Cancer Immunotherapy 2020

You might also like

Download as pptx, pdf, or txt
Download as pptx, pdf, or txt
You are on page 1of 76

Immunoreceptor tyrosine-

based activation motifs


(ITAMs)

TCR signaling is initiated by two tyrosine kinases: the Src family kinase Lck and ZAP-70.
Activated Lck phosphorylates ITAMs in the intracellular tails of chains of the TCR complex. Doubly
phosphorylated ITAMs recruit ZAP-70 from the cytosol to the plasma membrane. Tyr 315, Tyr 319,
and Tyr 493 of ZAP-70 are phosphorylated by Lck to fully activate ZAP-70, enabling ZAP-70 to
phosphorylate two scaffold proteins, LAT and SLP-76, leading to the recruitment of effector proteins
that stimulate T cell activation.
CD28 engagement also prevents apoptosis and acts synergistically with CD3-dependent
signals to ensure correct T cell proliferation, differentiation and growth
Mechanisms of B-cell tolerance in bone marrow and periphery

Both clonal deletion and anergy are mechanisms utilized in the primary as well as
peripheral lymphoid organs. Strength of BCR signaling affects the fate of the B cells
undergoing tolerance.
Receptor editing occurs primarily in the bone marrow.
Transitional B cells are found in the bone marrow and spleen.
Peripheral tolerance is primarily found in splenic transitional B cells, which depends
on tonic BCR signaling thresholds and survival signals from BAFF.
Tumor expansion
Interaction between PD-1/PD-L1 and T cell immune response. T cells are primed
and activated through the interaction with antigen-presenting cells. T cells recognizes
tumor via MHC-antigen interaction. Tumor PD-L1 and PD-L2 is upregulated by
interferon γ released from activated CD8 T cells. PD-L1 in turn inhibits T cells via
PD-1.
History of T-cell therapy in cancer
Strategies for use of T-cell therapies for cancer
Regulation of T-cell activation.

Padmanee Sharma et al. Cancer Discov 2021;11:838-857

©2021 by American Association for Cancer Research


PITFALLS WITH TILs:
A) cells were unable to persist in vivo, despite concomitant
administration of IL2, a T-cell growth factor.

B) Surgical resection required (expansion while preserving function)

C) Feasibility
 The tumor needs to be resected for isolation and expansion of TILs

 The patient needs to be “suitable” to tolerate lymphodepletion and


IL2-based treatments and remain suitable during the several-week
period required to expand the cells in vitro

 Although TILs can be isolated from many cancers, to date, only those
from melanomas consistently possess specific cytolysis against the
tumors from which they were generated
 Not so feasible for solid tumors
Approach advantageous for tumor susceptible to checkpoint blockade
Advantageous for tumors arising from EBV or papilloma virus infections
Genetically modified T cells
Approaches on the basis of genetic modification of normal peripheral-blood T cells

Common approaches for redirecting T-cell specificity:

(i) gene modification with TCRs directed against tumor-associated antigens

(ii) introduction of a CAR (Chimeric Antigen Receptor)

Different types of antigens can theoretically be used to redirect autologous T cells


against tumor cells:

a) tissue-specific differentiation antigens [such as melanocyte differentiation antigens


(MDA); e.g., gp100 or MART1, in melanoma and CD19 in B-cell malignancies], cancer
testis (germ cell) antigens (such as NY-ESO-1), which are detected in many tumors but
not in normal adult tissues, with the exception of the testis

b) overexpressed self-proteins (such as HER2), mutational antigens (such as BRAF-V600E)

c) viral antigens (such as EBV in Hodgkin disease and HPV in cervical cancer).
The presence of epitope-responsive T cells in the peripheral repertoire is
dependent on:

(A) the generation of appropriate TCR-α/β chains upon somatic recombination of V(D)J
gene segments during T-cell development

(B) survival of developing T cells during positive and negative selection in the thymus

