<<

Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Targeting PD-1 or PD-L1 in Metastatic – Combination Therapy in the First Line Setting

David H. Aggen, Charles G. Drake, and Brian I. Rini

Running Title: Combination PD-1 and PD-L1 for Kidney Cancer

Corresponding Author: David H. Aggen, MD, PhD [email protected] Memorial Sloan Kettering Cancer Center Sidney Kimmel Center for Prostate and Urologic Cancers 353 E. 68th St – Room 421 New York, NY 10065

Charles G. Drake, MD, PhD [email protected] Herbert Irving Cancer Center New York-Presbyterian/Columbia University Medical Center 177 Fort Washington Avenue, 6GN-435 New York, NY 10032

Brian I. Rini, MD [email protected] Vanderbilt University Medical Center Division of Hematology/Oncology 2220 Pierce Ave. 777 Preston Research Building Nashville, TN 37232

Keywords Kidney Cancer PD-1 PD-L1 Tyrosine Kinase Inhibitors Immunotherapy

Declaration of Interest/Disclosure D. Aggen: Consulting fees from Boehringer Ingelheim C. Drake: Consulting fees from BMS, Merck, Genentech/Roche, and B. Rini: Research funding and consulting fees from Merck, Pfizer, Ax, Peloton Therapeutics, BMS, and Genentech/Roche.

1

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Abstract: Recent FDA approvals of regimens targeting programmed death 1 (PD-1) in

combination with anti-CTLA-4 or with vascular endothelial growth factor (VEGF) tyrosine kinase

inhibitors (TKIs) are reshaping front-line therapy for metastatic kidney cancer. In parallel,

therapeutics specific for programmed death ligand 1 (PD-L1), one of the two major ligands for

PD-1, are under continued investigation. Surprisingly, not all PD-1 and PD-L1 agents lead to

similar clinical outcomes, potentially due to biological differences in the cellular expression and

regulation of these targets. Here, we review current clinical data on combination immune

therapy in metastatic kidney cancer and discuss the relevant biology of PD-

1 and PD-L1. The design of future rational combination therapy trials in metastatic renal cell

carcinoma will rely upon an understanding of this biology, along with an evolving understanding

of immune cell populations and their functional states in the tumor microenvironment.

Introduction

The discovery of PD-1 and PD-L1 as a mechanism of peripheral T cell tolerance spurred the development of multiple therapeutics blocking their critical interaction.(1) In the context of kidney cancer, the PD-1 specific therapies (2,3) and (4), as well as the

PD-L1 antibody (5) are FDA-approved in combination with other therapies for

metastatic (RCC). In the past 14 months, 4 phase III trials have tested the

hypothesis that immunotherapy (I/O)-based combinations are efficacious as first-line therapy in

metastatic RCC (Table 1). Combination therapies based on anti-PD-1 antibodies,

2

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

pembrolizumab plus and nivolumab plus , have improved overall (OS)

relative to .(3,4) In contrast, combination therapies with anti-PD-L1 antibodies

including avelumab plus axitinib (Javelin-101)(5) or plus (ImMotion

151)(6), have not yet demonstrated an overall survival benefit. Overall survival data in these

anti-PD-L1 combination trials is still immature, and ultimately a survival benefit might be

observed with further follow-up. Despite this finding, it is notable that occasional complete

responses were reported with both anti-PD-1 and anti-PD-L1 combination regimens. Caution

should be exercised in cross-trial comparison due to potential differences in baseline patient

characteristics. However, the OS benefits documented in anti-PD-1 combination therapy trials,

but not in anti-PD-L1 immunotherapy studies highlights potential advantages to targeting PD-1 as compared to PD-L1 in specific clinical contexts.

Observations in other tumor types support the concept of non-equivalence between PD-

1 and PD-L1 targeted therapeutics. In , the anti-PD-1 agent pembrolizumab

showed an OS benefit compared to 2nd line treatment in Keynote-045(7), whereas the anti-PD-

L1 antibody atezolizumab did not show an OS benefit when compared to 2nd line chemotherapy in a similar patient population in the IMVigor211 trial(8). In NSCLC, the anti-PD-1 antibody pembrolizumab improved OS relative to 2nd line chemotherapy in Keynote-010(9), whereas the anti-PD-L1 antibody avelumab failed to improve OS in a similar cohort of patients(10).

Notwithstanding the limitations of cross-trial comparisons, the discrepancies in clinical outcome

between PD-1 and PD-L1 antibodies beg a lingering question: are these therapeutics

equivalent?

3

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Fundamental differences in the biologic mechanisms of anti-PD-1 and anti-PD-L1 may

underlay these potentially disparate clinical outcomes; thus, understanding these nuances is

critical to the design of next generation combinatorial strategies. Herein, we describe some key

distinctions in PD-1 and PD-L1 biology in terms of cell type specific expression, differential regulation, and the physiologic effects of blockade. The relative contribution of antibody directed cell cytotoxicity (ADCC) for PD-L1 therapeutics is also discussed. Finally, we summarize ongoing clinical activity using these therapeutics in combination regimens.

The PD-1 / PD-L1 Interaction Attenuates a T Cell Response

T cell activation is initiated by the engagement of a T cell receptor (TCR) with its cognate

peptide-MHC complex - along with an appropriate costimulatory signal (Signal 2). In this

setting, the primary biologic function of PD-1 is to maintain a desirable range of T cell activation

so as to prevent rampant autoimmunity.(11) Upon T cell activation, PD-1 is upregulated within

12-36 hours and its interaction with PD-L1 and/or PD-L2 down-modulates T cell proliferation

and effector function (Figure 1).(12) Biochemically, PD-1 binding to either PD-L1 or PD-L2(13)

activates the tyrosine phosphatase SHP-2 in the PD-1 expressing cell; this directly dampens T

cell activation by dephosphorylating the TCR and costimulatory molecules like CD28.(14) In the setting of chronic stimulation, PD-1 preserves T cell clones that might otherwise undergo activation-induced cell death. As a consequence of its biology, PD-1 expression is both

a marker of initial T cell activation as well as a marker of several states of functional exhaustion.

