T cell therapies Flashcards

1
Q

Animal evidence for immune system role in cancer

A
  1. Mice with impaired innate or adaptive systems have increased spontaneous carcinogen-induced cancers
  2. In wt mice, cancer cells with immunogenic mutations are rapidly eliminated
  3. Cancers that develop in immunodeficient mice retain immunogenic mutations and are rapidly rejected by normal mice
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q

Human evidence for immune system role in cancer

A
  1. Patients with AIDs or on immunosuppressants have more cancers (including relapse of long-dormant cancers)
  2. Donor-origin tumours develop in immunosuppressed recipients of transplanted organs from asymptomatic donors
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q

Evidence for equilibrium between cancer and immunity

A

Injected MCA carcinogen into mice -> some develop cancer.

Some develop small tumours that do not grow progressively but if immunosuppressive, they grow.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q

CTLA4 inhibitors

A

Ipilimumab in clinical trials caused cancers to get bigger, so trials stopped, however doctors realised treated patients did better. Growth due to initial inflammation, then regress.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

PD1/PDL1 inhibitors

A

[Hodi et al 2010] - when work, checkpoint inhibitors induce stable remission

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

Factors contributing to efficacy of checkpoint inhibitors

A

Is PDL1 present in the tumour? Mutational load, T cell tumour infiltrate

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

Increase efficacy of checkpoint inhibitors by pre-treating cancer

A

Experiment: Oncolytic virus mutated so does not infect normal cells (due to tumour suppressor genes). Inject into tumour -> infects -> inflammation and cell death -> increased antigen presentation of tumour cell peptides

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Cancer vaccines

A
  1. Vaccinate against classical tumour antigens

2. Neoantigens: sequence exome/ RNA of cancer and compare vs normal cells. Then identify those highly expressed on MHC.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

Evidence for neoantigen vaccines

A

In mouse mutagen model, neoantigen-expressing tumour cells provoke T cell response and are eliminated.

In human tumours, there is a correlation between mutational load and T cell infiltration/ killing

Checkpoint inhibitor efficacy correlated with mutational load

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

Sahin et al 2017

A

Clinical trial of RNA mutanome neoantigen vaccine
Identify mutations by sequencing, predicting MHC presentation.
Chose 8 mutations then made RNA vaccine specific to patients -> inject -> compare frequency of metastatic events before and after (since no other control).
Now in phase 2 clinical trials.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

Redirected T cell therapy strategies

A
  1. Bispecific T cell engagers (BiTE)
  2. Immune mobilising T cells against cancer (ImmTAC)
  3. TCR engineered T cells
  4. Other engineered T cell strategies
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

BiTE

A

Soluble protein which connects T cell and cancer cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

Blinatumomab

A

First and only approved BiTE. Is an anti-CD19 and anti-CD3 single chain antibody, use to treat B cell malignancies.
Engages CD19 in low nM range (requires 1000s molecules to target robustly). Binds CD3epsilon in low nM range.

Recruits CD4 and CD8 T cells independently of TCR. The response is not induced unless both ends of blinatumomab are engaged.

Leads to T cell activation, proliferation and cytokine production, with CD19 target cell killing. T cells serially kill.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q

Dreier et al 2003

A

Preclinical data for blinatumomab.

Co-cultured blinatumomab with T cells and CD19+ cells => potent killing.

  • 2:1 effector:target killing
  • 1000s of CD19 molecules on target cells required for killing

Prevented tumour formation in vivo

  • cohorts of immunodeficient mice inoculated with NALM6 (tumour) cells
  • human PBMCs from healthy donors given (vs control)
  • different doses of blinatumomab or PBS vehicle control given d0, 1, 2, 3, 4 following tumour cell/ PBMC injection
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

ImmTAC

A

Soluble TCR-based targeting

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

Challenges of ImmTAC

A
  1. TCRs are membrane bound => engineer to be soluble
  2. TCRs have natural low affinity to targets, esp cancer targets (due to -ve thymic selection)
    HLA expression down regulated in cancers
    => increase affinity of TCR for its pMHC targets.
  3. Soluble TCRs lack avidity provided by coreceptors.
    A high affinity stable TCR on its own could target a cancer call but not kill it.
    => fuse TCR to an effector molecule (which binds CD3)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

