11
1 Mechanism of action of a new anti-CD38 antibody: enhancing myeloma immunotherapy Fabio Malavasi and Angelo Corso Faini Department of Medical Science University of Torino, Italy Running title: Anti-CD38 antibody in vivo therapy: not only target ligation Corresponding author: Fabio Malavasi, M.D. Department of Medical Sciences University of Torino Medical School Via Santena, 19 10126 TORINO Italy Phone: (+39) 011-860-7552 E-mail: [email protected] Funding: F.M. receives supports from the Fondazione Ricerca Molinette (Torino, Italy). Research. on March 18, 2021. © 2019 American Association for Cancer clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

1

Mechanism of action of a new anti-CD38 antibody: enhancing myeloma immunotherapy

Fabio Malavasi and Angelo Corso Faini

Department of Medical Science

University of Torino, Italy

Running title:

Anti-CD38 antibody in vivo therapy: not only target ligation

Corresponding author:

Fabio Malavasi, M.D.

Department of Medical Sciences

University of Torino Medical School

Via Santena, 19

10126 TORINO

Italy

Phone: (+39) 011-860-7552

E-mail: [email protected]

Funding:

F.M. receives supports from the Fondazione Ricerca Molinette (Torino, Italy).

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 2: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

2

Conflicts of interests:

F.M. has received honoraria for lectures and participation on the advisory boards of Janssen, Tusk

Therapeutics, Takeda and Sanofi, along with research agreements from Janssen and Tusk

Therapeutics.

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 3: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

3

Summary

Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM).

The logic behind it is relatively simple: a target molecule is selected because of its expression on

tumor cells, and the antibody delivers cytotoxic effects. Therapeutic results in MM indicate that

the anti-CD38 antibodies may have relevant immunotherapeutic properties.

Commentary on Moreno et al, this issue

In this issue of Clinical Cancer Research, Moreno and colleagues (1) provide an original analysis of

the characteristics of a new chimeric anti-CD38 antibody candidate for MM therapy.

The initial promise of monoclonal antibodies (mAb), heralded as a “magic bullet” for cancer

therapy, was not immediately fulfilled. Decades of frustrating results were necessary before the

clinical success of anti-CD20 antibodies finally rewarded the efforts of clinicians and

pharmaceutical companies, rekindling interest in pursuing the development of antibody-based

therapies. This required rethinking the role of antibodies in vivo: it is now accepted that some

antibodies block the function of a membrane receptor while others synergize it. Synergistic

antibodies occupy a domain that hosts the site of the natural ligand of the molecule (molecular

mimicry). When the target lies in close proximity to professional receptors it can trigger signals

(molecular parasitism). CD38, functionally associated with BCR, TCR and CD16A (the low-affinity

IgG FcR) (2), is one example.

These conclusions are relatively straightforward when the function of the target molecule is

known. However, such is not the case for CD38, whose precise roles are not yet fully known. The

results of antibody therapy following the introduction of Daratumumab (3) have provided useful

practical and theoretical information about the functions of the target and - at the same time -

revealed some unexpected mechanisms of action (MoAs) of therapeutic antibodies in vivo. The

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 4: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

4

findings presented by Moreno et al. indicate that Isatuximab has unique characteristics, only some

of which are shared with Daratumumab. In fact, given their different MoAs, the two antibodies

might be potentially valuable as therapeutic complements or alternatives in patients developing a

resistance to one of them. The particular benefit provided by Isatuximab is its sensitivity to the

number of CD38 molecules present on target cells. Isatuximab saturates membrane CD38 and can

be internalized. However, antibody-dependent cell cytotoxicity (ADCC), antibody-dependent cell

phagocytosis (ADCP) and complement-dependent cytoxicity (CDC) are triggered only when the

number of surface CD38 molecules reaches a threshold. The same thing happens in the induction

of direct apoptosis. Isatuximab does not alter the transcription on a high CD38+ myeloma line.

The existence of such a threshold was established by quantifying the number of CD38 molecules

expressed both by MM and by the majority of normal cell populations. Isatuximab may have a

lower depleting power than Daratumumab, but this feature could be protective for normal CD38+

cell populations. The action of Isatuximab is more complex when studying its effects on NK cells,

the major effectors of cytotoxicity. Moreno et al. tracked the events taking place in culture after

adding Isatuximab to MM in the presence of PBMC containing NK cells. The results are similar to

those reported with Daratumumab, with the number of circulating NK cells declining after

antibody infusion. Moreno et al. show that NK cells rapidly decrease in number after their initial

activation by Isatuximab. The authors ruled out the possibility that this decrease in NK cells [CD38+

(at a density lower than MM ) and CD16A+] is due to fratricide cytotoxicity (4). Isatuximab depletes

NK cells both in blood and bone marrow (BM) along with B progenitors and basophils. No effects

were observed in Treg populations. The experiments also highlighted the existence of cross-talk

between NK cells and Isatuximab, followed by their activation when exposed to MM. This cross-

talk includes Isatuximab’s exploitation of transmembrane signaling, which involves its Fc domain.