Those states are defined in part by the co-expression of additional immune checkpoint

molecules like LAG-3 and TIM-3.(15) Consequently, not all PD-1 expressing T cells behave as

4

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

functionally exhausted T cells, and additional cell surface markers and epigenetic signatures are

required to more completely define immune cell subsets with diminished effector capacity

(recently reviewed in detail).(16)

PD-1 and PD-L1 Protein Expression: Shared but Distinct Cellular Compartments

In general, PD-1 is expressed on activated / exhausted CD8 and CD4 T cells, although

expression has been reported on a number of other populations including B cells and

macrophages.(17) PD-L1 may be expressed on both tumor cells or other cells in the TME,

including dendritic cells, macrophages and other myeloid populations.(18) Controversy exists

regarding the most important PD-L1 expressing population – with some studies suggesting that

tumor expression is most critical(19,20) and other studies highlighting expression on myeloid

populations.(21,22) The second major PD-1 ligand, PD-L2, has a more restricted expression pattern with predominant expression on endothelial cells, monocytes, and dendritic cells. In a study evaluating PD-L2 expression in 7 distinct tumor types, RCC had amongst the lowest level

of tumor PD-L2 expression with relatively high stromal and endothelial cell expression of this

ligand.(23) The expression of PD-L2 in the TME is in general under-appreciated, especially as

PD-1 has a higher binding affinity for PD-L2 than for PD-L1.(24) Indeed, the potential of PD-L2

to promote T cell tolerance provides one potential explanation for the lack of overall survival

benefit with PD-L1 combination therapies in kidney cancer.

PD-1 and PD-L1 Are Dynamically Regulated by Cell Extrinsic and Intrinsic Factors

5

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

As described above, PD-1 expression is initiated by T cell activation. Expression is

further modulated by a number of signals in the TME including TGF-(25), and IFN-, which

promote upregulation of PD-1 on both T cells and macrophages.(26) PD-1 biology is somewhat

complex, with at least 10 transcriptional factor complexes that function in modulating PD-1

activity dependent on the state of T cell activation (reviewed in detail elsewhere).(27) In acute

infection, antigen clearance leads to eventual down-regulation of PD-1, while in the context of

cancer and chronic viral infection persistent antigen exposure drives continued PD-1 expression

on antigen-specific T cells.(28)

PD-L1 expression on immune and tumor cell subsets is largely induced by TH1 cytokines

like interferon-gamma (IFN-. Following IFN- exposure, tumor and immune cells up-regulate

PD-L1 through a transcriptional program involving the JAK1/STAT signaling pathway.(29)

Clinically, this is important, since mutations in the JAK/STAT pathway and antigen presentation machinery have been implicated in primary and acquired resistance to PD-1 therapy in .(30) At the genomic level, copy number alterations (CNA) in the PD-L1 gene in

tumor cells may also lead to increased levels of PD-L1 expression. CNA in PD-L1 at chromosome

9p24 are associated with increased tumor mutation burden(31) and are enriched in a rare but

unique RCC subset with sarcomatoid pathologic features(32). The latter is of keen interest as

gene signatures associated with sarcomatoid RCC pathology were enriched in patients

responding to atezolizumab and bevacizumab in IMMotion 151.(33) Subgroup analysis of

patients with sarcomatoid pathologic features from Checkmate 214(34) (nivolumab +

ipilimumab) and Keynote 426 (pembrolizumab + axitinib)(35) also demonstrated improved ORR

and OS relative to sunitinib.

6

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Additional tumor intrinsic factors may also drive PD-L1 expression to promote immune tolerance and tumor immune evasion. In clear cell RCC, HIF2 activation secondary to Von

Hippel Landau (VHL) deficiency promotes PD-L1 expression in vitro.(36,37) However, clinical data supporting this association are not yet available. VHL inactivation is estimated to occur in

>90% of RCC patients either through direct mutation or promoter hyper-methylation, and one would anticipate the number of PD-L1 expressing tumor samples in RCC would be dramatically higher if this association were absolute.(38) For example, in Checkmate 214, only 20-30% of

RCC patients had PD-L1 positive tumor cells.(3) Similarly, in the COMPARZ trial evaluating versus sunitinib, 36% of patients had PD-L1 positive specimens.(39) As a consequence, the association between PD-L1 expression and VHL deficiency certainly requires additional investigation.

Metabolic and Epigenetic Programs Modulated by the PD-1 / PD-L1 Axis

PD-1 ligation with PD-L1 or PD-L2 induces T cell functional exhaustion by causing distinct metabolic changes within the T cell. PD-1 binding switches the T cell energy source to fatty acid oxidation with concomitant attenuation of glycolysis.(40) This metabolic switch assists in determination of T cell effector versus memory cell fates and promotes the maintenance of functional CD8 exhaustion. Similarly, attenuation of glycolysis in CD4 T cells, which may or may not be independent of PD-1 signaling, promotes regulatory T cell commitment.(41) Thus PD-1 and additional costimulatory molecules, such as 4-1BB, are implicated in driving immune cell metabolic programs that lead to T cell dysfunction.

7

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Ongoing PD-1 engagement with its cognate ligands also results in epigenetic

reprogramming of T cells, which may prevent effective rescue by immune checkpoint blockade.

These observations were initially based on murine studies utilizing the LCMV virus that mimics

chronic antigen stimulation as is observed in cancerous states.(42) In LCMV murine models,

functionally exhausted T cell remained in a PD-1HI exhausted state even after clearance of

antigen, demonstrating that epigenetic mechanisms likely underlie long-lived functional

exhaustion and PD-1 expression.(43)

More recent data suggest that distinct epigenetic profiles define states of functional T

cell exhaustion.(16) Elegant work identified the nuclear transcription factor TOX as a central

regulator of epigenetic and transcriptional programs driving T cell exhaustion.(44,45) TOX

expression increases following chronic antigen stimulation, leading to a decrease in markers of

self-renewal in T cells - including the key transcription factor TCF1.(46) Conversely, deletion of

TOX restored CD8 T cell function and differentiation to effector and memory phenotypes. Taken

together, these studies show that that TOX is a critical driver of early T cell exhaustion.

