Boosting CAR T-Cell Therapy for Effective Cancer Treatment
eBook
Published: November 5, 2024
Credit: iStock
CAR T-cell therapy empowers the body’s own immune cells to target and destroy cancer. However, the process of creating CAR T-cells comes with challenges, from maintaining high cell quality to ensuring safe clinical application.
This eBook explores the critical steps in CAR T-cell production, focusing on key techniques that enhance efficacy and safety for patients.
Download this eBook to explore:
- Techniques for enriching T-cells for effective treatment
- Innovations in CAR design to overcome tumor evasion tactics
- Quality control measures that ensure safe, clinical-grade CAR T-cells
AN INTRODUCTION
TO CAR T-CELL
PRODUCTION
Page 3
Locked on Target:
T-Cell Receptor-Mediated
Cancer Cell Killing
Page 4
Start the CAR:
Constructing CAR-T Cells
Page 5
Production Pipeline
of Chimeric Antigen
Receptor T Lymphocytes
Page 6
Efficiency and Quality:
Manufacturing CAR-T
Cells for the Clinic
Sponsored By:Improve Reproducibility.
PHC Corporation of North America
1300 Michael Drive, Suite A, Wood Dale, IL 60191
Toll Free USA (800) 858-8442, Fax (630) 238-0074
www.phchd.com/us/biomedical
PHC Corporation of North America is a subsidiary of PHC Holdings Corporation, Tokyo, Japan, a global
leader in development, design and manufacturing of laboratory equipment for biopharmaceutical,
life sciences, academic, healthcare and government markets.
PHCbi brand Cell-IQ™ CO2 and O2 incubators are designed to deliver reproducibility
critical to immunotherapy and regenerative medicine from one day to the next.
Your desired temperature and gas concentrations replicate the in vivo model
of any cell culture environment with precision and predictability. Our SafeCell™
and inCu-saFe® technologies protect your work in vitro. Learn more at
www.phchd.com/us/biomedical/cell-culture-incubators.
1) Baker, Monya. “1,500 scientists lift the lid on reproducibility.”
Nature, no. 533 (May 26, 2016): 452-54. doi:10.1038/533452a.
Select from eight models including your choice of high heat
or H2O2 decontamination with CO2 or CO2/O2 control.
CELL CULTURE INCUBATORS
MCO-5ACL MCO-18ACL MCO-170AICUV MCO-230AICUV MCO-80IC
70% of surveyed scientists admitted that they could not replicate someone else’s research.1
50% admitted that they couldn’t replicate their own research.13
AN INTRODUCTION TO CAR T-CELL PRODUCTION
Locked on Target:
T-Cell ReceptorMediated Cancer
Cell Killing
Immunotherapy enlists and empowers components of an individual’s own immune system to attack cancer cells, in contrast to traditional cancer treatment plans that involve
surgery, drugs, and radiation. Among several types of adoptive cell
transfers (ACTs) where the patient’s immune cells are collected,
genetically modified, and re-infused to attack and eliminate
cancer cells, CAR-T immunotherapy shows exceptional promise
against both blood cancers and solid tumors.1 In a clinical trial
using CD19-targeted CAR T-cells,2 durable remission was
achieved in 27 of 30 patients with acute myelocytic leukemia.
However, the dynamic heterogeneity of cancer cells and their
complex microenvironment pose hurdles in the success of CAR-T
immunotherapy.
Role Of T Cells In Mounting An Immune Response
Against Cancer
T lymphocytes, a type of multifunctional white blood cell, play
an essential role in mounting adaptive immune responses. T
lymphocytes can gauge the state of a cell’s interior by scanning for
antigens on its surface. Cytotoxic lymphocytes (CTLs) or killer T
cells, hunt down and eliminate compromised cells (e.g., infected,
cancerous, etc.) while helper T cells regulate other T- and
B-lymphocyte subsets.
CTLs, a subset of the αβT lymphocytes expressing CD8 cellsurface co-receptors, harbor on their surface copies of a multisubunit receptor molecule, the T-cell receptor (TCR). TCRs,
when triggered by Major Histocompatibility Complex 1 (MHC1),
activate T lymphocytes. This, in turn, activates signaling cascades
leading to cell proliferation, differentiation, cytokine production,
and ultimately, the death of cancerous cells. Cancer cells must
present processed antigens bound to MHC1 on their surface,
together with co-stimulatory molecules (CD8 and CD28), to
activate CTLs.
