/
Immuno-oncology: immunomodulation and checkpoint inhibition Immuno-oncology: immunomodulation and checkpoint inhibition

Immuno-oncology: immunomodulation and checkpoint inhibition - PowerPoint Presentation

oryan
oryan . @oryan
Follow
350 views
Uploaded On 2022-04-07

Immuno-oncology: immunomodulation and checkpoint inhibition - PPT Presentation

Introduction Cancer immunotherapy aims to generate or augment an immune response unlike conventional treatments that directly target features of cancer cell growth such as survival proliferation or metabolism ID: 910506

immune cell cells cancer cell immune cancer cells 2019 checkpoint death ici inhibitor tumour protein myeloid programmed icis 2018

Share:

Link:

Embed:

Download Presentation from below link

Download Presentation The PPT/PDF document "Immuno-oncology: immunomodulation and ch..." is the property of its rightful owner. Permission is granted to download and print the materials on this web site for personal, non-commercial use only, and to display it on your personal computer provided you do not modify the materials and that you retain all copyright notices contained in the materials. By downloading content from our website, you accept the terms of this agreement.


Presentation Transcript

Slide1

Immuno-oncology:immunomodulation and checkpoint inhibition

Slide2

IntroductionCancer immunotherapy aims to generate or augment an immune response, unlike conventional treatments that directly target features of cancer cell growth such as survival, proliferation or metabolism

1

Checkpoint inhibition

is a form of immunomodulation that uses monoclonal antibodies to block inhibitory receptors on T cells and promote T cell activation, and is of considerable therapeutic interest1Targeting the immune system in this way is associated with a robust response that has the potential to stimulate persistent tumour regression mediated by immunological memory1,2A number of different strategies are under investigation to optimise outcomes with immunotherapeutic agents

1. Khalil DN, et al. Nat Rev Clin Oncol 2016;13(5):27390; 2. Locy H, et al. Front Immunol 2018;9:2909.

Slide3

Targeting immune checkpoint pathways in cancer

Slide4

Tumour cells suppress the antitumour immune response by inactivating T cellsT cell activation in response to tumour antigens is a complex process involving two independent pathways1Signal 1: MHC-mediated T cell receptor signalling in response to antigen stimulation

Signal 2: costimulation of the CD28 receptor by B7 ligands on APCs

Activated T cells drive

cytotoxic cell-mediated immunity, augment antibody responses and engage accessory cells such as macrophages2The activity of T cells is limited by inhibitory receptors, which downregulate signal 2 to maintain immune homeostasis and prevent autoimmunity3Tumours evade T cell immunosurveillance by activating inhibitory receptors, resulting in T cell apoptosis and depletion41. Bretscher PA, et al. Immunology 1999;96(1):18590; 2. Alberts B, Johnson A, Lewis J. Molecular Biology of the Cell. 4th

rev. ed. New York: Garland Science, 2002.3. Buckner JH, et al. Arthritis Res Ther 2004;6(5):21522; 4. Angelousi A, et al. Neuroendocrinology 2018;106(1):89100; 5. Sharma A, et al. Clinical Immunology Principles and Practice. 5

th

ed. Elsevier Health Sciences, 2019

.

APC, antigen-presenting cell; CD, cluster of differentiation; CTLA-4, cytotoxic T-lymphocyte-associated protein; MHC, major histocompatibility complex.

Figure adapted from Sharma A, et al.

2019.5

Inhibitory receptors downregulate T cell activity5

Slide5

Targeting lymphoid immune cells has been shown to elicit an antitumour responseImmune modulation targets immune cells rather than cancer cells to inhibit tumour progression

1

Immune modulation with

ICIs upregulates the immune response by blocking inhibitory receptors1,2Inhibitory immune checkpoints include PD-1, CTLA-4, LAG-3, TIGIT and TIM-3, among others1,2Checkpoint inhibition is not necessarily specific to any cancer type, as shown by the approval of ICIs across a range of cancer types1,41. Naidoo J, et al. Br J Cancer 2014;111(12):221419; 2. Qin S, et al. Mol Cancer 2019;18;155; 3. Dyck L, et al. Eur J Immunol 2017;47(5):76579; 4. Khalil DN, et al. Nat Rev Clin Oncol 2016;13(5):273

90; 5. PDQ Cancer Information Summaries. Bethesda (MD): National Cancer Institute (US), 2002.APC, antigen-presenting cell; CTLA-4, cytotoxic T-lymphocyte-associated protein 4,

ICI, immune checkpoint inhibitor; LAG-3, lymphocyte-activation gene 3; PD-1, programmed cell death protein 1; PD-L1, programmed cell death-ligand 1; TIGIT,

T cell immunoreceptor with Ig and ITIM domains; TIM-3,

T cell immunoglobulin and mucin-domain containing-3.