Advancements in single-cell analysis(47) and epigenetic profiling will be critical in further

defining the functional and phenotypic heterogeneity within these exhausted states, and

clinical interventions aimed at altering the epigenetic phenotype of T cells remains an area of

active interest.(48-50)

Functional Consequences of PD-1 / PD-L1 Blockade in the Clinic

Anti-PD-1 agents can restore the functionality of exhausted T cells through direct

ligation of PD-1 on CD4 and CD8 T lymphocytes, and based on that principle may rescue an immune response relatively independent of tumor PD-L1 expression (Figure 2A). Direct T cell

8

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

binding by an anti-PD-1 therapeutic may afford significant advantages relative to an anti-PD-L1

treatment, potentially via more rapid T cell expansion. In melanoma patients treated with anti-

PD-1, peripheral blood profiling showed that expansion of a PD-1+ effector T cell pool after

immune checkpoint blockade correlated with clinical response.(51) Relevant neoadjuvant

studies illustrate peripheral occupancy of PD-1, with peripheral blood responses detected

within 3 weeks on therapy.(52) Peripheral blood profiling also shows that anti-PD-1

therapeutics rapidly stimulate T cells in the periphery, enabling tumor cell lysis, relatively

independent of tumor volume/burden. The kinetics of T cell expansion mediated by direct

engagement of PD-1 on effector T cells may not be achievable with anti-PD-L1 agents targeting

tumor and immune cells. With adoption of immunotherapy into the neoadjuvant setting in

clinical trials of kidney cancer including PROSPER-RCC(53), we will gain further insights into the

mechanistic and kinetic differences in PD-1 and PD-L1 occupancy and immune blockade.

Similarly, adjuvant trials for high-risk RCC evaluating atezolizumab (NCT03024996) and pembrolizumab (NCT03142334) have completed accrual, and peripheral blood studies from these trials will enhance our understanding of the relative benefit of peri-operative PD-L1 and

PD-1 blockade.

PD-L1 targeted therapies, in contrast, can induce immune tumor rejection through

multiple mechanisms (Summarized in Figure 2B-2D). First, anti-PD-L1 therapies prevent ligation

with PD-1 on immune cells like anti-PD-1 therapeutics. PD-L1 blockade also prevents ligation

with the costimulatory molecule B7.1 (CD80) either in cis or in trans, which may provide a

secondary mechanism for T cell reinvigoration.(54) Secondly, anti-PD-L1 therapeutics may also

drive direct tumor cell killing through antibody-dependent cellular cytotoxicity (ADCC), in this

9

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

case via PD-L1 expressed on tumor cells (Figure 2C). In murine models, anti-PD-L1 that bind Fc

receptors that mediate ADCC let to tumor regression, whereas a similar effect was not

observed with anti-PD-1 therapies in those models.(55)

Despite these theoretical advantages, there are now 2 randomized phase III trials in

metastatic RCC using anti-PD-L1 therapeutics that have not yet shown an overall survival

benefit (Table 2). Overall survival data in these trials is still immature and longer follow-up is awaited. While the mechanisms underlying this difference may be challenging to dissect, one possibility is that interaction between PD-1 and PD-L2 is unaffected by PD-L1 blockade, such

that interactions between PD-L2 in the TME and PD-1 on T cells provides some level of ongoing

suppression. A 2nd theoretical concern involves binding to non-tumor cell expressing isoforms

of PD-L1, sequestering antibody that might be important in blocking the PD-1 / PD-L1

interaction. Accordingly, relevant data suggest that PD-L1 expression on exosomes(56) and

secreted variants of PD-L1(57) may suppress anti-PD-L1 responses. A final potential mechanism

of interest is that expression of PD-L1 on immune cells might deplete immune effector cells

through ADCC in certain circumstances (Figure 2D, reviewed below).

Antibody Isotype Effects on Clinical Activity

An underappreciated aspect of immune checkpoint blockade is the relative contribution

of T cell mediated tumor killing versus the potential for antibody-dependent cellular

cytotoxicity (ADCC) or complement dependent cytotoxicity. In ADCC, FC gamma receptors

(primarily FcRIII) on the surface of macrophages and NK cells bind to the Fc portion of

antibodies resulting in depletion of tumor or subsets of immune cells (Figure 2C and 2D).

10

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Specific IgG subtypes are more likely to promote ADCC, with IgG1 and IgG3 antibody subtypes with a higher binding affinity for Fc receptors (Supplemental Table 1).(58) Thus, anti-PD-L1 antibodies of the IgG1 isotype may lead to depletion of both tumor and immune cells. Indeed, avelumab, an IgG1 isotype antibody, can mediate ADCC and lead to effective direct tumor cell killing. In theory ADCC can also occur on PD-L1 positive CD8 effector cells leading to elimination of immune effectors. However, no definitive evidence of the latter phenomena has been appreciated clinically. Importantly, recently presented subgroup analysis from the Javelin-101 showed no difference in activity of combination avelumab plus axitinib treatment in patients with Fc-gamma receptor RIII polymorphisms, demonstrating that ADCC may be only a minor mechanism in anti-PD-L1 immunotherapy.(59)

Ongoing efforts are focused on improving the efficiency of antibody-induced cellular cytotoxicity with immune checkpoint blockade antibodies. Through modification of glycosylation and fucosylation sites, antibodies can be engineered to have differential effects on ADCC and cellular depletion.(60) To promote ADCC, an Fc-modified (non-fucosylated version) of anti-CTLA-4 is in early phase clinical trials with the goal of regulatory T cell depletion

(NCT#03110107). Future immunotherapy combination approaches may leverage the ability to selectively deplete immunosuppressive cell subsets and potentiate antitumor responses.

PD-1 vs. PD-L1 in combination with VEGF TKIs

In RCC, the addition of a VEGF TKI to an anti-PD-1 or PD-L1 antibody exploits a number of potentially synergistic mechanisms. VEGF in the tumor ecosystem promotes immunosuppression by decreasing T cell trafficking to tumors, increasing immunosuppressive

11

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

cytokines and initially increasing regulatory T cells. Treatment with anti-angiogenic therapies

mitigates a number of the immunosuppressive effects of VEGF in preclinical models.(61,62) For

example, the use of sunitinib in a preclinical RCC model decreases immunosuppressive myeloid-

derived suppressor cells (MDSCs), a potential mechanism of adaptive immune resistance to PD-

1 immunotherapy.(63) More recent preclinical data with axitinib showed anti-tumor efficacy

not only through vascular remodeling but also through depletion of tumor-promoting mast cells

and tumor-associated macrophages.(64) In human RCC specimens, treatment with antiangiogenic therapy increased infiltration of CD4 and CD8 effector T cells supporting the hypothesis that VEGF inhibition might potentiate the response to immune checkpoint blockade by promoting T cell infiltration.(65) Clearly, the immune effects of VEGF TKIs support non-

redundant mechanisms of immune activation distinct from the PD1/PD-L1 axis, with TKI

immune remodeling affecting the myeloid and T cell compartment.