Upon CTL activation, the microtubule organizing center
(MTOC) polarizes secretory granular traffic so that these granules
fuse with the plasma membrane and release degrading enzymes
like granzymes and perforins that effectively digest and eliminate
cancer cells. FasL and TRAIL expressed on reactivated CTLs are
also able to kill susceptible cancer cells through interaction with
death receptors or by inducing apoptotic pathways.
TCR: Components, Structure, Co-Receptors,
Signaling
The TCR heterodimer is composed of six peptide chains that
detect the antigen presented on cells by MHC molecules. The
alpha and beta chains have amino-terminal variable and constant
regions and are linked by disufide bonds. Each TCR provides
a single antigen-binding site. Since the cytoplasmic domains
of TCR are relatively short, the intracellular signaling steps
that follow TCR binding to the antigen are primarily carried
out by CD3. CD3 molecules assemble together with the TCR
and include immunoreceptor tyrosine-based activation motifs
(ITAMs).
Tumor Immune Evasion
Cancer cells are recognized by TCRs through the expression of
neo-antigens. In order to grow, cancer cells use multiple strategies
to neutralize and evade the host’s immune response.3 Cancer
cells escape immune recognition by generating neo-antigens
with weak immunogenicity, downregulating and modulating
the expression of neo-antigens on cancer cells, and synthesizing
immune suppressants that inhibit effector cell function and
generate a milieu of tumor tolerance. The immunosuppressive
microenvironment limits and impairs therapeutic CAR-T cells
as well. Studies show tumor-mediated blocking of indoleamine
2,3-dioxygenase (IDO), an enzyme that converts tryptophan into
metabolites, can impair CAR T-cell function.4 Therefore, one of
the primary considerations in designing CAR-T cells is to find
means to overcome the various ploys tumor cells use to avoid
detection and thrive.
For references, please see page 7.4
AN INTRODUCTION TO CAR T-CELL PRODUCTION
CARs, short for chimeric antigen receptors, are engineered molecules that consist of an extracellular ligand binding domain responsible for target specificity, a domain that
inserts the receptor into the cell membrane, and one or more
signaling domains that face the cell’s cytosol and are responsible
for activating T-cell division. Signaling domains in CARs may
include CD3ζ, CD27, CD28, ICOS, 4-IIB, and OX-40T. Cells that
express CARs are essentially engineered to kill cancer cells and
persist to patrol for emerging signs of tumor cells. CAR-T cells
were first cultured in 1989, when first generation chimeric TCR
genes were functionally expressed in T cells giving recipient T
cells the ability to recognize and respond to antigens in a nonMHC-restricted manner.1 Newer generations of CARs have
multiple co-stimulatory domains that enhance T-cell proliferation
and circulatory lifespan while curbing toxicity.
CAR-T Cells Counter Tumor Evasion
Tumor cells can evade T cells in the absence of robust tumor
recognition mechanisms. In CAR-T cell-mediated immunity,
a CAR’s antibody-like single-chain variable fragment (scFv)
engages antigens on tumor cell surfaces without the necessity
for MHC presentation. Moreover, tumor microenvironments
are characterized by T-cell exhaustion, a condition where
tumor-secreted immunosuppressive factors cause reduced
T-cell proliferation, increased expression of inhibitory receptors,
decreased production of stimulatory cytokines, and compromised
cytotoxicity. CAR-T cells designed to target multiple tumor cell
antigens can dramatically improve recognition specificity for
tumor-cells. In addition to directly killing tumor cells, CAR-T
cells are also engineered to deliver anti-tumor agents to kill cancer
clones or modify the tumor microenvironment.2
CAR: Design, Assembly, and Introduction Into T
Cells
T lymphocytes are induced to express CARs by delivering
and incorporating the CAR gene into the T-cell genome
using electroporation or disarmed viral vectors from murine
retroviruses or lentiviruses. It’s imperative to screen viral vectors
to ensure they do not replicate in the human host. Electroporation
results in transient expression but avoids the risk of developing
cancerous cells due to an insertional mutation. Advances in
CAR-T cells include other modifications in the T-cell genome
through gene editing and CRISPR-guided nucleases.
