overcoming acquired pd-1/pd-l1 resistance with cd38 blockade€¦ · views 1066 | cancer discovery...

4
VIEWS 1066 | CANCER DISCOVERY SEPTEMBER 2018 www.aacrjournals.org IN THE SPOTLIGHT Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade Deepak Mittal, Dipti Vijayan, and Mark J. Smyth Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. D. Mittal and D. Vijayan contributed equally to this article. Corresponding Author: Mark J. Smyth, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston 4006, Queensland, Australia. Phone: 617-8345-3957; Fax: 617-3362-0111; E-mail: Mark.Smyth@qimrberghofer. edu.au doi: 10.1158/2159-8290.CD-18-0798 ©2018 American Association for Cancer Research. Summary: Overexpression of CD38 after PD-1/PD-L1 blockade increases extracellular adenosine levels and may contribute to acquired resistance to anti–PD-1/PD-L1 therapy. Cancer Discov; 8(9); 1066–8. ©2018 AACR. See related article by Chen et al., p. 1156 (2). Therapies targeting the immune checkpoint molecule pro- grammed death 1 (PD-1) and its ligand, PD-L1, have revolu- tionized cancer treatment in the past decade. Although one third of patients with cancer have benefited from PD-1/ PD-L1 blockade, a large subset of patients remains refrac- tory to these monotherapies (innate resistance). Alarmingly, disease progression is also seen in patients who initially dem- onstrated a response to PD-1/PD-L1 therapy (acquired resist- ance; ref. 1). Understanding mechanisms underlying such innate and acquired resistance to PD-1/PD-L1 blockade and developing novel therapeutic approaches to overcome resist- ance is critical to significantly improve clinical outcomes. In this issue, Chen and colleagues highlight another molec- ular pathway that regulates extracellular adenosine levels and may restrict the long-term benefit of anti–PD-1/PD-L1 (2). This study determined that tumors treated with PD-1/ PD-L1–blocking antibodies developed resistance through the upregulation of CD38, which was induced by all-trans retinoic acid (ATRA) and type I IFN in the tumor microenvironment (Fig. 1). In vitro and in vivo studies demonstrate that CD38 inhibited CD8 + T-cell function via adenosine receptor sig- naling, and that CD38 or adenosine receptor blockade were effective strategies to overcome the resistance in the develop- ment of combination immunotherapeutic strategies (2). An innate anti–PD-1 resistance gene signature called IPRES was identified by transcriptomic analysis of mela- noma biopsies isolated from patients prior to anti–PD-1 treatment. IPRES includes a group of 26 transcriptomic pat- terns associated with regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. It was suggested that inhibition of biological processes relevant to the IPRES signature might enhance the efficacy of PD-1/PD-L1 blockade (3). In contrast, mechanisms of acquired resistance to immunotherapy have been discovered in patients through tumor sequencing and genetic screens, and most evidence so far largely points to tumor alterations that converge on the antigen presentation and IFNγ signaling pathways (4). CD8 + T cell–induced IFNγ initiates an antitumor response through cognate IFNγ recep- tor 1 and 2, and downstream signaling molecules JAK1/2 and IFN regulatory factor 1 (IRF1). Exposure to IFNγ is known to induce PD-L1 expression, and therefore mutations in any of these IFNγ-regulating genes limit the level of PD-L1 expres- sion, making these tumors resistant to anti–PD-1/PD-L1 treatment (4). Recently, the blockade of TNF signaling was shown to enhance anti–PD-1 treatment efficacy through attenuation of PD-1–mediated TIM3 expression and associ- ated activation-induced CD8 + T-cell death (5). A complete analysis of the downstream events following PD-1/PD-L1 inhibition and resistance will (i) help in the iden- tification of novel biomarkers that could predict response to PD-1/PD-L1 therapy and (ii) offer superior combination strategies that will augment efficacy of PD-1/PD-L1 blockade. To this end, Chen and colleagues used mouse models of lung cancer and melanoma to investigate gene signatures that were differentially regulated upon PD-1/PD-L1 blockade. CD38 emerged as a promising candidate whose expression was upregulated manifold in progressively anti–PD-L1-resistant tumors. CD38 is an ectoenzyme expressed by tumor cells and activated immune cells such as T cells, B cells, natural killer (NK) cells, and myeloid-derived suppressor cells (MDSC). CD38 catalyzes metabolically important NAD to ADP-ribose (ADPR) and cyclic ADPR, and this is further dephosphoryl- ated to adenosine in the presence of tumor-localized CD203a and CD73 (6). CD38 expression in the tumor microenviron- ment was not exclusive to anti–PD-1-resistant tumor cells but was also expressed on regulatory T cells and MDSC that had infiltrated the tumor microenvironment. To directly establish the link between PD-L1 and CD38, the authors generated PD-L1 knockout (KO) tumors and sorted these cells on the basis of CD38 expression. Interestingly, PD-L1 KO CD38 tumors failed to grow in wild-type mice compared with PD-L1 KO CD38 hi tumors. Consistently, CD38 knockdown tumors grew significantly slower than CD38 wild-type tumors in wild-type mice. This protection was abolished in the absence of CD8 + T cells, suggesting that CD38-expressing tumor cells impair CD8 + T-cell function. Mechanistically, the authors showed that CD38 promoted adenosine generation presum- ably through the CD38–CD203a–CD73 axis (ref. 7; Fig. 1). Research. on August 9, 2020. © 2018 American Association for Cancer cancerdiscovery.aacrjournals.org Downloaded from