Although the MDSC-specific effects of TKIs provide a good rationale for combining anti-

PD-L1 with VEGF therapy, there are some data suggesting that VEGF TKIs may dampen a T cell response to cancer. Chronic inhibition of VEGF with TKIs can induce a hypoxic state within the

TME with a concomitant accumulation of HIF-1.(66) Multiple studies show that HIF-1

accumulation induces a compensatory immunosuppressive state through recruitment of

MDSCs(67), tumor-associated macrophages(68,69), and Tregs(70). Additionally, accumulation

of HIF-1 alters PD-L1 expression on immune cell subsets.(71) That observation is supported by

data from human RCC samples showing that TKIs may decrease PD-L1 expression, rendering

anti-PD-L1 blockade more challenging. Of note, on-treatment biopsies from patients treated

12

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

with pazopanib or sunitinib showed transient decreases in PD-L1 expression by

immunohistochemistry.(72)

In some model systems, VEGF TKI’s also decrease immune cell PD-1 expression.(73)

The decrease in PD-1 expression, however, is not absolute, and blockade of remaining PD-1 on

T cells with anti-PD-1 therapeutics may explain the improved OS noted with combination anti-

PD-1 with VEGF TKIs. Taken together, these collected observations lend support to a hypothesis that limited or intermittent VEGF TKI therapy in combination regimens might allow an even greater immune response, but at present all TKI combinations in the phase III setting have been taken continuously. Further, HIF1 inhibition may be a therapeutic approach to enhance the clinical benefit of VEGF TKI-based combinations.

Consensus First-Line Therapy Approaches in Metastatic RCC

Both nivolumab plus ipilimumab and pembrolizumab plus axitinib are now consensus first line treatments for metastatic RCC. At present the choice of first line therapy for a given patient is not driven by a randomized, comparative trial, but rather by treatment side effect profile, prognostic risk group, perceived benefits of complete and overall response rate and

MD/patient preference. Avelumab plus axitinib is also FDA approved for first-line RCC, but so far, an overall survival benefit relative to sunitinib has not been documented. Finally, the FDA

application for drug approval of atezolizumab plus bevacizumab was withdrawn by the manufacturer, although there may in fact be subgroups of patients with specific gene signatures that benefit from this combination.(74) Taken together, these data support the use

13

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

of a PD-1 based immunotherapy combination, either with pembrolizumab plus axitinib or

nivolumab plus ipilimumab for first-line therapy of metastatic RCC.

Future Combination Therapy Approaches for Metastatic RCC

The impressive response rates and overall survival for patients treated with combination

anti-PD-1 plus anti-CTLA-4 or anti-PD-1 plus VEGF TKI therapy with a favorable side effect profile and tolerability begs the question of utilizing a triplet therapy in the first line setting.(75)

Combination nivolumab, ipilimumab, and has been administered safely across GU

malignancies, and the activity of this triplet will be tested in a phase II expansion cohort and a

randomized, phase 3 trial.(76) Triplet therapy, however, likely over treats some patients, such

that biomarker-based strategies to select patients for the appropriate mechanism and intensity

of therapy is an unmet need. One additional combination for first-line treatment,

pembrolizumab + , is currently being tested in large phase III trials. Table 3 provides a

complete listing of trials currently accruing for RCC.

A potential approach to mitigate the toxicities of I/O-I/O combinations is to incorporate

other anti-inflammatory medications into the first-line treatment regimens. To this end, clinical

trials are on-going exploring cytokine targets including anti-IL1NCT04028245), anti-IL-6, and

anti-IL8(77) (NCT03400332) to augment the immune response and potentially improve regimen

tolerability. Another potential approach might be to block TNF- in the combination therapy

setting. A recent publication in animal models highlighted this approach, demonstrating

increased activity of combination immunotherapy when TNF- blockade was added to anti-PD-

1 plus anti-CTLA-4.(78) The wealth of treatment options available for RCC also raises the

14

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

questions of optimal therapeutic sequencing which will be addressed in an upcoming trial.(79)

In contemporary cohorts of metastatic RCC patients, ~50% of patients will not receive a second

line treatment due to either disease progression or declining performance status.(80) In real-

world data sets, it is estimated that >80% of patients do not receive any second line

treatment.(80,81) As a consequence, maximizing the efficacy of first-line therapy is of utmost

importance.

Conclusions

There are now 3 FDA approved combination for the treatment of

first-line kidney cancer, but only anti-PD-1-based combinations to date have illustrated an

overall survival benefit. Blockade of PD-1 permits direct reprogramming of T cells, whereas

anti-PD-L1 exerts those effect in an indirect fashion and permits binding between PD-1 and PD-

L2. Although in contemporary models the activity of anti-PD-1 + VEGF TKI appears to be

additive, the remarkable gains in ORR, PFS and OS will likely necessitate that anti-PD-1 therapeutics remain the backbone of first-line treatment for renal cell carcinoma. For the foreseeable future, the selection of first-line treatment will be guided by side-effect profile, risk group and patient preference, while the next generation of first-line therapies may require clinically validated biomarkers to select the appropriate treatment regimen.