Modified T cells are then induced to divide clonally using an
artificial antigen-presenting system (aAPC) consisting of antiCD3/anti-CD28 beads or lentiviral-aAPCs. Contamination, a
risk in all ex vivo cultures, can be minimized by using closed
culture systems. Expansion and proliferation of CAR-T cells, if
uncontrolled in the circulation, can lead to toxicity and severe
adverse effects, including off-target attacks. Therefore, CAR
T-cell design must include feedback regulatory systems for
the optimization of therapeutic timing, strength of anti-tumor
activity, and target specificity.
CAR: Testing and Screening
CAR T-cells are tested for five major classes of functional
challenges to their therapeutic application: first, the capacity
of these cells to infiltrate tumor sites, particularly in the case of
solid tumors; second, their capacity to proliferate and persist in
circulation; third, their ability to recognize cancer cells; fourth,
their ability to function in an immunosuppressive environment;
and fifth, their capacity to limit their expansion through feedback
loops to curb toxicity and other adverse effects. Additionally,
CAR-T cells are screened for contaminants that may include other
T-cell subsets or tumor cells.
Multiple factors contribute to the variability observed in clinical
responses to CAR-T cell therapy. Preparative conditioning
regimes are administered to patients in order to reduce the
number of circulating T cells (lymphodepletion), promoting the in
vivo expansion of transferred CAR-T cells. Other factors include
the dosage of infused cells, the final steady-state number of cells,
the loss of tumor antigens, and the number of regulatory T cells in
the tumor microenvironment, among others.3 The development
of innovative strategies to regulate these diverse factors will
determine the long-term clinical benefits of CAR-T cells.
For references, please see page 7.
Start the CAR:
Constructing CAR-T
CellsWorkflow Key Considerations
AN INTRODUCTION TO CAR T-CELL PRODUCTION
The Chimeric Antigen Receptor (CAR) is an artificial assembly of an extracellular antigen binding domain, a transmembrane
domain, and an intracellular domain(s) that enables engineered T cells to home in on tumor targets with enhanced
specificity, proliferate rapidly in circulation, terminate tumor cells, and persist to patrol the body for emerging tumors.
Production Pipeline of Chimeric Antigen
Receptor T lymphocytes
Collecting T lymphocytes from
patients: Leukapheresis
Patients with relapsed cancer
have low T-cell counts making it
difficult to collect sufficient T cells.
Enrichment: Density gradient
centrifugation, elutriation,
immunomagnetic bead selection
Elimination of contaminants, like red blood
cells, platelets, monocytes, and tumor cells,
requires a multi-pronged approach.
Gene modification: Electroporation
and retroviral/lentiviral transduction
Viral vectors must be disarmed prior to
delivery. Next-generation CAR-T cells
include multiple genetic modifications to
increase their capacity to kill cancer cells
and persist in the circulation.
Activation and expansion: Polyclonal
activation through artificial antigen
presenting systems (anti-CD8/antiCD28 immunomagnetic beads/LV-APCs)
Consistency is achieved through
standardization and validation of raw
materials and protocols according to cGMPs
(current good manufacturing practices).
Quality Assurance: Testing for
viability, phenotyping, gram staining,
endotoxin, and bacterial, fungal, and
mycoplasma contaminants
Title 21 CFR, parts 210 and 211
outline the regulations for quality
management as set by FDA.
Formulation and Administration:
Testing for clinically prescribed dosage
and route of administration
Therapeutic cell preservation,
packaging, transport, receipt, and
administration must maintain product
stability and chain of custody.6
AN INTRODUCTION TO CAR T-CELL PRODUCTION
Manufacturing of CAR-T cells for clinical applications holds significant and unique challenges. The manufacturing workflow begins with collecting the
patient’s T lymphocytes and ends with an amplified population
of therapeutic CAR-T lymphocytes ready to be infused into
the patient as a living drug. The modification, activation, and
expansion of T lymphocytes require a high level of expertise
and sophisticated equipment that must meet the stringent
standards of quality control. Moreover, production of CAR-T cells
requires careful storage and handling to maintain stability of the
therapeutic cells and traceability.1
Improving CAR-T Production
Leukapheresis is the process by which mononuclear cells
(MNCs), including subsets of T cells, are collected from a target
patient, and it involves sustained bloodflow through closed-loop,
continuous or intermittent collection systems, and centrifugation.1
This is difficult in patients in advanced stages of malignancy who
have low peripheral blood access. Central venous catheterization
provides a steadier blood flow but introduces greater risk of
infection and trauma. In addition, chemotherapy and radiation
reduce lymphocyte count, and fewer T cells can be collected from
relapsed or refractory patients who have already passed through
multiple cycles of cytotoxic therapies.
