Download CD30/CD16A TandAb AFM13-Induced Target Cell Lysis

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Immunocontraception wikipedia , lookup

Antibody wikipedia , lookup

Immune system wikipedia , lookup

Adaptive immune system wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Innate immune system wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Immunomics wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Transcript
CD30/CD16A TandAb AFM13-Induced Target Cell Lysis By NK-Cells
Is Enhanced By CD137 Co-Stimulation And Blocking PD-1
Abstract No
2747
Xing Zhao1, Narendiran Rajasekaran1, Uwe Reusch2, Jens-Peter Marschner2, Martin Treder2#, Holbrook Edwin Kohrt1
for Clinical Sciences Research Stanford, Stanford, CA 94305-5151; 2Affimed GmbH, Im Neuenheimer Feld 582, 69120 Heidelberg, Germany
Establishment of a Hodgkin Lymphoma PDX model
The CD30/CD16A bi-specific tetravalent TandAb antibody AFM13 recruits and activates NK-cells by
specific binding to CD16A for targeted lysis of CD30+ tumor cells1. Given promising clinical activity
and safety profile of AFM13 and proof-of-mechanism demonstrating dependence on the immune
response2, potential synergy of AFM13 and checkpoint inhibitors was evaluated.
In vitro cytotoxicity assays demonstrated higher potency and efficacy of AFM13 relative to other antiCD30 antibody formats (CD30/CD16A diabodies, anti-CD30 IgGs)1. High potency and efficacy was
observed on different CD30+ tumor target cell lines using PBMC or enriched NK-cells as effector cells.
To evaluate a potential synergy of AFM13 and checkpoint inhibitors, chromium release assays with
effector cells that were cultured for 24 hours together with anti-CD30 and irradiated CD30+ lymphoma
tumor cells were performed. Pre-cultured PBMC or enriched NK-cells were used as effector cells at
various E:T ratios in 4 hour chromium release assay with 51Cr-labeled CD30+ lymphoma cells in media
alone, with AFM13, with checkpoint inhibitors, or with AFM13 in combination with checkpoint
inhibitors. Single treatment with AFM13 at suboptimal concentrations (1 pM) induced effector-to-target
cell-dependent lysis of CD30+ lymphoma cells up to 40% using enriched NK cells. Immune-modulating
antibodies alone mediated substantially lower lysis (<25%). However, the addition of anti-PD-1 or antiCD137 to AFM13 strongly enhanced specific lysis up to 70%, whereas the addition of anti-CTLA-4 to
AFM13 showed no beneficial effect. The most impressive increase of efficacy was observed when
AFM13 was applied together with a combination of anti-PD-1 and anti-CD137.
The promising in vitro results of the combination of AFM13 with checkpoint inhibitors and anti-CD137
co-stimulation prompted us to assess potential synergy of AFM13 with checkpoint inhibitors in an in
vivo PDX model.
AFM13 in combination with anti-PD-1 reduces PDX tumor growth
Day 28 + 2
Day 30
700
Ig G
Ig G
A FM 22
Inject AFM13; inject CPI/CPA Ab one day later
(both 5 mg/kg; i.p.)
Rag2-/- IL2Rgnull
Day 28+30
Day 58
700
600
A FM 22
600
a n t i- P D -1
a n t i- P D -1
A F M 2 2 + a n t i-P D -1
500
A F M 2 2 + a n t i-P D -1
500
A FM 13
A FM 13
A F M 1 3 + a n t i-P D -1
400
0
Engraftment of
tumor fragments
(8x8 mm)
Methods
Xenografted tumor fragments (8x8mm) derived from a surgical specimen of a newly diagnosed patient
with CD30+ lymphoma (including Hodgkin Disease), in Rag2-/-IL2Rγnull mice were observed for
engraftment and randomized into groups on day 28. Autologous PBMCs were infused on day 28 (2x106
PBMCs/mouse) intra-peritoneally. Antibody therapy with AFM13 and anti-CTLA-4, anti-CD137, or antiPD-1 began on day 28 and was continued weekly for a total of three intra-peritoneal injections. AntiCTLA-4 (Yervoy), anti-CD137 (Urelumab) or anti-PD-1 (Keytruda) were always dosed one day after
AFM13. An irrelevant IgG and an irrelevant CD16A-recruiting TandAb (AFM22) were use as a control.