15

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

References:

1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell 2015;27(4):450-61 doi 10.1016/j.ccell.2015.03.001. 2. Motzer RJ, Escudier B, McDermott DF, George S, Hammers HJ, Srinivas S, et al. Nivolumab versus in Advanced Renal-Cell Carcinoma. N Engl J Med 2015;373(19):1803-13 doi 10.1056/NEJMoa1510665. 3. Motzer RJ, Tannir NM, McDermott DF, Aren Frontera O, Melichar B, Choueiri TK, et al. Nivolumab plus Ipilimumab versus Sunitinib in Advanced Renal-Cell Carcinoma. N Engl J Med 2018;378(14):1277-90 doi 10.1056/NEJMoa1712126. 4. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med 2019;380(12):1116-27 doi 10.1056/NEJMoa1816714. 5. Motzer RJ, Penkov K, Haanen J, Rini B, Albiges L, Campbell MT, et al. Avelumab plus Axitinib versus Sunitinib for Advanced Renal-Cell Carcinoma. N Engl J Med 2019;380(12):1103-15 doi 10.1056/NEJMoa1816047. 6. Rini BI, Powles T, Atkins MB, Escudier B, McDermott DF, Suarez C, et al. Atezolizumab plus bevacizumab versus sunitinib in patients with previously untreated metastatic renal cell carcinoma (IMmotion151): a multicentre, open-label, phase 3, randomised controlled trial. Lancet 2019;393(10189):2404-15 doi 10.1016/S0140-6736(19)30723-8. 7. Bellmunt J, de Wit R, Vaughn DJ, Fradet Y, Lee JL, Fong L, et al. Pembrolizumab as Second-Line Therapy for Advanced Urothelial Carcinoma. N Engl J Med 2017;376(11):1015-26 doi 10.1056/NEJMoa1613683. 8. Powles T, Duran I, van der Heijden MS, Loriot Y, Vogelzang NJ, De Giorgi U, et al. Atezolizumab versus chemotherapy in patients with platinum-treated locally advanced or metastatic urothelial carcinoma (IMvigor211): a multicentre, open-label, phase 3 randomised controlled trial. Lancet 2018;391(10122):748-57 doi 10.1016/S0140- 6736(17)33297-X. 9. Herbst RS, Baas P, Kim DW, Felip E, Perez-Gracia JL, Han JY, et al. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet 2016;387(10027):1540-50 doi 10.1016/S0140-6736(15)01281-7. 10. Barlesi F, Vansteenkiste J, Spigel D, Ishii H, Garassino M, de Marinis F, et al. Avelumab versus docetaxel in patients with platinum-treated advanced non-small-cell lung cancer (JAVELIN Lung 200): an open-label, randomised, phase 3 study. The Lancet Oncology 2018;19(11):1468-79 doi 10.1016/S1470-2045(18)30673-9.

16

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

11. Baumeister SH, Freeman GJ, Dranoff G, Sharpe AH. Coinhibitory Pathways in Immunotherapy for Cancer. Annu Rev Immunol 2016;34:539-73 doi 10.1146/annurev- immunol-032414-112049. 12. Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, a novel member of the immunoglobulin gene superfamily, upon programmed cell death. EMBO J 1992;11(11):3887-95. 13. Latchman Y, Wood CR, Chernova T, Chaudhary D, Borde M, Chernova I, et al. PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2001;2(3):261-8 doi 10.1038/85330. 14. Yokosuka T, Takamatsu M, Kobayashi-Imanishi W, Hashimoto-Tane A, Azuma M, Saito T. Programmed cell death 1 forms negative costimulatory microclusters that directly inhibit T cell receptor signaling by recruiting phosphatase SHP2. J Exp Med 2012;209(6):1201-17 doi 10.1084/jem.20112741. 15. Woo SR, Turnis ME, Goldberg MV, Bankoti J, Selby M, Nirschl CJ, et al. Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 2012;72(4):917-27 doi 10.1158/0008-5472.CAN-11- 1620. 16. McLane LM, Abdel-Hakeem MS, Wherry EJ. CD8 T Cell Exhaustion During Chronic Viral Infection and Cancer. Annu Rev Immunol 2019;37:457-95 doi 10.1146/annurev- immunol-041015-055318. 17. Gordon SR, Maute RL, Dulken BW, Hutter G, George BM, McCracken MN, et al. PD-1 expression by tumour-associated macrophages inhibits phagocytosis and tumour immunity. Nature 2017;545(7655):495-9 doi 10.1038/nature22396. 18. Noguchi T, Ward JP, Gubin MM, Arthur CD, Lee SH, Hundal J, et al. Temporally Distinct PD-L1 Expression by Tumor and Host Cells Contributes to Immune Escape. Cancer Immunology Research 2017;5(2):106 doi 10.1158/2326-6066.CIR-16-0391. 19. Lau J, Cheung J, Navarro A, Lianoglou S, Haley B, Totpal K, et al. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice. Nature Communications 2017;8:14572 doi 10.1038/ncomms14572 https://www.nature.com/articles/ncomms14572#supplementary-information. 20. Juneja VR, McGuire KA, Manguso RT, LaFleur MW, Collins N, Haining WN, et al. PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. The Journal of experimental medicine 2017;214(4):895-904 doi 10.1084/jem.20160801. 21. Lin H, Wei S, Hurt EM, Green MD, Zhao L, Vatan L, et al. Host expression of PD-L1 determines efficacy of PD-L1 pathway blockade–mediated tumor regression. The Journal of Clinical Investigation 2018;128(2):805-15 doi 10.1172/JCI96113. 22. Tang H, Liang Y, Anders RA, Taube JM, Qiu X, Mulgaonkar A, et al. PD-L1 on host cells is essential for PD-L1 blockade–mediated tumor regression. The Journal of Clinical Investigation 2018;128(2):580-8 doi 10.1172/JCI96061. 23. Yearley JH, Gibson C, Yu N, Moon C, Murphy E, Juco J, et al. PD-L2 Expression in Human Tumors: Relevance to Anti-PD-1 Therapy in Cancer. Clin Cancer Res 2017;23(12):3158-67 doi 10.1158/1078-0432.CCR-16-1761.