(C) in the case of antigen-experienced T cells, on successful priming and proliferation


of the mature T cells in the lymphatic system
Developing genetically engineered TCRs for tumour antigen detection

Step 1: Direct TCR cloning system that would allow the unbiased analysis of the TCR
repertoire

Step 2: The retrieval of antigen-specific TCRαβ pairs

Step 3: Characterization of their function for future TCR gene therapy

MHC tetramers are used to detect MHC-epitope


reactive TCRs from the pool of Peripheral T cells

MHC tetramers that are bioengineered to present a specific peptide can be


used to find T-cells with receptors that match that peptide

The tetramers are labeled with a fluorophore allowing tetramer-bound T-cells to be


analyzed with flow cytometry

If a tetramer stain specific for a pathogenic peptide results in a positive signal, this
may indicate that the person’s immune system has encountered and built a
response to that pathogen.
MHC tetramer production

First MHC heavy chains


expressing a bacterial BirA-
recognition site are
synthesised.

The MHC heavy chain is then


folded with β2-
microglobulin (β2M; the
MHC light chain) and
synthetic peptide.

The enzyme BirA is then


added to biotinylate the
complex, adding a biotin
molecule to each MHC
monomer that is formed.

In the presence
of streptavidin which has
four biotin binding sites per
molecule, four MHC
monomers are joined
together to form a tetramer.
Whilst single MHC: peptide complexes and TCR bind weakly, MHC tetramers can bind up to
four TCRs simultaneously, creating a much stronger interaction.

MHC tetramers only bind to the relevant TCR and tetramers specific for any MHC heavy
chain (e.g. HLA A2, HLA B7) can be produced, coupled with any peptide epitope of choice
(e.g. viral, bacterial, tumor and autoimmune antigens).

The streptavidin molecule is routinely linked to a fluorochrome such as phycoerythrin, to


allow
for direct staining for use with flow cytometry.
‘hTEC10’ (human TCR efficient cloning within 10 d)
Most current therapies utilizing engineered T cells process autologous peripheral blood T
cells that are targeted to tumor antigens following retroviral transduction of a TCR or a
chimeric antigen receptor (CAR).

Genetically modified T cells (TCRs or CARs) are transfected using virus vectors
(retroviruses or lentiviruses) or a transposon system (Sleeping Beauty). Following
transfection, genetically modified T cells are expanded and transferred into patients
treated with preconditioning lymphodepletion similar to that used with TIL protocols

Cloning of tumor specific TCRs


TCRs are typically cloned from patient tumor-reactive T cell clones

T cells recognize processed peptide antigens expressed in the context of MHC

Advantages of genetically engineered TCR expressing T


cells

1) these cells can be produced from peripheral blood T cells


(unlike TILs)
2) can “see” intracellular antigens (unlike CARs)
Disadvantages of genetically modified TCRs

 TCRs recognizing tumor-associated antigens expressed on


healthy tissues induced on-target toxicity.

MDAs such as MART1 and gp100 are expressed by normal


melanocytes present in the skin, retina, and inner ear.

TCR gene therapy against MART1 and gp100 resulted in skin rash,
uveitis, and hearing loss

 off-target toxicity

Multiple myeloma: MAGE-A3–specific TCRs. Patients had fatal


cardiac toxicity due to unexpected recognition by the MAGE-A3
TCR of titin, a cardiac peptide antigen

 Can be used only for MHC compatible patients


single-chain variable fragment (scFv): formed by a variable fragment of heavy
(VH) and light (VL) chains of a mAb and fused to a flexible linker
CAR is comprised of antigen-recognition domain (scFv), a hinge domain or
spacer (CD28, CD8α, CD7, IgG4, and IgG1), a transmembrane domain (CD28,
CD8α, CD7, CD4, CD3z, and OX40), a co-stimulatory domain (CD244, CD28,
CD27, OX40, ICOS, and CD137), and a signaling domain (CD3z, DAP10, and
DAP12).
The Role of OX40-Mediated Co-stimulation in T-Cell Activation and Survival

This process is
antigen dependent
Effects of OX40 ligation on effector versus regulatory T cells

Crit Rev Immunol . 2009 ; 29(3): 187–201.


