material, and primary packaging
material) based on an understanding of their impact on random causes present and/or adjusting the process mean
processability or product quality towards the preferred target value to the benefit of the patient.
& Product specification(s) In a non-QbD approach, common cause variation is more likely
& Controls for unit operations that have an impact on to be discovered during commercial production and may
downstream processing or product quality (e.g., the impact interrupt commercial production and cause drug shortage when
of drying on degradation and particle size distribution of it will require a root cause analysis.
the granulate on dissolution) Process capability can be used to measure process
& In-process or real-time release testing in lieu of end-product improvement through continuous improvement efforts that
testing (e.g., measurement and control of CQAs during focus on removing sources of inherent variability from the
processing) process operation conditions and raw material quality.
& A monitoring program (e.g., full product testing at regular Ongoing monitoring of process data for Cpk and other
intervals) for verifying multivariate prediction models
measures of statistical process control will also identify when
special variations occur that need to be identified and
Process Capability and Continual Improvement corrective and preventive actions implemented.
Continuous improvement is a set of activities that the
Process capability measures the inherent variability of a applicant carries out in order to enhance its ability to meet
stable process that is in a state of statistical control in requirements. Continual improvements typically have five
relation to the established acceptance criteria. Table II phases as follows (32):
shows the definition, calculation formula, and description of
process capability indices (30) that are useful for monitoring & Define the problem and the project goals, specifically
the performance of pharmaceutical manufacturing process- & Measure key aspects of the current process and
es. Calculations based on the inherent variability due to collect relevant data
common cause of a stable process (i.e., in a state of & Analyze the data to investigate and verify cause-and-
statistical control) result in process capability (Cp and Cpk) effect relationships. Determine what the relationships
indices. When the process has not been demonstrated to be are, and attempt to ensure that all factors have been
in a state of statistical control, the calculation needs to be considered. Seek out root cause of the defect if any.

Table II. Process Capability Indices and Their Measures

Index Description
ðUSL−LSLÞ
Cp ¼ Estimates process capability when the data mean is centered between upper and lower specification limits.
6b
σ

Cpkl ¼ ðMean−LSLÞ Estimates process capability when the data mean is not centered between upper and lower specification limits or when
3b
σ
specifications consist of a lower limit only.
Cpku ¼ ðUSL−MeanÞ Estimates process capability when the data mean is not centered between upper and lower specification limits or when
3b
σ
specifications consist of an upper limit only.

USL upper specification limit, LSL lower specification limit, σ


b (sigma hat) inherent variability due to common cause of a stable process
Understanding Pharmaceutical Quality by Design 781

& Improve or optimize the current process based upon The purpose of risk assessment prior to development
data analysis using techniques such as design of studies is to identify potentially high-risk formulation and
experiments to create a new, future state process. process variables that could impact the quality of the drug
Set up pilot runs to establish process capability. product. It helps to prioritize which studies need to be conducted
& Control the future state process to ensure that any and is often driven by knowledge gaps or uncertainty. Study
deviations from target are corrected before they results determine which variables are critical and which are not,
result in defects. Implement control systems such as
which facilitates the establishment of a control strategy. The
statistical process control, production boards, visual
outcome of the risk assessment is to identify the variables to be
workplaces, and continuously monitor the process.
experimentally investigated. ICH Q9 (4) provides a
nonexhaustive list of common risk assessment tools as follows:

In addition, continuous improvement can apply to legacy & Basic risk management facilitation methods (flow-
products. Legacy products usually have a large amount of charts, check sheets, etc.)
historical manufacturing data. Using multivariate analysis to & Fault tree analysis
examine the data could uncover major disturbances in the form & Risk ranking and filtering
of variability in raw materials and process parameters. Contin- & Preliminary hazard analysis
uous improvement could be achieved by reducing and control- & Hazard analysis and critical control points
ling this variability. Newer processes associated with a design & Failure mode effects analysis
space facilitate continuous process improvement since appli- & Failure mode, effects, and criticality analysis
cants will have regulatory flexibility to move within the design & Hazard operability analysis
space (ICH Q8). & Supporting statistical tools

PHARMACEUTICAL QUALITY BY DESIGN TOOLS


It might be appropriate to adapt these tools for use in specific
Prior Knowledge areas pertaining to drug substance and drug product quality.