Upload: others

Post on 08-Jul-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade€¦ · VIeWs 1066 | CANCER DISCOVERY September 2018 IN THE SPOTLIGHT Overcoming Acquired PD-1/PD-L1 Resistance with CD38

VIeWs

1066 | CANCER DISCOVERY September 2018 www.aacrjournals.org

IN THE SPOTLIGHT

Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade Deepak Mittal , Dipti Vijayan , and Mark J. Smyth

Immunology in Cancer and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia. D. Mittal and D. Vijayan contributed equally to this article. Corresponding Author: Mark J. Smyth, QIMR Berghofer Medical Research Institute, 300 Herston Road, Herston 4006, Queensland, Australia. Phone: 617-8345-3957; Fax: 617-3362-0111; E-mail: [email protected] doi: 10.1158/2159-8290.CD-18-0798 ©2018 American Association for Cancer Research.

summary: Overexpression of CD38 after PD-1/PD-L1 blockade increases extracellular adenosine levels and may contribute to acquired resistance to anti–PD-1/PD-L1 therapy. Cancer Discov; 8(9); 1066–8. ©2018 AACR.

See related article by Chen et al., p. 1156 (2).

Therapies targeting the immune checkpoint molecule pro-grammed death 1 (PD-1) and its ligand, PD-L1, have revolu-tionized cancer treatment in the past decade. Although one third of patients with cancer have benefi ted from PD-1/PD-L1 blockade, a large subset of patients remains refrac-tory to these monotherapies (innate resistance). Alarmingly, disease progression is also seen in patients who initially dem-onstrated a response to PD-1/PD-L1 therapy (acquired resist-ance; ref. 1 ). Understanding mechanisms underlying such innate and acquired resistance to PD-1/PD-L1 blockade and developing novel therapeutic approaches to overcome resist-ance is critical to signifi cantly improve clinical outcomes.

In this issue, Chen and colleagues highlight another molec-ular pathway that regulates extracellular adenosine levels and may restrict the long-term benefi t of anti–PD-1/PD-L1 ( 2 ). This study determined that tumors treated with PD-1/PD-L1–blocking antibodies developed resistance through the upregulation of CD38, which was induced by all-trans retinoic acid (ATRA) and type I IFN in the tumor microenvironment (Fig.  1). In vitro and in vivo studies demonstrate that CD38 inhibited CD8 + T-cell function via adenosine receptor sig-naling, and that CD38 or adenosine receptor blockade were effective strategies to overcome the resistance in the develop-ment of combination immunotherapeutic strategies ( 2 ).

An innate anti–PD-1 resistance gene signature called IPRES was identifi ed by transcriptomic analysis of mela-noma biopsies isolated from patients prior to anti–PD-1 treatment. IPRES includes a group of 26 transcriptomic pat-terns associated with regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. It was suggested that inhibition of biological processes relevant to the IPRES signature might enhance the effi cacy of PD-1/PD-L1 blockade ( 3 ). In contrast, mechanisms of acquired resistance to immunotherapy have

been discovered in patients through tumor sequencing and genetic screens, and most evidence so far largely points to tumor alterations that converge on the antigen presentation and IFNγ signaling pathways ( 4 ). CD8 + T cell–induced IFNγinitiates an antitumor response through cognate IFNγ recep-tor 1 and 2, and downstream signaling molecules JAK1/2 and IFN regulatory factor 1 (IRF1). Exposure to IFNγ is known to induce PD-L1 expression, and therefore mutations in any of these IFNγ-regulating genes limit the level of PD-L1 expres-sion, making these tumors resistant to anti–PD-1/PD-L1 treatment ( 4 ). Recently, the blockade of TNF signaling was shown to enhance anti–PD-1 treatment effi cacy through attenuation of PD-1–mediated TIM3 expression and associ-ated activation-induced CD8 + T-cell death ( 5 ).