CAR-T cells Manufacturing process
anti-CD3/CD28 antibody-
coated magnetic beads are
used for selection and ex
vivo T-cell activation

Expansion protocols for


CAR-T cells rely typically on
cytokines, such as IL-2, IL-7,
IL-15, and IL-21.

CAR-T therapy starts with accumulating the patient’s white blood cells by leukapheresis. The apheresis products are enriched
or deleted for a specific cell subset and then activated by one of following methods, including interleukins (IL-2, IL-7, and IL-
15), anti-CD3/CD28 antibody-coated magnetic beads, soluble CD3 antibody, artificial antigen-presenting cells, plate-bound
antibodies, and adhesion molecules.

The activated T cells are introduced with the CAR transgene through lentiviral or retroviral and non-viral methods
(electroporation of naked DNA and plasmid-based transposon/transposase).

Afterwards, the engineered-T cells undergo an expansion process in static culture bags or dynamic culture vessels or rotating
bioreactors. Eventually, cell numbers are calculated based on the patient’s disease burden, weight, and another formulation.
The CAR modified-T cells transfer to either a container for infusion purposes or cryopreserved for storage.
CAR-T cells for autoimmune disease

Multiple Sclerosis

Transduced CD4+ T cells using a lentiviral vector system encoding the myelin oligodendrocyte glycoprotein
(MOG) CAR gene and Foxp3 gene. The main role of MOG CAR-Tregs is to localize and bind to
MOG+ oligodendrocytes in CNS, providing a protective shield for these cells against immune attacks

T cell expressing CAAR specific for B cell receptors targets aberrant B cells.

Further, it inhibits B cell development from secreting autoreactive antibodies and prevents B cells from
presenting autoantigens to autoreactive T cells, which leads to suppression of T cell-mediated
autoimmune diseases.
CAR T cells
CAR T cells are constructed by fusing an antibody-derived single-chain variable
fragment (scFv) to T-cell intracellular signaling domains

Advantages:
a) T cells recognize cell surface antigens in a non–MHC-restricted manner

b) They do not depend on antigen processing and presentation.

c) CARs can be used to treat all patients in an HLA-unrestricted manner.

d) can target tumors that have downregulated HLA class I molecules or fail to process or
present proteins.

e) rapidly generate tumor-targeted T cells, bypassing the obstacles that preclude the
induction and execution of effective immune responses in vivo

f) Can be used allogenic and do not have genetic barriers (as off the shelf therapy)

Single chain variable fragment (scFv) is derived from a monoclonal antibody or an antigen-
binding region isolated from an immunoglobulin (Ig) heavy and light chain library
The identification of appropriate tumor antigens (TAA) is the greatest challenge
now facing researchers in the field of ACT. Identifying TAAs not present on
vital organs is critical.
The antigen recognition of T cells and CARs

(A) Antigens inside the cell are delivered to the cell surface as peptide fragments by the
MHC and T cells interact with antigens through the TCR. The subsequent costimulatory
signal is also necessary for optimal T-cell proliferation, differentiation, and survival. CD28
and 4-1BB interact with CD80 and 4-1BBL, respectively.
(B) CARs react directly with native tumor antigens expressed on the surface of a tumor cell
in a non-MHC-restricted manner. CARs are composed of antibody-binding domains
fused to T-cell signaling domains. The target binding module of the single-chain
antibody-variable fragment (scFv) are originated from a tumor-specific antibody
Limitations of CAR-T cells:
 tumor escape by loss of target expression