How to solubilise TCRs for ImmTAC

A

Truncate TM domains and relocate stabilising inter-chain disulphide bond.
Produce in bacteria so can increase quantity.
- the two mTCR chains, alpha and beta, are made in E coli as inclusion bodies, then refolded in vitro, purified
- mTCRs are stable at room temperature for months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

How to enhance TCR affinity for ImmTAC

A

TCR contacts pMHC via 6 CDRs- CDR3s mainly contact the peptide and CDR2s the HLA.

[Yi et al 2005] - Phage display to increase affinity. Random mutation of different CDR loops, then select CDR combinations.

Affinity enhanced TCRs show increased antigen residence times.
- Wt half-life 6s vs 8hrs for 3 mutant CDRs combined. Wt binds in micro molar range vs pico molar range for engineered TCR.

[Crean et al 2020] - Affinity-enhanced TCRs have increased buried hydrophobic content at the binding interface (likely entropically favourable due to expulsion of H2O molecules that typically surround hydrophobic groups). So brings existing TCR-pMHC contacts closer rather than making more contacts.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

How to enhance TCR affinity for ImmTAC

A

TCR contacts pMHC via 6 CDRs- CDR3s mainly contact the peptide and CDR2s the HLA.

[Yi et al 2005] - Phage display to increase affinity. Random mutation of different CDR loops, then select CDR combinations.

Affinity enhanced TCRs show increased antigen residence times.
- Wt half-life 6s vs 8hrs for 3 mutant CDRs combined. Wt binds in micro molar range vs pico molar range for engineered TCR. Does this actually increase sensitivity? -> modified version of KPR (modified receptors less sensitive than wt -> TCR internalisation?)

[Crean et al 2020] - Affinity-enhanced TCRs have increased buried hydrophobic content at the binding interface (likely entropically favourable due to expulsion of H2O molecules that typically surround hydrophobic groups). So brings existing TCR-pMHC contacts closer rather than making more contacts.

[Holland et al 2020] - TCR have different pMHC binding mode to Ab TCR mimetic scaffolds.
- Contact points much more dispersed with TCR vs Ab mimetic -> specificity advantage (e.g. over BiTE)

20
Q

How fuse TCR to effector molecule in ImmTAC

A

TCR fused to anti-CD3epsilon scFV (binds CD3 with nM affinity).
Non cancer-specific T cell then kills the cancer cell in an antigen-specific manner.

21
Q

Liddy et al 2012

A

ImmTAC potency is driven by affinity -> increases sensitivity => can target cancers expressing only a few target molecules.

Looked at dose-response (target peptide conc vs IFNy release from CD8 T cells).

22
Q

Damato et al 2019

A

ImmTAC Tebentafusp redirects CD8 T cells to kill melanoma cells, sparing antigen-negative bystander cells.
Expected to be approved this year for uveal melanoma.

23
Q

What next for soluble T cell redirection agents (BiTE and ImmTAC)?

A
  1. Solid tumours- targets different e.g. EpCAM, PSMA, CEA for BiTEs. Or cancer testis antigens (NY-ESO1, MAGE, PRAME) for soluble TCRs.
  2. Combination treatments to enhance effect
    - enhance T cell access to tumours
    - relieve inhibitory tumour microenvironment
24
Q

TCR engineered cells

A

CAR-T cells

Adoptive TCR T cell therapy

25
Q

Adoptive TCR T cell therapy

A

Extract patient immune cells, isolate T cells, edit to express chosen TCRs (e.g. via viral vector) -> expand -> deliver to patient.

4-6 weeks

TCR editing by phage display to increase affinity. Must test specific potency to avoid off-target effects.