Although not officially defined, the term “prior knowl- Mechanistic Model, Design of Experiments, and Data
edge” has been extensively used in workshops, seminars, Analysis
and presentations. In regulatory submissions, applicants
often attempt to use prior knowledge as a “legitimate” Product and process understanding is a key element of
reason for substitution of scientific justifications or QbD. To best achieve these objectives, in addition to mechanis-
conducting necessary scientific studies. tic models, DoE is an excellent tool that allows pharmaceutical
Knowledge may be defined as a familiarity with someone scientists to systematically manipulate factors according to a
or something, which can include information, facts, descrip- prespecified design. The DoE also reveals relationships between
tions, and/or skills acquired through experience or education. input factors and output responses. A series of structured tests
The word “prior” in the term “prior knowledge” not only are designed in which planned changes are made to the input
means “previous,” but also associates with ownership and variables of a process or system. The effects of these changes on
confidentiality, not available to the public. Thus, for the a predefined output are then assessed. The strength of DoE over
purpose of this paper, prior knowledge can only be obtained the traditional univariate approach to development studies is the
through experience, not education. Knowledge gained ability to properly uncover how factors jointly affect the output
through education or public literature may be termed public responses. DoE also allows us to quantify the interaction terms
knowledge. Prior knowledge in the QbD framework general- of the variables. DoE is important as a formal way of
ly refers to knowledge that stems from previous experience maximizing information gained while minimizing the resources
that is not in publically available literature. Prior knowledge required. DoE studies may be integrated with mechanism-based
may be the proprietary information, understanding, or skill studies to maximize product and process understanding.
that applicants acquire through previous studies. When DoE is applied to formulation or process devel-
opment, input variables include the material attributes (e.g.,
Risk Assessment particle size) of raw material or excipients and process
parameters (e.g., press speed or spray rate), while outputs
ICH Q9 quality risk management indicates that “the are the critical quality attributes of the in-process materials or
manufacturing and use of a drug product, including its final drug product (e.g., blend uniformity, particle size or
components, necessarily entail some degree of risk.… The particle size distribution of the granules, tablet assay, content
evaluation of the risk to quality should be based on scientific uniformity, or drug release). DoE can help identify optimal
knowledge and ultimately link to the protection of the patient conditions, CMAs, CPPs, and, ultimately, the design space.
and the level of effort, formality, and documentation of the FDA scientists have shown the use of DoE in product and
quality risk management process should be commensurate with process design in recent publications (33–39).
the level of risk (4).” The purpose of ICH Q9 is to offer a
systematic approach to quality risk management and does not Process Analytical Technology
specifically address risk assessment in product development.
However, the risk assessment tools identified in ICH Q9 are The application of PAT may be part of the control
applicable to risk assessment in product development also. strategy (28). ICH Q8 (R2) identifies the use of PAT to
782 Yu et al.