T-cell–driven selective pressure allows emergence of CD19-negative populations leading to


tumor escape. This has been observed in several patients with ALL treated with CD19 CARs.

 on-target/off-tumor toxicity

 cytokine release syndrome (CRS)

Manifestations of CRS include fever, increased cytokine concentrations (IL6, IFNγ),


hypotension, hypoxia, and neurologic symptoms

Solutions: treated with glucocorticoids and/or, preferentially, antibody to IL6 receptor


(tocilizumab)
Clinical trials

NY-ESO-1 TCR LARGE SCALE ENGINEERING OF T CELLS


CD19 CAR T cells
Problems with TCR based or CAR T cell therapies:

 Majority of clinical responses are short-lived

 ultimate tumor relapse

 relatively limited in vivo survival or suppression or exhaustion of


infused engineered T cells

 ex vivo culture in supra-physiologic concentrations of IL-2

Increases the cell numbers by several orders of magnitude.


But
driving cells to expand under these conditions “ages the cells from
a more naive and replicative phenotype to late-stage effectors”

another theoretical concern for the use of mature T cells as targets for anti-cancer TCRs is the
potential for mis-pairing: that is, the pairing of endogenous cellular TCR chains with
introduced transgenic chains.
Immunosuppressive state of effector CD8þ T cells in the tumor microenvironment

How tumors ‘‘escape’’ the immune system?

Reduction of MHC expression diminishes presentation of target antigens.

The expression of Fas ligand on tumor cells induces apoptosis in CTLs.

TGFb and IL-10 drive the differentiation of CD4+ T cells toward the regulatory phenotype which results in
T-cell inhibition.

The engagement of PD-1 with its ligand programmed cell death protein ligand 1 (PD-L1) inhibits the
proliferation and effector function of T cells through the recruitment of SHP2, which inactivates TCR
mediated signaling
T-cell exhaustion during chronic antigen exposure

The presentation of antigenic peptides in the context of MHC class I triggers the priming of naive CD8 + T cells and
their differentiation into effector CD8+ T cells. After antigen clearance, effector CD8+ T cells differentiate into
memory CD8 T cells. In contrast, during continuous exposure to antigen, T cells gradually lose effector functions
and proliferation capacity and finally are eliminated via apoptosis. The gain of cell surface inhibitory receptors
such as PD-1 correlated with T-cell exhaustion reduces antitumor immune function.
Different germ line recombination possibilities results in a huge TCR a and b chain repertoire
T cells undergo positive and negative selection in the thymic cortex and medulla,
respectively
mTECs: Medullary
Thymic Epithelial cells

Positive selection involves the


selection of cells which are
functional

A promiscuous
expression of a wide
array of self-antigens
in the thymus is
essential for the
negative selection of
self-reactive T cells
and the
establishment of
central tolerance.
Autoreactive T cells are negatively selected in the medulla through mTEC expression of self-antigens
such as tissue-restricted antigens (TRAs)

TRAs are promiscuously expressed under the control of the autoimmune regulator Aire and by the transcription factor Fezf2 in mTECs
POSITIVE and Negative SELECTION OF T CELLS

Double positive abTCRlow cells must successfully undergo positive and negative selection before they can
leave the thymus

Cells which have successfully rearranged abTCR will die in the thymus cortex if they do not bind self
MHC within 3-4 days

Positive selection occurs when double positive T cells bind cortical epithelial cells expressing Class I or
Class II MHC plus self peptides with a high enough affinity to get the survival signal.

Positive selection also determines whether the T cell will become a helper or a cytotoxic T cell. Positive
selection on Class I MHC will produce a CD8 Tc cell, while positive selection on Class II MHC will yield a
CD4 Th cell.

Expression of co-receptor (CD4 or CD8) and its binding to MHC is also required for positive selection; mice
expressing a defective CD4 that cannot bind Class II also fail to produce mature Th cells.