17

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

24. Youngnak P, Kozono Y, Kozono H, Iwai H, Otsuki N, Jin H, et al. Differential binding properties of B7-H1 and B7-DC to programmed death-1. Biochem Biophys Res Commun 2003;307(3):672-7. 25. Park BV, Freeman ZT, Ghasemzadeh A, Chattergoon MA, Rutebemberwa A, Steigner J, et al. TGF-β1-Mediated Smad3 Enhances PD-1 Expression on Antigen-Specific T Cells in Cancer. Cancer Discovery 2016:CD-15-1347 doi 10.1158/2159-8290.CD-15-1347. 26. Terawaki S, Chikuma S, Shibayama S, Hayashi T, Yoshida T, Okazaki T, et al. IFN-alpha directly promotes programmed cell death-1 transcription and limits the duration of T cell-mediated immunity. J Immunol 2011;186(5):2772-9 doi 10.4049/jimmunol.1003208. 27. Bally APR, Austin JW, Boss JM. Genetic and Epigenetic Regulation of PD-1 Expression. The Journal of Immunology 2016;196(6):2431-7 doi 10.4049/jimmunol.1502643. 28. LaFleur MW, Muroyama Y, Drake CG, Sharpe AH. Inhibitors of the PD-1 Pathway in Tumor Therapy. J Immunol 2018;200(2):375-83 doi 10.4049/jimmunol.1701044. 29. Garcia-Diaz A, Shin DS, Moreno BH, Saco J, Escuin-Ordinas H, Rodriguez GA, et al. Interferon Receptor Signaling Pathways Regulating PD-L1 and PD-L2 Expression. Cell Rep 2017;19(6):1189-201 doi 10.1016/j.celrep.2017.04.031. 30. Zaretsky JM, Garcia-Diaz A, Shin DS, Escuin-Ordinas H, Hugo W, Hu-Lieskovan S, et al. Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med 2016;375(9):819-29 doi 10.1056/NEJMoa1604958. 31. Budczies J, Bockmayr M, Denkert C, Klauschen F, Groschel S, Darb-Esfahani S, et al. Pan- cancer analysis of copy number changes in programmed death-ligand 1 (PD-L1, CD274) - associations with gene expression, mutational load, and survival. Genes Chromosomes Cancer 2016;55(8):626-39 doi 10.1002/gcc.22365. 32. Gupta S, Cheville JC, Jungbluth AA, Zhang Y, Zhang L, Chen YB, et al. JAK2/PD-L1/PD-L2 (9p24.1) amplifications in renal cell carcinomas with sarcomatoid transformation: implications for clinical management. Mod Pathol 2019 doi 10.1038/s41379-019-0269-x. 33. Rini BI, Huseni M, Atkins MB, McDermott DF, Powles TB, Escudier B, et al. LBA31Molecular correlates differentiate response to atezolizumab (atezo) + bevacizumab (bev) vs sunitinib (sun): Results from a phase III study (IMmotion151) in untreated metastatic renal cell carcinoma (mRCC). Annals of Oncology 2018;29(suppl_8) doi 10.1093/annonc/mdy424.037. 34. McDermott DF, Choueiri TK, Motzer RJ, Aren OR, George S, Powles T, et al. CheckMate 214 post-hoc analyses of nivolumab plus ipilimumab or sunitinib in IMDC intermediate/poor-risk patients with previously untreated advanced renal cell carcinoma with sarcomatoid features. Journal of Clinical Oncology 2019;37(15_suppl):4513- doi 10.1200/JCO.2019.37.15_suppl.4513. 35. Rini BI, Plimack ER, Stus V, Gafanov R, Hawkins R, Nosov D, et al. Pembrolizumab (pembro) plus axitinib (axi) versus sunitinib as first-line therapy for metastatic renal cell carcinoma (mRCC): Outcomes in the combined IMDC intermediate/poor risk and sarcomatoid subgroups of the phase 3 KEYNOTE-426 study. Journal of Clinical Oncology 2019;37(15_suppl):4500- doi 10.1200/JCO.2019.37.15_suppl.4500. 36. Ruf M, Moch H, Schraml P. PD-L1 expression is regulated by hypoxia inducible factor in clear cell renal cell carcinoma. Int J Cancer 2016;139(2):396-403 doi 10.1002/ijc.30077.

18

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

37. Messai Y, Gad S, Noman MZ, Le Teuff G, Couve S, Janji B, et al. Renal Cell Carcinoma Programmed Death-ligand 1, a New Direct Target of Hypoxia-inducible Factor-2 Alpha, is Regulated by von Hippel-Lindau Gene Mutation Status. Eur Urol 2016;70(4):623-32 doi 10.1016/j.eururo.2015.11.029. 38. Cancer Genome Atlas Research N. Comprehensive molecular characterization of clear cell renal cell carcinoma. Nature 2013;499(7456):43-9 doi 10.1038/nature12222. 39. Choueiri TK, Figueroa DJ, Fay AP, Signoretti S, Liu Y, Gagnon R, et al. Correlation of PD-L1 tumor expression and treatment outcomes in patients with renal cell carcinoma receiving sunitinib or pazopanib: results from COMPARZ, a randomized controlled trial. Clin Cancer Res 2015;21(5):1071-7 doi 10.1158/1078-0432.CCR-14-1993. 40. Patsoukis N, Bardhan K, Chatterjee P, Sari D, Liu B, Bell LN, et al. PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun 2015;6:6692 doi 10.1038/ncomms7692. 41. Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J Immunol 2011;186(6):3299-303 doi 10.4049/jimmunol.1003613. 42. Wherry EJ, Ha SJ, Kaech SM, Haining WN, Sarkar S, Kalia V, et al. Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 2007;27(4):670-84 doi 10.1016/j.immuni.2007.09.006. 43. Pauken KE, Sammons MA, Odorizzi PM, Manne S, Godec J, Khan O, et al. Epigenetic stability of exhausted T cells limits durability of reinvigoration by PD-1 blockade. Science 2016;354(6316):1160-5 doi 10.1126/science.aaf2807. 44. Scott AC, Dündar F, Zumbo P, Chandran SS, Klebanoff CA, Shakiba M, et al. TOX is a critical regulator of tumour-specific T cell differentiation. Nature 2019;571(7764):270-4 doi 10.1038/s41586-019-1324-y. 45. Khan O, Giles JR, McDonald S, Manne S, Ngiow SF, Patel KP, et al. TOX transcriptionally and epigenetically programs CD8+ T cell exhaustion. Nature 2019;571(7764):211-8 doi 10.1038/s41586-019-1325-x. 46. Kratchmarov R, Magun AM, Reiner SL. TCF1 expression marks self-renewing human CD8(+) T cells. Blood Adv 2018;2(14):1685-90 doi 10.1182/bloodadvances.2018016279. 47. Yao C, Sun H-W, Lacey NE, Ji Y, Moseman EA, Shih H-Y, et al. Single-cell RNA-seq reveals TOX as a key regulator of CD8+ T cell persistence in chronic infection. Nature Immunology 2019;20(7):890-901 doi 10.1038/s41590-019-0403-4. 48. Bengsch B, Ohtani T, Khan O, Setty M, Manne S, O'Brien S, et al. Epigenomic-Guided Mass Cytometry Profiling Reveals Disease-Specific Features of Exhausted CD8 T Cells. Immunity 2018;48(5):1029-45 e5 doi 10.1016/j.immuni.2018.04.026. 49. Miller BC, Sen DR, Al Abosy R, Bi K, Virkud YV, LaFleur MW, et al. Subsets of exhausted CD8(+) T cells differentially mediate tumor control and respond to checkpoint blockade. Nat Immunol 2019;20(3):326-36 doi 10.1038/s41590-019-0312-6. 50. Sen DR, Kaminski J, Barnitz RA, Kurachi M, Gerdemann U, Yates KB, et al. The epigenetic landscape of T cell exhaustion. Science 2016;354(6316):1165-9 doi 10.1126/science.aae0491.