Figure adapted from PDQ Cancer Information Summaries.

2002.

5

Upregulating the immune response with checkpoint inhibitors5

Binding of PD-L1 antigen to PD-1 blocks T cell mediated killing of cancer cell

Blocking PD-1 or PD-L1 reinvigorates the T cell response

Slide6

Inhibitory receptors on myeloid cells are also of interest as therapeutic targetsCancer-associated inflammation upregulates production of immunosuppressive myeloid cells including MDSCs and TAMs1

MDSCs suppress effector T cells and

NK cells, and express

high levels of PD-L12The potent immunosuppressive effect of myeloid immune cells is a major barrier to cancer immunotherapy1This has generated interest in ICIs that target myeloid immune cells, and combination treatments with other myeloid and/or lymphoid modulators1,21. Nakamura K, et al. Cell Mol Immunol 2020;17(1):112; 2. Park SM, et al. Arch Pharm Res 2019;42(7):56066.

ICI, immune checkpoint inhibitor; MDSC, myeloid-derived suppressor cell; NK, natural killer; PD-L1, programmed cell death-ligand 1; TAM, tumour-associated macrophage; TME, tumour microenvironment. Figure adapted from Nakamura K, et al. 2019.1

Cancer-associated inflammation drives

myeloid immunosuppression

1

Slide7

In recent years, ICIs have emerged as front-line treatment for a range of different cancer typesA number of different ICIs have been approved as first- and later-line options in a wide range of cancer types, including

metastatic melanoma, non-small cell lung cancer, renal cell carcinoma

and

bladder/urothelial cancer151. Park Y-J, et al. Exp Mol Med 2018;50(8):109; 2. Alsaab HO, et al. Front Pharmacol 2017;23;8:561; 3. Korman AJ, et al. Adv Immunol 2006;90:297339; 4. Boehringer Ingelheim. Data on file; 5. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/results?cond=checkpoint+inhibitor&Search=Apply&recrs=a&recrs=f&recrs=d&age_v=&gndr=&type=&rslt= (Accessed: February 2020).

CTLA-4, cytotoxic T-lymphocyte-associated protein 4, ICI, immune checkpoint

inhibitor;

PD-1

,

programmed cell death protein

1; PD-L1, programmed cell death-ligand 1

.

Slide8

Dual targeting of different inhibitory pathways may augment treatment outcomes with ICIsPrimary and acquired

treatment resistance limit the effectiveness

of ICI monotherapy

1The synergistic effect of targeting different immune inhibitory pathways has the potential to overcome resistance and improve outcomes1The therapeutic potential of ICI combinations has been validated by the approval of the anti-CTLA-4/anti-PD-1 combination regimen in a range of different indications21. Fares CM, et al. Am Soc Clin Oncol Educ Book 2019;39:14764; 2. Rotte A, et al. J Exp Clin Cancer Res 2019;38(1):255;

3. Marin-Acevedo JA, et al. J Hematol Oncol 2018;11(1):39.APC, antigen-presenting cell; CD, cluster of differentiation; CTLA-4, cytotoxic T-lymphocyte-associated protein 4

;

ICI,

immune checkpoint inhibitor; KIR, killer Ig-like receptors; LAG-3

, lymphocyte-activation gene 3; MHC, major histocompatibility complex; PD-1, programmed cell death protein 1; PD-L1/2, programmed cell death-ligand 1/2; TIGIT, T cell immunoreceptor with Ig and ITIM domains;

T cell immunoreceptor with Ig and ITIM domains; TIM-3, T cell immunoglobulin and mucin-domain containing-3; TME, tumour microenvironment.Figure adapted from Marin-Acevedo JA, et al. 2018.3

Immune interactions between APCs or tumour cells and T cells

3

Slide9

ICIs in combination with cancer cell-directed therapies may also have synergistic potentialCancer cells use many pathways to sustain growth, replicate, and resist cell death1

Combining ICIs with cancer cell-directed therapies

has the potential to augment the antitumour effect through discrete targeting

2,3Inhibition of the Wnt/-catenin pathway may improve outcomes with checkpoint inhibitors by improving T cell infiltration and priming3p53-targeted agents may augment outcomes by sensitising tumours to ICIs41. Hanahan D, et al. Cell 2011;144(5);646–74; 2. Patel SA, et al. Immunity 2018;48(3):41733; 3. Gopalkrishna S, et al. J Hematol Oncol 2017;10:101; 4. Kim SS, et al. Oncoimunology 2018;7(10):e1484982

.ICI, immune checkpoint inhibitor; KRAS; Kirsten Rat Sarcoma; MYC, MYC proto-oncogene, basic helix-loop-helix transcription factor.