Negative selection occurs when double positive T cells bind to bone-marrow derived APC (macrophages
and dendritic cells) expressing Class I or Class II MHC plus self peptides with a high enough affinity to
receive an apoptosis signal. Note that selection occurs on self peptides in the thymus; MHC presents self
peptides in the absence of pathogen.
Development of T cells (milestones):

 HSC-derived thymus-seeding progenitors (TSPs) migrate into the thymus and


differentiate into an Early Thymic Progenitor (ETP) upon rearrangement of the
diversity (D) and joining (J) regions of the TCR β locus.

 ETPs progress to a pre-T cell state expressing CD1a and CD5. At this stage,
recombination of the variable (V) region of the TCR β locus to form a complete
rearranged VDJ TCR β locus occurs almost simultaneously with the rearrangement of
the gene segments encoding the γδ TCR.

 Depending on the outcome of the TCR segment rearrangements, the cells can then
follow an αβ or a γδ differentiation path.
 A successful TCR β rearrangement leads to the process of β-selection and
emergence of a CD4+ immature single positive (ISP) T cell.

 The CD4 ISP cell then develops into a double-positive (DP) cell that expresses
both CD4 and CD8 and has begun to rearrange the V and J regions of the TCRα
locus.

 Clonotypic TCRs after positive and negative selection


The life span of DP thymocytes is limited as they quickly proceed to apoptosis if
they do not receive a TCR-mediated survival signal provided by the self-HLA molecules
of the thymic epithelium before maturing into CD4 +CD8− and CD4− CD8+ single-
positive (SP) T cells.
Emergence of single positive T cells T cell priming and expansion
after positive selection

 The resulting T cells are “self-restricted and tolerant” of self tissues.

 The newly generated T cell clones, known as naive T cells, initially circulate throughout
the body at low frequency.

 Antigen specific T cell expansion/priming

Upon encountering antigen, T cells expand and acquire effector and/or memory functions
This T cell priming requires
a) TCR engagement by HLA-peptide complexes on APCs
b) Concomitant ligation by co-stimulatory ligands on APCs
Human T cell development
Memory T cell subtypes:

Central memory T cells (TCM cells)

Express CD45RO, C-C chemokine receptor type 7 (CCR7), and L-selectin (CD62L)
Central memory T cells also have intermediate to high expression of CD44
Commonly found in the lymph nodes and in the peripheral circulation

CD44 expression is used to distinguish between naïve from memory T cells

Effector memory T cells (TEM cells and TEMRA cells)

Express CD45RO but lack expression of CCR7 and L-selectin.


Have intermediate to high expression of CD44
“Lack lymph node-homing receptors” and are thus found in the peripheral circulation
and tissues

Stem memory TSCM cells


Like naive T cells, TSCM cells are CD45RO−, CCR7+, CD45RA+, CD62L+ (L-selectin), CD27+,
CD28+ and IL-7Rα+
Express large amounts of CD95, IL-2Rβ, CXCR3, and LFA-1, and show numerous functional
attributes distinctive of memory cells
These cells are capable of generating TCM and
In preclinical studies, adoptively transferred T confer
TEM subsets
superior
while maintaining themselves
immunity compared with other T memory subsets
SCM
T-cell memory is organized in subsets that are selectively distributed
in the body according to their effector functions
Different signatures of T cells with defined cytokine potential
Exploitation of fully rejuvenated CTLs (rejCTLs) from iPSCs

Reprogramming via iPSC technology generates T-iPSCs from antigen-specific CTLs. These T-iPSCs can
redifferentiate into CTLs that retain ancestral antigen specificity and cytotoxicity. Rejuvenated CTLs have higher
proliferative capacity and longer telomeres than do the original exhausted CTLs
TCR genes from original CTLs are retained in T-iPSCs

The TCR a- and b-chain genes (TCRA and TCRB) are rearranged during normal ab T-cell
development in the thymus.