Optimal affinity where enhance TCR recognition of cancer targets without compromising specificity

26
Q

Linette GP et al 2015

A

NY-ESO-1 specific TCR transducer T cells cross-reactive to Titin protein in HEP2 heart cells.
Measured T cell activation by cytokine production e.g. IL2, IFNy

27
Q

Afami-cel

A

Expected to be first TCR-engineered T cell therapy approved this year for synovial carcinoma.

Target Ag is MAGE-A4.

Overall 34% response rate (16/47), including 36% (14/39) synovial carcinoma and 25% for MRCLS (2/8)

28
Q

Challenges of adoptive TCR T cell therapy

A
1. Efficacy in solid tumours
=> most TCR-T cell therapies target HLA class I, so can engineer CD4 T cells to also express CD8
  1. Off-target toxicity
    => comprehensive preclinical safety analysis (empirical testing and in silico analysis to identify molecular mimicry peptides)
    * Mispairing? Possible that transgenic TCR chains may mispair with endogenous TCR chains -> off-target toxicity. Observed in mice, resulted in severe autoimmunity [Bendle et al 2010]
  2. Want off-the-shelf treatments
    => universal/ allogeneic T cells more scalable, cheaper, immediate access vs autologous T cells. But challenge of this is GVHD
29
Q

Other T cell redirection strategies

A
  1. HiT T cell

2. TRuC T cell

30
Q

HiT T cell

A

Natural TCRs that bind cell surface targets (not pMHC) independently of TCR expression.
Use phage display to optimise target affinity.

Preclinical data in mouse models demonstrate tumour response with mesothelia-targeting HiT T cells

31
Q

TRuC T cell

A

Fuse Ab binding domain to CD3epsilon.

Hypothesis: engagement of entire TCR complex may facilitate a broad and controlled response, allowing more potent tumour cell killing, faster migration to tumour and longer persistence.

A mesothelia TRuC is in clinical development with responses observed in ovarian cancer patients [Patrick A et al 2019]

32
Q

Discovery early 1990s

A

Zeta chain on TCR discovered. Has 3 ITAMs + short EC domain so difficult to study function by giving antibody.

Instead, fused zeta chain to other proteins for which they had Abs. Then activate T cell this way without engaging the TCR.

33
Q

Eshhar et al 1993

A

Made chimeric genes = single chain Fv domain of Ab linked to gamma or zeta chains.
Expressed chimeric genes as functional surface receptors in a cytosolic T cell hybridoma.
Triggered IL2 secretion upon entering Ag and mediated non-MHC restricted hasten-specific target-cell lysis.
Used FACS, immunoblotting/ gel electrophoresis.

34
Q

CAR T cell therapy success in CD19+ B cell leukaemia

A

Cancer caused by genetic modifications e.g. generation of fusion genes creating active kinases -> ^B cells.
Chemo to kill malignant BM cells + imatinib (kinase inhibitor).
Median 6 month survival in adults.

35
Q

Zhao et al 2015

A

Efficacy of CAR T cells with different signalling domains.

  • EC domain targeting CD19 fused to zeta chain
  • Murine model shows increased survival using CAR-T cells with co-stimulation
  • CD28 co-stimulation allows faster cancer killing by CAR-T cells but have shorter persistence
  • Authors used flow cytometry to show CAR and LNGFR expression. Measured cytotoxic activity using a (51)Cr release assay and bioluminescence assay (NALM6 as target cells)
36
Q

Brentjens et al 2013

A

Flow cytometry showed that after 11 days of treatment with CAR T cells for acute lymphoblastic leukaemia, B cell markers not detected.
This observation persists for 50 days.

37
Q

FDA approved 2 CAR T cell therapies for B cell leukaemia in 2017

A

Kymriah for ‘ALL’

Yescarta for B cell lymphoma

38
Q

CAR T cell therapy is the culmination of advances in several fields

A
  • Genetic engineering
  • Target selection (e.g. CD19 paradigm)
  • Tumour immunology
  • Synthetic receptor design
  • Cell manufacturing
39
Q

Park et al 2018

A

Combined analysis of all CAR-T cell patients in a particular hospital (n = 53).
14/53 -> severe cytokine release syndrome
44/53 -> complete remission
Overall median survival = 12.9 months (i.e. patients relapse)
Patients with a low disease burden (<5% of B< are active B cells) have median survival of 20.1 months

40
Q

Roybal et al 2016

A

Want to increase specificity of CAR T cells to selectively kill cancer cells.