19

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

51. Huang AC, Postow MA, Orlowski RJ, Mick R, Bengsch B, Manne S, et al. T-cell invigoration to tumour burden ratio associated with anti-PD-1 response. Nature 2017;545(7652):60-5 doi 10.1038/nature22079. 52. Huang AC, Orlowski RJ, Xu X, Mick R, George SM, Yan PK, et al. A single dose of neoadjuvant PD-1 blockade predicts clinical outcomes in resectable melanoma. Nat Med 2019;25(3):454-61 doi 10.1038/s41591-019-0357-y. 53. Harshman LC, Puligandla M, Haas NB, Allaf M, Drake CG, McDermott DF, et al. A phase III randomized study comparing perioperative nivolumab vs. observation in patients with localized renal cell carcinoma undergoing nephrectomy (PROSPER RCC). Journal of Clinical Oncology 2017;35(15_suppl):TPS4596-TPS doi 10.1200/JCO.2017.35.15_suppl.TPS4596. 54. Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7-1 costimulatory molecule to inhibit T cell responses. Immunity 2007;27(1):111-22 doi 10.1016/j.immuni.2007.05.016. 55. Dahan R, Sega E, Engelhardt J, Selby M, Korman Alan J, Ravetch Jeffrey V. FcγRs Modulate the Anti-tumor Activity of Antibodies Targeting the PD-1/PD-L1 Axis. Cancer Cell 2015;28(3):285-95 doi 10.1016/j.ccell.2015.08.004. 56. Poggio M, Hu T, Pai CC, Chu B, Belair CD, Chang A, et al. Suppression of Exosomal PD-L1 Induces Systemic Anti-tumor Immunity and Memory. Cell 2019;177(2):414-27 e13 doi 10.1016/j.cell.2019.02.016. 57. Gong B, Kiyotani K, Sakata S, Nagano S, Kumehara S, Baba S, et al. Secreted PD-L1 variants mediate resistance to PD-L1 blockade therapy in non-small cell lung cancer. J Exp Med 2019;216(4):982-1000 doi 10.1084/jem.20180870. 58. Vidarsson G, Dekkers G, Rispens T. IgG Subclasses and Allotypes: From Structure to Effector Functions. Frontiers in Immunology 2014;5(520) doi 10.3389/fimmu.2014.00520. 59. Choueiri TK, Albiges L, Haanen JBAG, Larkin JMG, Uemura M, Pal SK, et al. Biomarker analyses from JAVELIN Renal 101: Avelumab + axitinib (A+Ax) versus sunitinib (S) in advanced renal cell carcinoma (aRCC). Journal of Clinical Oncology 2019;37(15_suppl):101- doi 10.1200/JCO.2019.37.15_suppl.101. 60. O’Connell A, AlDeghaither D, Zahavi D, Weiner LM. Enhancing antibody-dependent cell- mediated cytotoxicity: a strategy for improving antibody-based immunotherapy. Antibody Therapeutics 2018;1(1):7-12 doi 10.1093/abt/tby002. 61. Ozao-Choy J, Ma G, Kao J, Wang GX, Meseck M, Sung M, et al. The novel role of tyrosine kinase inhibitor in the reversal of immune suppression and modulation of tumor microenvironment for immune-based cancer therapies. Cancer Res 2009;69(6):2514-22 doi 10.1158/0008-5472.CAN-08-4709. 62. Shrimali RK, Yu Z, Theoret MR, Chinnasamy D, Restifo NP, Rosenberg SA. Antiangiogenic agents can increase lymphocyte infiltration into tumor and enhance the effectiveness of adoptive immunotherapy of cancer. Cancer Res 2010;70(15):6171-80 doi 10.1158/0008- 5472.CAN-10-0153. 63. Ko JS, Zea AH, Rini BI, Ireland JL, Elson P, Cohen P, et al. Sunitinib mediates reversal of myeloid-derived suppressor cell accumulation in renal cell carcinoma patients. Clin Cancer Res 2009;15(6):2148-57 doi 10.1158/1078-0432.CCR-08-1332.