A complete analysis of the downstream events following PD-1/PD-L1 inhibition and resistance will (i) help in the iden-tifi cation of novel biomarkers that could predict response to PD-1/PD-L1 therapy and (ii) offer superior combination strategies that will augment effi cacy of PD-1/PD-L1 blockade. To this end, Chen and colleagues used mouse models of lung cancer and melanoma to investigate gene signatures that were differentially regulated upon PD-1/PD-L1 blockade. CD38 emerged as a promising candidate whose expression was upregulated manifold in progressively anti–PD-L1-resistant tumors. CD38 is an ectoenzyme expressed by tumor cells and activated immune cells such as T cells, B cells, natural killer (NK) cells, and myeloid-derived suppressor cells (MDSC). CD38 catalyzes metabolically important NAD to ADP-ribose (ADPR) and cyclic ADPR, and this is further dephosphoryl-ated to adenosine in the presence of tumor-localized CD203a and CD73 ( 6 ). CD38 expression in the tumor microenviron-ment was not exclusive to anti–PD-1-resistant tumor cells but was also expressed on regulatory T cells and MDSC that had infi ltrated the tumor microenvironment.

To directly establish the link between PD-L1 and CD38, the authors generated PD-L1 knockout (KO) tumors and sorted these cells on the basis of CD38 expression. Interestingly, PD-L1 KO

CD38 − tumors failed to grow in wild-type mice compared with PD-L1 KO CD38 hi tumors. Consistently, CD38 knockdown tumors grew signifi cantly slower than CD38 wild-type tumors in wild-type mice. This protection was abolished in the absence of CD8 + T cells, suggesting that CD38-expressing tumor cells impair CD8 + T-cell function. Mechanistically, the authors showed that CD38 promoted adenosine generation presum-ably through the CD38–CD203a–CD73 axis (ref. 7 ; Fig. 1).

Research. on August 9, 2020. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 2: Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade€¦ · VIeWs 1066 | CANCER DISCOVERY September 2018 IN THE SPOTLIGHT Overcoming Acquired PD-1/PD-L1 Resistance with CD38

views

September 2018 CANCER DISCOVERY | 1067

This subsequently led to the activation of the immunosuppres-sive adenosine receptors A2AR and A2BR in T cells and stunted cytotoxic T-cell functions (Figure 1).

The immunosuppressive effects of adenosine on the tumor microenvironment are well documented (7). This study pro-vides a major role for CD38-mediated adenosine in restricting anti–PD-1/PD-L1 efficacy. Similarly, blocking adenosine gen-eration or signaling via CD73 or A2AR, respectively, increased the sensitivity of tumors to anti–PD-1 therapies (7). Con-versely, adenosine also increases PD-1 levels on CD8+ T cells (7). This further suggests that a vicious cycle exists between PD-1/PD-L1 and adenosine in the tumor microenvironment, justify-ing the need to cotarget PD-1/PD-L1 and adenosine pathways.

Given this striking relationship between PD-L1 and CD38, and CD38 and adenosine, Chen and colleagues next tested whether combining anti–PD-L1 with anti-CD38 antibodies or with adenosine receptor antagonists might improve thera-peutic outcome in mouse cancer models (2). Coinhibition of PD-L1 and CD38 or PD-L1 with the combination of A2A/A2B adenosine receptor antagonists showed superior antitumor efficacy over monotherapy alone. This was also accompanied by marked increase in the percentage of CD8+ effector T cells, and significant reduction in the percentage of CD4+ regulatory T cells and CD45+ MDSC. The authors used the combination of both A2A and A2B adenosine recep-

tor inhibitors in their experiments; therefore, it is difficult to evaluate the relevance of A2A versus A2B receptor expres-sion. Because A2A receptor expression was relatively greater than A2B receptor expression on CD8+ T cells and A2A is a higher-affinity receptor than A2B, it is likely that the major-ity of antitumor effects observed were associated with A2AR on CD8 T cells.

The anti-CD38 antibody daratumumab is an effective treatment for patients with relapsed or refractory multiple myeloma; however, a proportion of patients develop therapy resistance. A detailed investigation into these myeloma cells indicated that CD38 expression was significantly reduced 14 weeks after daratumumab infusion. Exogenous addition of ATRA restored CD38 expression on these bone mar-row–derived myeloma cells, thus making these cells again sensitive to daratumumab therapy (8). An exciting hypothesis would be to test whether reduced CD38 expression following daratumumab affected PD-1/PD-L1 expression in the tumor microenvironment, thus sensitizing myeloma cells to anti–PD-1/PD-L1 treatment.