At the indicated time points, tumor volumes were determined, and mice were sacrificed for the
assessment of tumor-infiltrating lymphocytes and intra-tumoral cytokines.
Time (days)
28
58
Sacrifice mice with
700% tumor size;
monitor others up to 6
months
Inject autologous
PBMCs
(i.p.)
200
100
0
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
-1 0
4
n = 5 m ic e /P D X s u b je c t
300
R e la t iv e t o B a s e l in e
Tumor growth
300
A F M 1 3 + a n t i-P D -1
400
n = 1 5 m ic e /P D X s u b je c t
P e r c e n t C h a n g e in T u m o r V o lu m e
P e r c e n t C h a n g e in T u m o r V o lu m e
Background
R e la t iv e t o B a s e l in e
1Center
200
100
0
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
-1 0
-2 0
-2 0
-3 0
-3 0
-4 0
-4 0
-5 0
-5 0
• Tumor growth control was investigated in four independent in vivo PDX studies on day 30 and day 58.
• AFM13 reduced tumor growth in all four PDX experiments (1-4) on day 58.
• The inhibitory effect on tumor growth of the anti-PD-1 antibody alone was weaker than that of AFM13 alone, but augmented AFM13 efficacy.
• Therapy with AFM13 and anti-CTLA-4, anti-CD137, or anti-PD-1 began on day 28 and continued
• IgG and an irrelevant CD16A TandAb (AFM22) did not affect tumor growth.
• In 3/4 cases, the combination of AFM13 with anti-PD-1 induced tumor shrinkage.
weekly for a total of three intraperitoneal injections.
• As a control, mice were treated with irrelevant IgG or an irrelevant TandAb (AFM22).
• Tumor volumes, tumor infiltrating lymphocytes and cytokines were assessed at the indicated time
Combination of AFM13 with anti-PD-1 enhances cross-talk between NK- and T-cells
points.
a n ti- C T L A -4
600
16
500
C D 3+C D 4+
C D 3 -C D 5 6 + C D 6 9 +
(d a y 3 0 )
8
4
2
1
0 .5
16
8
0 .2 5
C D 3+C D 4+
C D 3 -C D 5 6 + C D 6 9 +
4
2
1
0 .5
0 .2 5
Ig G
a n t i- P D -1
Day 28+30
C D 3+C D 8+
(d a y 5 8 )
C D 3+C D 8+
32
P o p u la tio n C o m p a r e d to Ig G T r e a tm e n t
A FM 13
700
AF M 2 2
a n t i- P D - 1
AF M 2 2
AF M 1 3
+ a n t i- P D - 1
AF M 1 3
Ig G
AF M 2 2
a n t i- P D - 1
+ a n t i- P D - 1
AF M 2 2
AF M 1 3
+ a n t i- P D - 1
AF M 1 3
+ a n t i- P D - 1
a n t i- C D 1 3 7
400
R e la t iv e t o B a s e l in e
CD30
800
% C h a n g e in T u m o r V o lu m e
CD30/CD16A TandAb structure and mechanism of action
Ig G
P o p u la tio n C o m p a r e d to Ig G T r e a tm e n t
AFM13 combined with CD137 co-stimulation or blocking PD-1 results in PDX
tumor regression
Conclusion
Engagement of NK-cells using CD30/CD16A TandAbs by binding to CD16A does not completely
exploit the efficacy of NK-cells. Combination of AFM13 with immune-modulating anti-CTLA-4, antiCD137, and anti-PD-1 antibodies not only enhanced the anti-tumor activity of NK-cells but also
stimulated infiltration of T-cells and cytokine release in the tumors supporting cross-talk between
innate and adaptive immunity. Therefore combination trials with companion intra-tumoral assessment
may personalize dual antibody therapy and augment the efficacy of AFM13 and CPIs.