Activation of cancer

cell signalling

Potential targets

Cancer

hallmarks

Upregulation of oncogenic

gene expression

Evading cell

death (including immunogenic cell death)

KRAS

ErbB

Wnt/

β

-catenin

Cell death pathways

p53

MYC

Epigenetic regulators

Slide10

Combinations of ICIs with novel targets

Slide11

Novel lymphoid and myeloid checkpoint-related agents could improve outcomes with approved immunotherapiesThe TME promotes T cell impairment

and immune escape

1

Novel targets in the lymphoid and myeloid TME are being explored in combination with anti-CTLA-4 and anti-PD-1/PD-L1 agents1Combinations with novel targets have the potential to counteract immune escape with acceptable toxicity profiles11. Khair DO, et al. Front Immunocol 2019;10:453; 2. Roslan Z, et al. J Oncol 2019;4536302; 3. Shangary S, et al. Clin Cancer Res 2008;14(17):5318

24.

AE, adverse event

;

Ang 2, angiopoietin

2; CD47, cluster of differentiation 47; CTLA-4

, cytotoxic T-lymphocyte-associated protein 4; DIABLO, direct inhibitor of apoptosis-binding protein with low pI; ICI, immune checkpoint inhibitors; LAG-3, lymphocyte-activation gene 3; LRP, low density lipoprotein receptor-related protein; MDM2, mouse double minute 2 homolog; PD-L1

, programmed cell death

ligand;

SIRP

α

, signal regulatory protein-

α

;

Smac, second mitochondria-derived activator of caspase; TIGIT, T cell immunoreceptor with Ig and ITIM domains; TIM-3, T cell immunoglobulin and mucin-domain

containing-3; TME, tumour

microenvironment; VEGF, vascular endothelial growth

factor.

Figure

adapted from Khair DO, et al. 2019.

Examples of novel lymphoid and myeloid targets in the TME

1

3

Slide12

SIRPα inhibition may have a synergistic antitumour effect when combined with ICIsInfiltrating

myeloid cells promote immune evasion

, and this has generated interest in

myeloid-immune targets1,2The CD47–SIRPα interaction transduces inhibitory signals on macrophages and other myeloid cells2Preclinical studies have indicated that CD47 or SIRPα blockade in combination with ICIs may have a synergistic antitumour effect31. Khair DO, et al. Front Immunocol 2019;10:453; 2. Weiskopf K, et al. Eur J Cancer 2017;76:1009; 3. Murata Y, et al. Cancer Sci 2018;109(8):2349

57; 4. Boehringer Ingelheim. Data on file.CD, cluster of differentiation; ICI, immune checkpoint inhibitor; SIRP

α

, signal regulatory protein-

α

.

The use of SIRP

α antagonists to enhance antitumour immunity is currently being explored

4

Slide13

Combining ICIs with p53-targeted therapy may augment the innate and adaptive immune response to cancerMDM2 inhibits the tumour suppressor p53, which is critical for regulating genes involved in DNA repair, senescence and apoptosis

1

When amplified,

MDM2 facilitates p53 degradation, promoting the development and maintenance of many cancers1Reactivation of p53 can promote innate and adaptive immunity, and decrease expression of PD-L12The cytotoxic potential of a PD-1 inhibitor in combination with p53-targeted therapy is currently being explored in clinical trials31. Kato S, et al, 2018. JCO Precis Oncol 2018;2018. 10.1200/PO.17.00235; 2. Sanz G, et al. J Mol Cell Biol 2019;11(7):58699; 3. Fang DD, et al. . Abstract 3192 at American Association for Cancer Research 2019.

https://cancerres.aacrjournals.org/content/79/13_Supplement/3192 (Accessed: February 2020).ICI, immune checkpoint inhibitor; MDM2, mouse double minute 2 homolog; MHC, major histocompatibility complex; NK, natural killer; PD-1, programmed cell death protein 1; PD-L1, programmed cell death-ligand 1.

Figure adapted from Sanz G, et al.

2019.

2

p53 promotes senescence and apoptosis, and the release of cytokines from senescent cells activates immune cells

2

Slide14

LRP 5/6 receptor antagonists inhibit Wnt-induced tumour growth and are being explored in combination with an ICIThe Wnt/

-catenin pathway

is one of the most important oncogenic pathways related to immune evasion

1Wnt receptor LRP5 and LRP6 expression promotes Wnt signalling1,2An LRP5/6 antagonist has been developed, and is a potent blocker of Wnt signalling2The effectiveness of combining an LRP5/6 antagonist with a PD-1 inhibitor is being investigated21. Pai SG, et al. J Hematol Oncol 2017;10:101; 2. Zinzalla V, et al. Abstract DDT01-01 at American Association for Cancer Research 2019. https://cancerres.aacrjournals.org/content/79/13_Supplement/DDT01-01 (accessed: February 2020

).ICI, immune checkpoint inhibitor; LEF, lymphoid enhancer factor; LPR, low-density lipoprotein receptor; LRP, low density lipoprotein receptor-related protein 1; P, phosphoryl group; PD-1, programmed cell death protein 1; TEF, transcriptional enhancer factor.