Besides CD38, the CD39–CD73 axis can also generate adenosine, albeit through a different catalytic pathway (6, 7). How these nonoverlapping and adenosine-generating path-ways are regulated in patients displaying adaptive anti–PD-1/PD-L1 resistance would also need assessment. Importantly,

Anti–PD-1/PD-L1

A Anti–PD-1/PD-L1 therapy

PD-1

CD8+Anti–PD-1/

PD-L1

A Anti–PD-1/PD-L1 therapy

D 1/

PPPDDD--111

CCCCCCCCCDCCCCDDDDD888888888++++++++++

PD-L1

IFNγTumorcelldeath

B Anti–PD-1/PD-L1 mediated acquired resistance

Anti–PD-1/PD-L1

An

PD-1

CD8+

Tumor cell death

PD-PPD-PDD---D----1-1

CD8+

Tumorcells

Tumorcells

PD-1

PD-L1

PD-L1

Anti–PD-1/PD-L1 ATRA

1

IFNβIFNβ

1111

RARα

2

3

CD38

NAD+

ADPR

CD203a

AMPAAAMAMAMAAMAMAMA

CD73

Adenosine

4

Antitumorfunctions

22

Anti–PD-1/PD-L1

C Anti-CD38 therapy in anti–PD-1/PD-L1-resistant tumors

PD-1

CD8+Anti–PD-1/A

PD-L1

C Anti-CD38 therapy in anti–PD-1/PD-L1-resistant tumors

D 1/

PPPDDD-111

CCCCCCCCCDCDCCCDDDDC 8888888888++++++++

PD-L1

Tumorcells

CD38

Anti-CD38

Anti-tumorimmunity

1

2

CD8+

A2AR

A2BR5

Figure 1.   Anti–PD-1/PD-L1–mediated acquired resistance is dependent on CD38-generated adenosine in the tumor microenvironment. A, Blocking PD-1/PD-L1 interactions increases T-cell infiltration in the tumor microenvironment and induces CD8+ T cell–dependent tumor cell control. B, Anti–PD-1/PD-L1–resistant tumors produce soluble mediators such as IFNβ and ATRA (1) that lead to increased expression of CD38 on tumor cells via RARα (2 and 3) that may catalyze NAD+ to immunosuppressive adenosine via the CD38/CD203a/CD73 pathway (4). Adenosine activates A2AR and A2BR adenosine receptors on CD8+ T cells to suppress their antitumor function (5). C, Anti-CD38 mAb in combination with PD-1/PD-L1 blockade improves antitumor immune responses.

Research. on August 9, 2020. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 3: Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade€¦ · VIeWs 1066 | CANCER DISCOVERY September 2018 IN THE SPOTLIGHT Overcoming Acquired PD-1/PD-L1 Resistance with CD38

Views

1068 | CANCER DISCOVERY September 2018 www.aacrjournals.org

CD38 is also a receptor for CD31 or PECAM1, a marker of endothelial cells. High CD31 expression is related to tumor cell migration and may correlate with poor survival in patients with cancer (9). It is hence likely that the antitumor activity observed with antagonistic CD38 antibody might occur through additional nonadenosine pathways such as reduced angiogenesis.

The authors also probed whether these findings could be translated to humans by analyzing tumor specimens from patients with lung cancer and melanoma. CD38 expression was associated with an active immune infiltrate and also coin-cided with the expression of FOXP3, CTLA4, TIM3, PD-1, PD-L2, LAG3, BTLA, CCL2, HVEM, and IDO. Furthermore, CD38 expression was significantly increased in patients on treatment receiving nivolumab (anti–PD-1 therapy) compared with pretreatment groups. This provides further rationale to trial anti-CD38 antibody in patients developing acquired resistance to anti–PD-1 or anti–PD-L1 therapy. Although the anti-CD38 antibody daratumumab is clinically in use for multiple myeloma, the value of blocking CD38 is yet to be tested in patients with solid tumors. Interestingly, intracellu-lar CD38 is also critical for the antitumor effects of NK cells (10). NK cell–tumor cell interaction activates intracellular CD38 in NK cells by protein kinase A, which results in the production of ADPR that targets transient receptor potential melastatin 2 (TRPM2), Ca2+ permeable channels, on cytolytic granules. This TRPM2-mediated Ca2+ is important for NK-cell degranulation and their antimetastatic function (10). This contrasts with Chen and colleagues (2), who show that CD38 on tumor cells produces immunosuppressive adeno-sine. Therefore, in-depth research is required to delineate the role of CD38 on tumor versus immune cells, and whether CD38 blocking will be truly therapeutic in metastatic malig-nancies may need further investigation.