Indeed, these effects are diminished in the presence of Fc blockers. The same signals were

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 5: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

5

investigated at a molecular level by examining the transcription of NK cells after exposure to a

myeloma line in the presence of Isatuximab. The 70 genes identified are classified as being

involved in chemotaxis, cytolysis and defense response. One of them, CD137 (Tumor Necrosis

Factor Receptor Super Family 9, TNFRSF9), is a gene that controls an inducible co-stimulatory

molecule and also a player in anti-CD20 therapy. The combination of Isatuximab with anti-CD137

did not provide the expected prolongation of life of NK cells in this MM model. Similar negative

results were obtained by combining Isatuximab with lenalidomide and proteasome inhibitors. Co-

cultures with SLAMF7 (a target of an antibody used in MM therapy and reported as being involved

in ADCP) likewise proved not to alter the depletion of NK cells.

CD38 is a phylogenetically ancient molecule whose functions as an ectoenzyme, an adhesion

molecule and a receptor are well known (2). The work by Moreno et al. significantly expands

investigation of the actual role of CD38. It is still difficult to reconcile the use of CD38 as a tumor

target given its almost ubiquitous presence on the surface of normal cells, including B, T, and

myeloid regulatory populations. Other cells (e.g., erythrocytes and platelets) also express CD38,

although at very low densities. It is also unclear how the same antibody can deliver toxic hits to

the tumor, while also driving different effects on positive and negative effectors. Differences in the

surface levels of the molecule, very high on MM and low on effectors, may account for its distinct

functional effects. The interactions taking place between the target epitope of the therapeutic

antibodies and the different IgG Fc Receptors (FcR) give raise to alternative hypotheses. The

epitope recognized by Isatuximab encompasses the catalytic domain of the molecule and is

different from that of Daratumumab (5). The main CD38 substrate is ATP, with the production of

cADPR and ADPR, cytoplasmic messengers regulating Ca2+. In selected environments such as the

myeloma niche in BM, CD38 may also use NAD+, especially in acidic conditions. pH modification is

one of the evasion strategies implemented by MM and induces the expression of CD203a, an

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 6: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

6

ectonucleotide pyrophosphatase/phosphodiesterase 1 (NPP1), also known as Plasma Cell-1 (PC-1).

This ectoenzyme cooperates with CD38 to produce adenosine, a potent immune suppressant.

Isatuximab is reportedly one of the most efficient inhibitors of the enzymatic features exerted by

CD38, only now being evaluated for its therapeutic potential.

While the role of the IgG FcRs has been analyzed in different tumor models and with different

antibodies (6), there has been no systematic analysis of its role in anti-CD38 therapy to date. The

structural differences between Isatuximab (chimeric) and Daratumumab (human) may account for

distinct functional interactions with the IgG FcRs. Experience with individual FcRs shows that the

membrane dynamics of myeloma cells change when Daratumumab is presented by antibody-

armed FcR+ effectors, mimicking the events taking place in vivo (7). The outcome of the

interactions of the two different antibodies may explain the differences observed in terms of

membrane dynamics, with Isatuximab leading to internalization and Daratumumab to generation

of membrane-derived vesicles.

It seems intuitively difficult to rationalize a scenario in which the antibody kills the tumor, depletes

cells populations, activates T effectors and blocks suppressors. A hypothesis that may help in

explaining it is that the distinct signals may result from the ligation of the target molecule by the

antigen-binding site and from the simultaneous engagement of the IgG Fc domain by FcRs (figure

1). The formation of a trimolecular complex (the “scorpion effect”) may lead to activatory or

inhibitory signals, according to the target cells (8).

The fate of the therapeutic antibody may also be an issue. The life of Daratumumab in vivo is

longer than that of normal IgG. An attractive hypothesis is that the low-CD38 density erythrocytes

and platelets act as a carrier of the therapeutic antibodies in the biological fluids, protecting them

from elimination. Recent studies on neonatal FcR, the physiological regulator of homeostasis of

IgG and albumin (9), may offer some related insights.

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 7: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

7

The contribution by Moreno et al. is noteworthy in that it links the threshold of in vivo levels of

CD38 to the predicted efficacy of antibody treatment in MM. The peculiar ability of Isatuximab to

react only with cells expressing high levels of CD38 may lead to the definition of signals in NK cells

capable of sustaining their activation and duration in vivo. The relationship between a given

target for a therapeutic antibody and the FcR co-expressed on the target cell raises the possibility

that the “scorpion effect” might be exploited when engineering the Fc domain to enhance the

therapeutic effects.

No less importantly, the clinical benefits of antibody-mediated therapy in MM are paralleled by its

wide acceptance by patients, another powerful argument for seeking further advances in the field.

Acknowledgements

F.M. is greatly indebted to Professor M. Cragg (University of Southampton, UK) for suggestions

and to Laura McLean and Marzia Roccia for editorial assistance in the preparation of the

manuscript.