The collected sample then needs to be enriched for T cells.
Density gradient centrifugation efficiently removes erythrocyte
and granulocyte contaminants. However, commonly used
Ficoll-Paque gradients are unable to separate T cells from
monocytes. Methods based on cell size and density (elutriation)
remove monocyte contaminants but tumor cells and unwanted
lymphocytes may be retained. Immunomagnetic beads can isolate
T-cell subsets with a high degree of specificity. Optimal methods
for enrichment depend on the quality of the starting sample.
Scaling up requires the validation of essential culture ingredients
and protocols and development of standard operating procedures
to ensure consistency and dosage specificity as per cGMPs.
The phenotypic heterogeneity of solid tumors makes them
difficult for CAR-T cells to detect. Fourth generation CARs
bypass this limitation by including co-stimulatory domains and
CAR-inducible IL-12.2 This allows NK cells and macrophages
to mount a second wave of attack against cancer cells that would
be undetectable by earlier generations of CAR-T cells. Another
recent advance involves the modification of CARs to induce
phagocytosis (CAR-P), directing macrophages to engulf target
cells including cancer cells.2
Quality Concerns in Clinical Applications
Quality management is regulated by the Foundation for
the Accreditation of Cellular Therapy (FACT) or the Joint
Accreditation Committee (JACIE), governed by the Center
for Biologics Evaluation and Research (CBER) of the Food and
Drug Administration (FDA) of the United States. Specifications
delineated in the FDA’s investigational new drug (IND)
application ensure continuous control, traceability, documentation,
standards, product safety, identity, purity, sterility, and potency
through large-scale clinical trials. Although clinical trials are
costly and time consuming, lengthening the time from bench to
bedside, they play a vital role in identifying product contaminants
and potency and lend insight into underlying mechanisms. Two
CAR T-cell therapies have been approved by the FDA, one for
pediatric acute lymphoblastic leukemia, and the other for adults
with advanced non-Hodgkin's lymphomas.
Once the therapeutic cellular product is formulated, samples
are tested for optimal dosage, route of delivery, and archiving.
Cells can be introduced either directly into cancerous tissue or
through intravenous infusions. Logistical concerns in this final
stage include optimizing methods of cryopreservation, storage,
packaging, transport, thawing, and infusion that preserve the
stability and efficacy of the therapeutic cells.3 Impressive clinical
responses have led the FDA to designate several CAR-T cell
therapies as breakthroughs in cancer treatment.4 For CAR-T
cell therapy to move from the echelons of frontier research to
being adopted as a viable option in cancer treatment globally,
CAR-T cellular products will need to be manufactured under
standardized, industry-grade conditions.
For references, please see page 7.
Efficiency
and Quality:
Manufacturing CAR-T
Cells for the Clinic7
AN INTRODUCTION TO CAR T-CELL PRODUCTION
Article 1 - Locked on Target: T-Cell Receptor-Mediated
Cancer Cell Killing
References
1. D. Wang, et al., “Glioblastoma-targeted CD4+ CAR T cells mediate
superior antitumor activity.” JCI Insight. 3:10, 2018.
2. M. Sharpe, et al., “Genetically modified T cells in cancer therapy:
opportunities and challenges.” Dis Model Mech. 8, 337-350, 2015.
3. S. Maude, et al., “Current status of chimeric antigen receptor therapy
for haematological malignancies.” Br J Haematol. 172, 11-22, 2016.
4. S. Ninomiya, et al., "Tumor indoleamine 2,3-dioxygenase (IDO) inhibits
CD19-CAR T cells and is downregulated by lymphodepleting drugs." Blood.