Together, the findings of Chen and colleagues strongly suggest that targeting adenosine may offer therapeutic ben-efit for patients displaying progressively anti–PD-1/PD-L1–resistant tumors. Experimental evidence supports that other adenosinergic molecules such as CD39, CD73, and adenosine receptors may be critical in anti–PD-1/PD-L1 resistance (7). A comparative analysis of these molecules across human tumor

types is warranted, and further mechanism-of-action studies must be performed to determine how redundant these mol-ecules might be with respect to anti–PD-1/PD-L1 resistance.

Disclosure of Potential Conflicts of InterestM.J. Smyth reports receiving commercial research grants from

Bristol-Myers Squibb, Tizona Therapeutics, and Aduro Biotech and is a consultant/advisory board member for Tizona Therapeutics. No potential conflicts of interest were disclosed by the other authors.

Published online September 4, 2018.

RefeRenCes 1. O’Donnell JS, Long GV, Scolyer RA, Teng MW, Smyth MJ. Resistance

to PD1/PDL1 checkpoint inhibition. Cancer Treat Rev 2017;52:71–81. 2. Chen L, Diao L, Yang Y, Yi X, Rodriguez BL, Li Y, et al. CD38-medi-

ated immunosuppression as a mechanism of tumor cell escape from PD-1/PD-L1 blockade. Cancer Discov 2018;8:1156–75.

3. Hugo W, Zaretsky JM, Sun L, Song C, Moreno BH, Hu-Lieskovan S, et al. Genomic and transcriptomic features of response to anti-PD-1 therapy in metastatic melanoma. Cell 2016;165:35–44.

4. Shin DS, Zaretsky JM, Escuin-Ordinas H, Garcia-Diaz A, Hu-Liesko-van S, Kalbasi A, et al. Primary resistance to PD-1 blockade mediated by JAK1/2 mutations. Cancer Discov 2017;7:188–201.

5. Bertrand F, Montfort A, Marcheteau E, Imbert C, Gilhodes J, Filleron T, et  al. TNFalpha blockade overcomes resistance to anti-PD-1 in experimental melanoma. Nat Commun 2017;8:2256.

6. Horenstein AL, Chillemi A, Zaccarello G, Bruzzone S, Quarona V, Zito A, et  al. A CD38/CD203a/CD73 ectoenzymatic pathway inde-pendent of CD39 drives a novel adenosinergic loop in human T lymphocytes. Oncoimmunology 2013;2:e26246.

7. Vijayan D, Young A, Teng MWL, Smyth MJ. Targeting immunosup-pressive adenosine in cancer. Nat Rev Cancer 2017;17:709–24.

8. Nijhof IS, Casneuf T, van Velzen J, van Kessel B, Axel AE, Syed K, et al. CD38 expression and complement inhibitors affect response and resistance to daratumumab therapy in myeloma. Blood 2016;128: 959–70.

9. Deaglio S, Aydin S, Grand MM, Vaisitti T, Bergui L, D’Arena G, et al. CD38/CD31 interactions activate genetic pathways leading to pro-liferation and migration in chronic lymphocytic leukemia cells. Mol Med 2010;16:87–91.

10. Rah SY, Kwak JY, Chung YJ, Kim UH. ADP-ribose/TRPM2-mediated Ca2+ signaling is essential for cytolytic degranulation and antitumor activity of natural killer cells. Sci Rep 2015;5:9482.

Research. on August 9, 2020. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from

Page 4: Overcoming Acquired PD-1/PD-L1 Resistance with CD38 Blockade€¦ · VIeWs 1066 | CANCER DISCOVERY September 2018 IN THE SPOTLIGHT Overcoming Acquired PD-1/PD-L1 Resistance with CD38

2018;8:1066-1068. Cancer Discov   Deepak Mittal, Dipti Vijayan and Mark J. Smyth  BlockadeOvercoming Acquired PD-1/PD-L1 Resistance with CD38

  Updated version

  http://cancerdiscovery.aacrjournals.org/content/8/9/1066

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerdiscovery.aacrjournals.org/content/8/9/1066.full#ref-list-1

This article cites 10 articles, 3 of which you can access for free at:

   

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

  SubscriptionsReprints 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://cancerdiscovery.aacrjournals.org/content/8/9/1066To request permission to re-use all or part of this article, use this link

Research. on August 9, 2020. © 2018 American Association for Cancercancerdiscovery.aacrjournals.org Downloaded from