Germline-configuration TCR a and b loci are present in iPSCs derived from CTLs, which
retain the same rearranged sequences of TCR a- and b-chain genes as those in the original
antigen-specific CTLs.

CDR3, the most diversifiable antigen-recognition site of the TCR, spans the
V(D)J-junction region, where several random nucleotides are inserted.

CDR3 sequences from T-iPSCs and their original CTL clones were completely identical at
both the TCRA and the TCRB loci, evidence that antigen specificity encoded in the genomic
DNA of original CTLs is retained throughout cloning, reprogramming, and expansion.
TCR genes from original CTLs are retained in T-iPSCs

The TCR a- and b-chain genes (TCRA and TCRB) are rearranged during normal ab T-cell
development in the thymus.

Germline-configuration TCR a and b loci are present in iPSCs derived from CTLs, which
retain the same rearranged sequences of TCR a- and b-chain genes as those in the original
antigen-specific CTLs.

CDR3, the most diversifiable antigen-recognition site of the TCR, spans the
V(D)J-junction region, where several random nucleotides are inserted.

CDR3 sequences from T-iPSCs and their original CTL clones were completely identical at
both the TCRA and the TCRB loci, evidence that antigen specificity encoded in the genomic
DNA of original CTLs is retained throughout cloning, reprogramming, and expansion.
TCR rearrangements in iPSC-derived CTLs during
lymphoid differentiation
Concern

Additional rearrangement of TCRA may occur randomly during lymphoid differentiation in


iPSC-derived CTLs

Such TCR rearrangements of TCR may give rise to nonspecific polyclonal T cells that
could occasion ‘‘autoimmune’’ reactions if administered

Successful interaction of the TCR with MHC molecules during thymic selection
downregulates the expression of the recombination activating genes 1 and 2 (RAG-1 and
RAG-2) and thereby prevents further endogenous TCRA rearrangements

# Trials of RAG down-regulation with inhibitors or with gene-editing techniques such as


those involving CRISPR/Cas9 to stop further TCRA rearrangement are under way

Redifferentiated CD8+ CTLs bind to multimers of their respective target MHC-peptide


complex and express the desired TCRs
Generation of antigen-specific CTLs from T-iPSCs: methods
Step I: Antigen-specific CTL clones first are established from antigen-specific CTLs obtained
from peripheral blood. These clones are reprogrammed into iPSCs (T-iPSCs) by transduction
with vectors that contain the four Yamanaka reprogramming factors.

Step II: Hematopoietic differentiation

a) T-iPSCs are cultured on C3H10T1/2 feeder cells for 2 weeks, yielding iPSC-derived sacs that contain
many hematopoietic progenitor cells

b) Hematopoietic progenitor cells extracted from sacs are transferred onto Notch-ligand-expressing
CH310T1/2 feeder cells

c) After 28 days of culture, T-lineage cells are harvested and stimulated, yielding huge numbers of
rejuvenated antigen-specific CTLs that were generated from T-iPSCs.
Generation of antigen-specific CTLs from T-iPSCs: methods

+ Fms-related tyrosine kinase 3


ligand, stem cell factor, and IL-7
+ Vascular growth factor

mixed with irradiated peripheral blood


mononuclear cells, and stimulated with
IL-7, IL-15, and phytohemagglutinin
After 28 days T cell
lineage cells harvested

Step III: antigen specificity of these iPSC-derived rejCTLs is determined by multimer staining
and interferon-g enzyme-linked immunospot assays used to confirm that the specificity of
the original CTL clone is maintained.

Step IV: Banking of T-iPSCs.