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 8: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

8

REFERENCES

1) Moreno et al, Clinical Cancer Research, this issue

2) Malavasi F, Deaglio S, Funaro A, Ferrero E, Horenstein AL, Ortolan E, et al Evolution and function of the ADP ribosyl cyclase/CD38 gene family in physiology and pathology. Physiol Rev. 2008 Jul;88(3):841-86 3) van de Donk NWCJ, Richardson PG, Malavasi F. B CD38 antibodies in multiple myeloma: back to the future. Blood. 2018 Jan 4;131(1):13-29 4) Wang Y, Zhang Y, Hughes T, Zhang J, Caligiuri MA, Benson DM et al. Fratricide of NK Cells in Daratumumab Therapy for Multiple Myeloma Overcome by Ex Vivo-Expanded Autologous NK Cells. Clin Cancer Res. 2018 Aug 15;24(16):4006-4017 5) Deckert J, Wetzel MC, Bartle LM, Skaletskaya A, Goldmacher VS, Vallée F et al. SAR650984, a novel humanized CD38-targeting antibody, demonstrates potent antitumor activity in models of multiple myeloma and other CD38+ hematologic malignancies. Clin Cancer Res. 2014 Sep 1;20(17):4574-83 6) Roghanian A, Teige I, Mårtensson L, Cox KL, Kovacek M, Ljungars A, et al. Antagonistic human FcγRIIB (CD32B) antibodies have anti-tumor activity and overcome resistance to antibody therapy in vivo. Cancer Cell. 2015 Apr 13;27(4):473-88 7) Malavasi F, Castella B, Schiavoni I, Incarnato D, Oliva S, Horenstein AL CD38 and antibody therapy: what can basic science add? [Abstract]. Blood 2016; 128:SCI-36 8) Hogarth PM, Pietersz GA. Fc receptor-targeted therapies for the treatment of inflammation, cancer and beyond. Nat Rev Drug Discov. 2012 Mar 30;11(4):311-31 9) Pyzik M, Rath T, Lencer WI, Baker K, Blumberg RS. FcRn: The Architect Behind the Immune and Nonimmune Functions of IgG and Albumin. J Immunol. 2015 May 15;194(10):4595-603

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 9: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

9

Figure 1. The left side of the figure illustrates the key effects of the anti-CD38 Isatuximab MoA on

the tumor target and on the main functional effectors (NK cells). On the right, a diagram of a

hypothetical extension of the MoA by the major therapeutic antibodies specific for target

molecules of lymphoid neoplasia. The hypothesis rests mainly on the functional network

implemented between the antibodies and their IgG Fc Receptors (FcR) expressed at various levels

by lymphoid and myeloid effectors. Included is the possibility that the antibodies may react

simultaneously on the same cell via Fab and via FcR, through the so-called scorpion effect. Most

steps in the diagram have been experimentally validated and others await verification.

Abbreviations: ITAM, immunoreceptor tyrosine-based activation motif; ITIM, immunoreceptor

tyrosine-based inhibitory motif

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 10: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

Figure 1:

© 2019 American Association for Cancer Research

Isatuximab MoA Hypothesis for an extension of MoA

FcR+

e�ectorFcR+

e�ector

ITAM ITAM

ITIM ITIM

FcR FcRCD38 CD38

Scorpion e�ect

CD38

E�ects onFcR- target

Myeloma

CD38

Signals

ADCCCDCet al.

or or

- Quantitative levels of antibody

Isatuximab

IsatuximabIsatuximab

Isatuximab

Isatuximab

Isatuximab

Deprivation of B-cell precursorsand basophils

ADCC (on high and low CD38 levels)CDC (on high CD38 levels)ADCP (on high CD38 levels)Direct apoptosis (on high CD38 levels)Sensitization of high CD38 cells tobortezomib+dexamethasone in thepresence of stromaTranscription unaltered

Events followed by exhaustion andphagocytosis

Ex vivo depletion of CD38+ cells, witha Fc-dependent activation (andmodification of transcription)

Myeloma

NK cells

Bone marrow

- Activation/di�erentiation and number of target molecules

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260

Page 11: Mechanism of action of a new anti-CD38 antibody: enhancing ... · 07-03-2019  · Antibody therapy is a treatment option for several diseases, including multiple myeloma (MM). The

Published OnlineFirst March 7, 2019.Clin Cancer Res   Fabio Malavasi and Angelo Corso Faini  myeloma immunotherapyMechanism of action of a new anti-CD38 antibody: enhancing

  Updated version

  10.1158/1078-0432.CCR-19-0260doi:

Access the most recent version of this article at:

  Manuscript

Authorbeen edited. Author manuscripts have been peer reviewed and accepted for publication but have not yet

   

   

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected] at

To order reprints of this article or to subscribe to the journal, contact the AACR Publications

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://clincancerres.aacrjournals.org/content/early/2019/03/07/1078-0432.CCR-19-0260To request permission to re-use all or part of this article, use this link

Research. on March 18, 2021. © 2019 American Association for Cancerclincancerres.aacrjournals.org Downloaded from

Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on March 7, 2019; DOI: 10.1158/1078-0432.CCR-19-0260