ensure that the process remains within an established design (Helen) Teng, Youmin Wang, Huiquan Wu, Abhay Gupta,
space (3). PAT can provide continuous monitoring of CPPs, Ziyaur Rahman, and Naiqi Ya for their valuable suggestions.
CMAs, or CQAs to make go/no go decisions and to
demonstrate that the process is maintained in the design
REFERENCES
space. In-process testing, CMAs, or CQAs can also be
measured online or inline with PAT. Both of these applica-
1. Juran JM. Juran on quality by design: the new steps for planning
tions of PAT are more effective at detecting failures than end- quality into goods and services. New York: The Free Press; 1992.
product testing alone. In a more robust process, PAT can 2. Woodcock J. The concept of pharmaceutical quality. Am Pharm
enable active control of CMAs and/or CPPs, and timely Rev 2004; 1–3.
adjustment of the operating parameters if a variation in the 3. U. S. Food and Drug Administration. Guidance for Industry: Q8
(2) Pharmaceutical Development. 2009
environment or input materials that would adversely impact 4. U. S. Food and Drug Administration. Guidance for Industry: Q9
the drug product quality is detected. Quality Risk Management. 2006.
Application of PAT involves four key components as 5. U. S. Food and Drug Administration. Guidance for Industry:
follows (40): Q10 pharmaceutical quality system. 2009.
6. U. S. Food and Drug Administration. Guidance for Industry: Q8,
& Multivariate data acquisition and analysis Q9, and Q10 questions and answers. 2011.
& Process analytical chemistry tools 7. ICH Quality Implementation Working Group. Points to consider.
ICH-endorsed guide for ICH Q8/Q9/Q10 implementation. 2011.
& Process monitoring and control 8. U. S. Food and Drug Administration. Guidance for Industry:
& Continuous process optimization and knowledge Q11 development and manufacture of drug substance. 2012.
management 9. U. S. Food and Drug Administration. FDA-EMA parallel
assessment of Quality-By-Design elements of marketing appli-
Multivariate data acquisition and analysis requires building cations. http://www.fda.gov/Drugs/DrugSafety/ucm365524.htm.
scientific understanding about a process and identifying critical Accessed 16 Nov 2013
10. Yu LX. Pharmaceutical quality by design: product and process
material attributes and process parameters that affect product development, understanding, and control. Pharm Res. 2008;25:781–91.
quality and integrating this knowledge into the process control, 11. Lionberger R, Lee S, Lee L, Raw A, Yu LX. Quality by design:
which is essentially the same as the process understanding in the concepts for ANDAs. AAPS J. 2008;10:268–76.
12. Raw AS, Lionberger R, Yu LX. Pharmaceutical equivalence by
context of QbD. Process analytical chemistry tools provide real- design for generic drugs: modified-release products. Pharm Res.
time and in situ data about the status of the process. Multivariate 2011;28:1445–53.
data analysis takes the raw information from the PAT tools and 13. Rathore AS, Winkle H. Quality by design for biopharmaceuticals.
connects it to CQAs. Based on the outcome of the data analysis, Nat Biotechnol. 2009;27:26–34.
14. U. S. Food and Drug Administration. Guidance for Industry: tablet
process controls adjust critical variables to assure that CQAs are scoring: nomenclature, labeling, and data for evaluation. 2013.
met. The information collected about the process provides a basis 15. U. S. Food and Drug Administration. Guidance for Industry: Size of
for further process optimization. Studies in FDA laboratories Beads in Drug Products Labeled for Sprinkle. January, 2011.
indicated the promise of several PAT tools and chemometric 16. U. S. Food and Drug Administration. Summary Minutes of the
Advisory Committee for Pharmaceutical Science and Clinical
approaches (41–44). Pharmacology. July 26, 2011. http://www.fda.gov/downloads/
AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/
CONCLUSION AdvisoryCommitteeforPharmaceuticalScienceandClinicalPharm-
acology/UCM272111.pdf. Accessed 13 Aug 2013.
The goals of implementing pharmaceutical QbD are to 17. U. S. Food and Drug Administration. Guidance for industry:
immediate release solid oral dosage forms scale-up and postapproval
reduce product variability and defects, thereby enhancing changes: chemistry, manufacturing, and controls, in vitro dissolution
product development and manufacturing efficiencies and testing, and in vivo bioequivalence documentation. 1995.
postapproval change management. It is achieved by designing 18. U. S. Food and Drug Administration. Guidance for industry:
a robust formulation and manufacturing process and establish- modified release solid oral dosage forms scale-up and postapproval
changes: chemistry, manufacturing, and controls, in vitro dissolution
ing clinically relevant specifications. The key elements of
testing, and in vivo bioequivalence documentation. 1997.
pharmaceutical QbD can include the QTPP, product design 19. U. S. Food and Drug Administration. Guidance for industry:
and understanding, process design and understanding, and scale CMC postapproval manufacturing changes to be documented in
up, control strategy, and continual improvement. Prior knowl- annual reports. 2014.
edge, risk assessment, DoE, and PAT are tools to facilitate 20. U. S. Food and Drug Administration. Quality by design for ANDs: an
example for immediate-release dosage forms. 2012. http://www.fda.gov/
QbD implementation. Finally, product and process capability is
downloads/Drugs/DevelopmentApprovalProcess/
assessed and continually improved postapproval during product HowDrugsareDevelopedandApproved/ApprovalApplications/
lifecycle management. AbbreviatedNewDrugApplicationANDAGenerics/UCM304305.pdf.
Accessed 16 Nov 2013.
21. U. S. Food and Drug Administration. Quality by design for ANDs:
an example for modified-release dosage forms. 2011. http://
ACKNOWLEDGMENT www.fda.gov/downloads/Drugs/DevelopmentApprovalProcess/
HowDrugsareDevelopedandApproved/ApprovalApplications/
AbbreviatedNewDrugApplicationANDAGenerics/
The authors would like to thank Lane V. Christensen, UCM304305.pdf. Accessed 16 Nov 2013.
Devinder Gill, Frank Holcombe Jr, Robert Iser, Khalid Khan, 22. CMC Biotech Working Group. A Mab: A case study in
Robert Lionberger, Jennifer Maguire, Christine Moore, Yingxu b i o p r o c e s s d e v e l o p m e n t . h t t p : / / w w w. g o o g l e . c o m /
(Daniel) Peng, Andre Raw, Bhagwant Rege, Susan url?sa=t&rct=j&q=&esrc=s&frm=1&source=web&cd=8&ved=0CF8-
QFjAH&url=http%3A%2F%2Fwww.ispe.org%2Fpqli%2Fa-
Rosencrance, Vilayat Sayeed, Paul Schwartz, Glen Smith, Yue mab-case-study-version2.1&ei=f6qIUpXqKuLlsATlioHgDA
Understanding Pharmaceutical Quality by Design 783

& u s g = A F Q j C N H 9 J B 1 7 H 4 g s s C d 48 9 d l O w V 4 x o A m D Q . 34. Rahman Z, Siddiqui A, Khan MA. Assessing the impact of