Figure adapted from Pai SG, et al.

2017.

1

LRP5 and LRP6 are transmembrane proteins

that bind Wnt

ligand and drive transcription of

Wnt target genes1

Slide15

VEGF suppresses the tumour immune response and combining anti-VEGF agents with ICIs may augment their activityAngiogenesis suppresses the immune response

to cancer cells

1

Anti-angiogenic treatments may therefore encourage a shift to an immunosupportive state1Anti-VEGF/Ang2 agents promote DC differentiation and decrease the accumulation of MDSCs1A PD-1 ICI combined with an anti-VEGF agent was superior to monotherapy in a clinical study (NCT02853331), and received FDA approval2,3Other trials are ongoing41. Yi M, et al. Mol Cancer 2019;18:60; 2. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT02853331 (Accessed: February 2020); 3. Food and Drug Administration. FDA approves pembrolizumab plus axitinib for advanced renal cell carcinoma. https

://www.fda.gov/drugs/drug-approvals-and-databases/fda-approves-pembrolizumab-plus-axitinib-advanced-renal-cell-carcinoma (Accessed: February 2020); 4. ClinicalTrials.gov. https://clinicaltrials.gov/ct2/show/NCT03468426 (Accessed: February

2020).

Ang 2, angiopoietin

2; DC

, dendritic cell; FDA, US Food and Drug Administration; ICI

, immune checkpoint inhibitor; MDSC, myeloid-derived suppressor cell; PD-1, programmed cell death protein

1; PD-L1, programmed cell death-ligand 1; TAM, tumour-associated macrophage; Treg, regulatory T cell; VEGF, vascular endothelial growth factor.Figure adapted from Yi M, et al. 2019.1

Angiogenesis contributes to immunosuppression

through multiple mechanisms

1

Slide16

Smac/DIABLO is a proapoptogenic protein that is being explored in combination with ICIsResistance to apoptosis is a key driver of cancer development, generating interest in mimicking endogenous promoters of apoptosis

(including components of the Smac/DIABLO pathway)

1,2

Smac mimetic compounds sensitise tumours to TNF-α-induced apoptosis and block TNF-α-induced growth3Smac mimetic compounds have demonstrated a synergistic effect in combination with ICIs in preclinical studies31. Philchenkov A, et al. Crit Rev Oncog 2016;21(34):185202; 2. Martinez-Ruiz G, et al. J Exp Clin Cancer Res 2008;27(1):48; 3. Beug ST, et al. Nat Commun 2017;8. doi: 10.1038/ncomms14278; 4. Dougan SK, et al. Sci Signal 2019;12(596):1

2.BAFF, B cell-activating factor; CD, cluster of differentiation; DIABLO, direct inhibitor of apoptosis-binding protein with low pI; IL, interleukin; NKT, natural killer cell; Smac, second mitochondria-derived activator of caspase; ICI,

immune checkpoint inhibitor;

TNF, tumour necrosis factor.

Figure

adapted from Dougan SK, et al,

2019.

4Smac mimetic compounds promote lymphocyte

expansion and differentiation4

Slide17

Summary

Slide18

SummaryImmune modulation aims to control tumour growth and progression by

targeting immune cells

, and it is therefore different to traditional cancer chemotherapy, which targets cancer cells directly

1ICIs are a form of immune modulation that augment the antitumour immune response by interrupting co-inhibitory signalling pathways that enable cancer cells to evade immune surveillance2ICIs have emerged as first-line treatment options for a range of different cancer types, including metastatic melanoma, non-small cell lung cancer, renal cell carcinoma and bladder/urothelial cancer3

Dual ICI combination treatment has the potential to improve treatment outcomes, and has been validated by the approval of combined anti-CTLA-4/anti-PD-1 regimens in specific cancer types4

The benefits of

combining ICI inhibitors with novel checkpoint-related molecules

and other agents that target cancer growth pathways are being explored

5

1. Naidoo J, et al. Br J Cancer 2014;111(12):2214

9; 2. Darvin P, et al. Exp Mol Med 2018;13;50(12):

165; 3. Alsaab HO, et al. Front Pharmacol 2017;23;8:561; 4. Rotte A, et al. J Exp Clin Cancer Res 2019;38(1):255; 5. Khair DO, et al. Front Immunocol 2019;10:453.

CTLA-4

, cytotoxic T-lymphocyte-associated protein 4

; ICI

, immune checkpoint

inhibitor;

PD-1, programmed cell death protein

1.