Day 28 + 2
32
F o ld C h a n g e in In tr a tu m o r a l L y m p h o c y te
The efficacy of AFM13 was augmented by each CPI (anti-CTLA-4, anti-PD-1, or anti-CD137) tested, but
most impressive and obvious with anti-PD-1. The enhanced antitumor activity of AFM13 in
combination with immune-modulating antibodies was associated with higher numbers of tumorinfiltrating NK- and T-cells and augmented release of pro-inflammatory cytokines. Treatment with
control IgG or irrelevant CD16A-recruiting TandAbs did not induce an unspecific immune cell
activation supporting strict target-dependent NK-cell activation by AFM13.
F o ld C h a n g e in In tr a tu m o r a l L y m p h o c y te
Results
A F M 1 3 + a n ti-C T L A -4
300
A F M 1 3 + a n t i-P D -1
• Treatment with AFM13 alone initially stimulates NK-cell infiltration into tumors, after 58 days infiltration of both NK-cells and T-cells.
200
A F M 1 3 + a n ti-C D 1 3 7
• Combining AFM13 with anti-PD-1 potentiates not only infiltration of NK-cells (CD3-CD56+CD69+), but also migration of CD4+ & CD8+ T-cells
100
0
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
1
2
3
4
into tumors.
• These findings support AFM13-stimulated cross-talk between innate and adaptive immunity that is further augmented by anti-PD-1.
-2 0
• An irrelevant CD16A-recruiting TandAb (AFM22) and/or anti-PD-1 did not substantially enhance the number of infiltrating T- or NK-cells.
-4 0
Intra-tumoral cytokine assessment on day 58
-1 0 0
Stage 1
Stage 2
Stage 3
Stage 4
NK-cell with
CD16A and tumor
cell with CD30
receptors
TandAb locks NKcell and tumor cell
in close proximity
and activates NKcell
NK-cell releases
perforin, creating
pores in tumor cell
membrane for
granzyme entry
Granzyme and
caspase action
trigger apoptosis
of tumor cell
# Corresponding author
• AFM13 reduced tumor growth in all four independent HL PDX studies (1-4).
• The effect of the immune-modulating antibodies on tumor growth was weaker than AFM13 and
strongest for anti-PD-1.
• AFM13 efficacy was augmented by each combination tested, but most impressive in all four
independent studies with anti-PD-1.
• Treatment with AFM13 or anti-PD-1 induces intra-tumoral release of IFN-g, TNF-a
and IL-2.
• The combination therapy with AFM13 and anti-PD-1 increases the release.
• Treatment with irrelevant CD16A TandAb (AFM22) did not induce substantial
intra-tumoral cytokine release
7
IF N - g
c o m p a r e d t o Ig G ( d a y 5 8 )
-8 0
m e a n f o ld c h a n g e in c y t o k in e p r o f il e
-6 0
6
T N F -a
5
IL - 2
4
3
2
1
0
Ig G
AFM 22
a n ti-P D -1
AFM 22
+ a n ti-P D -1
AFM 13
AFM 13
+ a n ti-P D -1
References: 1. Reusch, U. et al. A novel tetravalent bispecific TandAb (CD30/CD16A) efficiently recruits NK cells for the lysis of CD30+ tumor cells. Mabs 6(3): 728–739 (2014).
2. Rothe, A. et al. A phase I study of the bispecific anti-CD30/CD16A antibody construct AFM13 in patients with relapsed or refractory Hodgkin lymphoma. Blood 125, 4024–4032 (2015).