Authentication of rejuvenated CTLs:

 although the original CTL clone expressed PD-1, a marker


of exhausted T cells, iPSC-derived rejCTLs did not.

 rejCTLs included central-memory-phenotyped cells (CCR7, CD27 and CD28)

Single unifying theme for all memory T cell subtypes:

a) they are long-lived

b) can quickly expand to large numbers of effector T cells upon re-exposure to their
cognate antigen. By this mechanism they provide the immune system with
"memory" against previously encountered pathogens.

c) Memory T cells may be either CD4+ or CD8+ and usually express CD45RO
Despite having the functional capacity of memory cells, T SCM cells retain a core
of genes expressed by TN cells and share the recirculation patterns and
distribution of TN cells in vivo
Specific characteristics of iPSC-derived T cells

 Specific to iPSC-derived T cells (antigen-specific CTLs and CAR-expressing T cells) is the


expression of CD3, TCRab, CD56, and CD8a, but not CD8b)
Suicide-gene based safeguards in iPSC-derived T-cell therapy

fusion protein iC9: An FK506-binding


protein (FKBP12) replaces the caspase
recruitment domain, allowing
conditional dimerization

Activation of iC9 with a specific


chemical inducer of dimerization (CID)
initiates a caspase cascade, activates
caspase-3 directly, and induces
apoptosis swiftly in iC9-transduced cells

Induction of apoptosis by activating iC9

Once the tumor burden is relived these cells can be induced to undergo suicide

In the intrinsic cell death pathway, activated BAX or BID translocates to the mitochondria,
forming pores that allow cytochrome C to move from mitochondrial intermembrane space
to cytosol. There, cytochrome C activates caspase-9, which in turn activates caspase-3 and
leads to apoptosis.
Hot tumours have immune infiltration whereas cold tumours' are
not infiltrated by immune cells in particular T cells

T cells

'Hot' versus 'Cold' Tumors. Images of tumors representing three patterns of T


infiltration visualized by CD3 + staining of T cells
Oncolytic viruses make tumors 'hot' and suitable for checkpoint blockade cancer
immunotherapies.

OV: Oncolytic virus; NK cell: Natural killer cell; PD-1: Programmed death-1; PD-L1:
Programmed death ligand-1; CTLA-4: Cytotoxic T-lymphocyte-associated protein 4; IFN-
a/b: Interferon-alpha or beta; CCL3/4: Chemokine (C-C motif) ligand 3 or 4.
Mechanism of converting a cold tumour to hot tumour by
Oncolytic Viruses

Immune checkpoint blockade is inefficient in 'cold' tumors, which are poorly


infiltrated by immune cells and also have low expression of PD-L1 on their surface. In
the absence of available targets, immune checkpoint blockers like anti-PD-L1 (targeting
PD-L1 expressed at the surface of cancer cells or on antigen-presenting cells), alone or in
combination with anti-CTLA-4, remain therapeutically inefficient

Therapeutic administration of oncolytic viruses (OV) into tumors promotes strong


antiviral immune response accompanied by the production of cytokines such as type-1
interferons and chemokines.

Type-1 interferons promote the expression of PD-L1 on the surface of cancer cells, while
chemokines like CCL3 and CCL4 attract immune cells which often express PD-1 or
CTLA-4.

##Thus, antiviral immunological events inflame the tumor and make it 'hot'. When
checkpoint inhibitors are administered subsequently, they can bind to their respective
targets on either cancer or immune cells. As a final result, oncolytic viruses sensitize
tumors to the therapeutic effects of immune checkpoint inhibitors
Schematic showing the engineering of Talimogene laherparepvec (T-VEC)

The backbone is the JS17 strain of herpes simplex virus, type 1 in which the two viral ICP34.5 genes have bee
deleted and replaced with copies of the hGM-CSF genes under control of a CMV promoter. In addition, th
viral ICP47 gene is deleted transitioning the viral US11 gene to an immediate-early promoter.
Converting Cold into Hot Tumors by Combining Immunotherapies

Talimogene laherparepvec is a human


herpes simplex virus (HSV) that selectively
infects and replicates in human cancer cells

Schematic Course of Events following Oncolytic Virus and Anti-PD-1 Treatment in Metastatic Melanoma

You might also like