125, 3905-3916, 2015.
Article 2 - Start the CAR: Constructing CAR-T Cells
References
1. G. Gross, et al., "Expression of immunoglobulin-T-cell receptor
chimeric molecules as functional receptors with antibody-type
specificity." Proc Natl Acad Sci U S A. 86:10024-10028, 1989.
2. B. Ye, et al., "Engineering chimeric antigen receptor-T cells for cancer
treatment." Mol Cancer. 17:32, 2018.
3. J.A. Fraietta, et al., "Improving therapy of Chronic Lymphocytic
Leukemia (CLL) with Chimeric Antigen Receptor (CAR) T cells."
Semin Oncol. 43(2): 291-299, 2016.
Article 3 - Efficiency and Quality: Manufacturing CAR-T
Cells for the Clinic
References
1. U. Kohl, et al., "CAR T cells in trials: recent achievements and
challenges that remain in the production of modified T cells for
clinical applications." Hum Gene Ther. 29: 559-568, 2018.
2. M. Chmielewski, et al., "IL-12 release by engineered T cells
expressing chimeric antigen receptors can effectively muster an
antigen-independent macrophage response on tumor cells that have
shut down tumor antigen expression." Cancer Res. 71: 5697-5706, 2011.
3. P.P. Zheng, et al., "Approved CAR T cell therapies: ice bucket
challenges on glaring safety risks and long-term impacts." Drug Discov
Today. 23, 1175-1182, 2018.
4. J.A. Fraietta, et al., "Improving therapy of Chronic Lymphocytic
Leukemia (CLL) with Chimeric Antigen Receptor (CAR) T cells."
Semin Oncol. 43(2): 291-299, 2016.PHC Corporation of North America
1300 Michael Drive, Suite A, Wood Dale, IL 60191
Toll Free USA (800) 858-8442, Fax (630) 238-0074
www.phchd.com/us/biomedical
PHC Corporation of North America is a subsidiary of PHC Holdings Corporation, Tokyo, Japan, a global
leader in development, design and manufacturing of laboratory equipment for biopharmaceutical,
life sciences, academic, healthcare and government markets.
VIP® Series ultra-low temperature freezers provide reliable long-term storage
of immunotherapy and regenerative medicine specimens at -86°C.
Learn more about the benefits of purpose-built compressors and patented
VIP Plus composite thin-wall insulation in a high performance ultra-low
temperature storage system at www.phchd.com/us/biomedical/vip.
There is nothing sustainable
or energy efficient about
losing your life’s work.
Protection Without Compromise.
For a cost-effective alternative to LN2 vapor-phase storage, learn about our model
MDF-C2156VANC-PA air phase -150°C cryogenic freezer at www.phchd.com/us/biomedical.
ULT FREEZERS
MDF-U76VA
MDF-U76VC
MDF-U702VXC MDF-U502VH MDF-U702VH MDF-U5586SC MDF-U7586SC MDF-DU900VC MDF-DC700VXC
Select from five sizes of VIP Series freezers from
compact 3.0 cu.ft. to large 29.8 cu.ft. freezers
designed for high volume storage in biorepositories
and biobanking.
ULT FREEZERS
MDF-C8V1 MDF-U502VXC MDF-U53VA MDF-U76VA
MDF-U76VC
MDF-U302VX MDF-U702VXC MDF-U502VH MDF-U702VH MDF-U5586SC MDF-U7586SC MDF-DU900VC MDF-DC700VXC
ULT FREEZERS
F-C8V1 MDF-U502VXC MDF-U53VA MDF-U76VA
MDF-U76VC
MDF-U302VX MDF-U702VXC MDF-U502VH MDF-U702VH MDF-U5586SC MDF-U7586SC MDF-DU900VC MDF-DC700VXC
ULT FREEZERS
MDF-U502VXC MDF-U53VA MDF-U76VA
MDF-U76VC
MDF-U702VXC MDF-U502VH MDF-U702VH MDF-U5586SC MDF-U7586SC MDF-DU900VC MDF-DC700VXC
Brought to you by
Download the eBook for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.
Experiencing issues viewing the form? Click here to access an alternate version