Accessed 16 Nov 2013. nimodipine devitrification in the ternary cosolvent system through
23. USP 34—NF 29 (United States Pharmacopeial Convention). quality by design approach. Int J Pharm. 2013;455(1–2):113–23.
Chapter 1078. Good manufacturing practice for bulk pharma- 35. Rahman Z, Siddiqui A, Khan MA. Orally disintegrating tablet of
ceutical excipients. Rockville, MD: USP; 2011, pp. 1415–1420 novel salt of antiepileptic drug: formulation strategy and
24. USP 34—NF 29 (United States Pharmacopeial Convention). evaluation. Eur J Pharm Biopharm. 2013;85:1300–9.
USP and NF Excipients, Listed by Category. Rockville, MD: 36. Zidan AS, Sammour OA, Hammad MA, Megrab NA, Habib
USP; 2011, pp. 583–595. MJ, Khan MA. Quality by design: understanding the formulation
25. U. S. Food and Drug Administration. Inactive Ingredient Search variables of a cyclosporine, a self-nanoemulsified drug delivery
for Approved Drug Products. http://www.accessdata.fda.gov/ systems by Box-Behnken design and desirability function. Int J
scripts/cder/iig/index.cfm, Accessed 13 Aug 2013. Pharm. 2007;332(1–2):55–63.
26. Nazzal S, Nutan M, Palamakula A, Shah R, Zaghloul AA, 37. Xu X, Khan MA, Burgess DJ. A Quality by design (QbD) case
Khan MA. Optimization of a self-nanoemulsified tablet study on liposomes containing hydrophilic API: II. Screening of
dosage form of ubiquinone using response surface methodol- critical variables, and establishment of design space at laboratory
ogy: effect of formulation ingredients. Int J Pharm. scale. Int J Pharm. 2012;423(2):543–53.
2002;240:103–14. 38. Yerlikaya F, Ozgen A, Vural I, Guven O, Karaagaoglu E, Khan
27. Awotwe-Otoo D, Agarabi C, Wu GK, Casey E, Read E, Lute S, MA, et al. Development and evaluation of paclitaxel nanoparti-
et al. Quality by design: impact of formulation variables and their cles using a quality-by-design (QbD) approach. J Pharm Sci.
interactions on quality attributes of a lyophilized monoclonal 2013;102(10):3748–61.
antibody. Int J Pharm. 2012;438(1–2):167–75. 39. Rahman Z, Khan MA. Hunter screening design to understand
28. U.S. Food and Drug Administration CDER. Guidance for the product variability of solid dispersion formulation of a
industry: PAT—a framework for innovative pharmaceutical peptide antibiotic. Int J Pharm. 2013;456(2):572–82.
development, manufacturing, and quality assurance. 2004. 40. Yu LX, Lionberger RA, Raw AS, D'Costa R, Wu H, Hussain
29. Glodek M, Liebowitz S, McCarthy R, McNally G, Oksanen C, AS. Application of process analytical technology to crystalliza-
Schultz T, et al. Process robustness—a PQRI white paper. Pharm tion process. Adv Drug Deliv Rev. 2004;56(3):349–69.
Eng. 2006;26:1–11. 41. Wu H, Khan MA. Quality-by-design (QbD): an integrated
30. NIST/SEMATECH e-Handbook of Statistical Methods. What is multivariate approach for the component quantification in
process capability? http://www.itl.nist.gov/div898/handbook/pmc/ powder blends. Int J Pharm. 2009;372(1–2):39–48.
section1/pmc16.htm. Accessed on 13 Aug 2013. 42. Wu H, Khan MA. Quality-by-design (QbD): an integrated process
31. ASTM E2281—08a (2012)e1 Standard practice for process and analytical technology (pat) approach to determine the nucleation
measurement capability indices. http://www.astm.org/Standards/ and growth mechanisms during a dynamic pharmaceutical co-
E2281.htm. Accessed 13 Aug 2013. precipitation process. J Pharm Sci. 2011;100(5):1969–86.
32. De Feo JA, Barnard W. JURAN Institute's six sigma breakthrough 43. Rahman Z, Siddiqui A, Khan MA. Characterization of a
and beyond—quality performance breakthrough methods. India: nonribosomal peptide antibiotic solid dispersion formulation by
Tata McGraw-Hill Publishing Company Limited; 2005. process analytical technologies sensors. J Pharm Sci.
33. Wu H, Khan MA. Quality-by-design (QbD): an integrated 2013;102(12):4337–46.
approach for evaluation of powder blending process kinetics 44. Xu X, Siddiqui A, Khan MA. Focused beam reflectance
and determination of powder blending end-point. J Pharm Sci. measurement to monitor nimodipine precipitation process. Int J
2009;98(8):2784–98. Pharm. 2013;456(2):353–6.

You might also like