(in vivo murine model)
Authors took Ag fragment, fused to IC domain of notch receptor (TF that is cleaved when recognises ligand) rather than zeta chain. Transduced a CAR in this cell.

CD19 -> up regulation of CAR. Mesothelia -> activate custom response.

No IL2 produced if just target one of the two antigens. High IL2 production when target both. Showed T cell activation through CAR-GFP expression and CD69.

Problems:

  1. If T cell in tissue expression both Ag, even if by different cells
  2. Whenever T cell expresses CAR, is liable to response to any cell expressing mesothelin. So authors measured CAR surface expression half life (with CAR-GFP) -> 8hrs.
41
Q

Wu et al 2015

A

CAR T cell therapy can cause life threatening cytokine storms so can increase safety though remote-controlled CAR T cell activation. For example, CAR with no ITAMs but has heterodimerisation domain so can only activate cell in presence of heterodimeriser small molecule ‘Rapalog’.

Authors took mice, injected with CD19+ and tumour cells (both with fluorescent protein), then give CAR T cells, with aforementioned architecture, that target CD19. Reported robust IL2 release only when had CD19 and dimeriser.

42
Q

Eyquem et al 2017

A

Want to increase potency, since many patients relapse. T cells thought to become exhausted by chronic stimulation, and normally TCR down regulation is a mechanism of reducing stimulation. Expression of TCRs (+CARs) partly regulated by production and Ag-induced internalisation/degradation. Viral transduction of CAR into T cells -> random integration (decoupled from normal TCR expression).

Authors used CRISPR in mouse model of B cell leukaemia to KO TCR and introduce CAR to TRAC locus so have TCR-CAR+ T cells.
Improved survival in mouse model of B-ALL. Normal CAR T cells -> mice die after ~20days; mice survive > 60 days when under control of TCR promotor. These CARs maintain a lower level of expression -> may prevent exhaustion.

43
Q

Extend Ag repertoire by targeting intracellular Ag

A
  1. Generate Ab specific for pMHC
    [Maus et al 2016] - made Ab that binds HLA-A2 expressing NY-ESO-1 peptide
  2. Generate a TCR-based CAR.
    [Stone et al 2014] - Studied in vitro and in vivo effectiveness of single chain TCR => growth of metastatic tumours significantly inhibited. Tumour cells which escaped were Ag-loss variants.
44
Q

Boice et al 2016

A

Want to target solid tumours. CAR T cells can be engineered to manipulate the tumour microenvironment, acting as ‘micro pharmacies’.

Used B cell lymphoma system. HVEM (TNF receptor) expression is dysregulated. HVEM binds the inhibitor receptor BTLA on B cells.
Knock-down of HVEM exacerbates the cancer.
Soluble HVEM treatment reduces lymphoma burden.
CAR-T cells engineered to constitutively secrete HVEM +> reduced tumour volume between than standard CAR T cells.

45
Q

Decrease costs of CAR T cell/ adoptive TCR T cell therapies

A

Autologous T cells expensive and not always available (e.g. if T cells depleted by radio/chemotherapy).
Allogeneic T cells -> rejection or GVHD?

[Qasim et al 2017]

  1. Deplete patients own cells with alemtuzumab which targets CD52
  2. Prepare T cells with CD52-KO, transduced with CAR and ‘suicide gene’ so could deplete specifically if adverse reaction.

Want target Ag to be expressed on cancer but not healthy cells. And ideally not be completely unique to one patient.

  • High specificity e.g. neoantigens, viral antigens, gremlin antigens e.g. NY-ESO-1 testis antigen (developmental Ag expressed in some cancers)
  • Low specificity e.g. tumour differentiation antigens (CD19, CEA), over expressed antigens (ERBB2 over expressed in epithelial tumours)