20

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

64. Laubli H, Muller P, D'Amico L, Buchi M, Kashyap AS, Zippelius A. The multi-receptor inhibitor axitinib reverses tumor-induced immunosuppression and potentiates treatment with immune-modulatory antibodies in preclinical murine models. Cancer Immunol Immunother 2018;67(5):815-24 doi 10.1007/s00262-018-2136-x. 65. Liu XD, Hoang A, Zhou L, Kalra S, Yetil A, Sun M, et al. Resistance to Antiangiogenic Therapy Is Associated with an Immunosuppressive Tumor Microenvironment in Metastatic Renal Cell Carcinoma. Cancer Immunol Res 2015;3(9):1017-29 doi 10.1158/2326-6066.CIR-14-0244. 66. Bergers G, Hanahan D. Modes of resistance to anti-angiogenic therapy. Nat Rev Cancer 2008;8(8):592-603 doi 10.1038/nrc2442. 67. Corzo CA, Condamine T, Lu L, Cotter MJ, Youn JI, Cheng P, et al. HIF-1alpha regulates function and differentiation of myeloid-derived suppressor cells in the tumor microenvironment. J Exp Med 2010;207(11):2439-53 doi 10.1084/jem.20100587. 68. Doedens AL, Stockmann C, Rubinstein MP, Liao D, Zhang N, DeNardo DG, et al. Macrophage expression of hypoxia-inducible factor-1 alpha suppresses T-cell function and promotes tumor progression. Cancer Res 2010;70(19):7465-75 doi 10.1158/0008- 5472.CAN-10-1439. 69. Imtiyaz HZ, Williams EP, Hickey MM, Patel SA, Durham AC, Yuan LJ, et al. Hypoxia- inducible factor 2alpha regulates macrophage function in mouse models of acute and tumor inflammation. J Clin Invest 2010;120(8):2699-714 doi 10.1172/JCI39506. 70. Clambey ET, McNamee EN, Westrich JA, Glover LE, Campbell EL, Jedlicka P, et al. Hypoxia-inducible factor-1 alpha-dependent induction of FoxP3 drives regulatory T-cell abundance and function during inflammatory hypoxia of the mucosa. Proc Natl Acad Sci U S A 2012;109(41):E2784-93 doi 10.1073/pnas.1202366109. 71. Noman MZ, Desantis G, Janji B, Hasmim M, Karray S, Dessen P, et al. PD-L1 is a novel direct target of HIF-1alpha, and its blockade under hypoxia enhanced MDSC-mediated T cell activation. J Exp Med 2014;211(5):781-90 doi 10.1084/jem.20131916. 72. Sharpe K, Stewart GD, Mackay A, Van Neste C, Rofe C, Berney D, et al. The effect of VEGF- on biomarker expression in sequential tissue from patients with metastatic clear cell renal cancer. Clin Cancer Res 2013;19(24):6924-34 doi 10.1158/1078-0432.CCR-13-1631. 73. Voron T, Colussi O, Marcheteau E, Pernot S, Nizard M, Pointet AL, et al. VEGF-A modulates expression of inhibitory checkpoints on CD8+ T cells in tumors. J Exp Med 2015;212(2):139-48 doi 10.1084/jem.20140559. 74. McDermott DF, Huseni MA, Atkins MB, Motzer RJ, Rini BI, Escudier B, et al. Clinical activity and molecular correlates of response to atezolizumab alone or in combination with bevacizumab versus sunitinib in renal cell carcinoma. Nat Med 2018;24(6):749-57 doi 10.1038/s41591-018-0053-3. 75. Cella D, Grunwald V, Escudier B, Hammers HJ, George S, Nathan P, et al. Patient- reported outcomes of patients with advanced renal cell carcinoma treated with nivolumab plus ipilimumab versus sunitinib (CheckMate 214): a randomised, phase 3 trial. Lancet Oncol 2019;20(2):297-310 doi 10.1016/S1470-2045(18)30778-2. 76. Nadal RM, Mortazavi A, Stein M, Pal SK, Davarpanah NN, Parnes HL, et al. Results of phase I plus expansion cohorts of cabozantinib (Cabo) plus nivolumab (Nivo) and

21

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

CaboNivo plus ipilimumab (Ipi) in patients (pts) with with metastatic urothelial carcinoma (mUC) and other genitourinary (GU) malignancies. Journal of Clinical Oncology 2018;36(6_suppl):515- doi 10.1200/JCO.2018.36.6_suppl.515. 77. Bermejo IM, Jaffee EM, Davar D, Cardarelli J, Williams D, Phillips P, et al. Phase 1b/2 study of nivolumab in combination with an anti–IL-8 , BMS-986253, in a biomarker-enriched population of patients with advanced cancer. Journal of Clinical Oncology 2018;36(15_suppl):TPS3109-TPS doi 10.1200/JCO.2018.36.15_suppl.TPS3109. 78. Perez-Ruiz E, Minute L, Otano I, Alvarez M, Ochoa MC, Belsue V, et al. Prophylactic TNF blockade uncouples efficacy and toxicity in dual CTLA-4 and PD-1 immunotherapy. Nature 2019;569(7756):428-32 doi 10.1038/s41586-019-1162-y. 79. Zhang T, Ballman KV, Choudhury AD, Chen RC, Watt C, Wen Y, et al. PDIGREE: An adaptive phase 3 trial of PD-inhibitor nivolumab and ipilimumab (IPI-NIVO) with VEGF TKI cabozantinib (CABO) in metastatic untreated renal cell cancer (Alliance A031704). Journal of Clinical Oncology 2019;37(15_suppl):TPS4596-TPS doi 10.1200/JCO.2019.37.15_suppl.TPS4596. 80. Motzer RJ, Barrios CH, Kim TM, Falcon S, Cosgriff T, Harker WG, et al. Phase II randomized trial comparing sequential first-line everolimus and second-line sunitinib versus first-line sunitinib and second-line everolimus in patients with metastatic renal cell carcinoma. J Clin Oncol 2014;32(25):2765-72 doi 10.1200/JCO.2013.54.6911. 81. Wagstaff J, Jones R, Hawkins R, Porfiri E, Pickering L, Bahl A, et al. Treatment patterns and clinical outcomes in patients with renal cell carcinoma in the UK: insights from the RECCORD registry. Ann Oncol 2016;27(1):159-65 doi 10.1093/annonc/mdv504.

22

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure Legends

Figure 1. PD-1 / PD-L1 Targeted Therapeutics in Renal Cell Carcinoma. Overview of current immunotherapy targets in renal cell carcinoma. The PD-1 antibodies pembrolizumab, nivolumab, and prevent interaction with PD-L1 and PD-L2. In contrast, the PD-L1 antibodies avelumab, atezolizumab, and prevent PD1 ligation, but leave PD-1 and

PD-L2 ligation unopposed.

Figure 2. Mechanisms of PD-1 and PD-L1 Targeting Immunotherapy. A) PD-1 blockade exerts direct effects on immune cells upon ligation by driving distinct metabolic and epigenetic programs that reverse T cell dysfunction. B) PD-L1 blockade on immune cells masks PD-1 ligation. PD-1 can bind PD-L2. C) Antibody directed cell cytotoxicity (ADCC) from PD-L1 ligation on tumor cells permits tumor cell killing. D) PD-L1 expression on T cells permits ADCC of immune cells overexpressing PD-L1.

23

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Tables

Table 1. Summary of Completed Phase III Trials in Metastatic RCC Evaluating Combination

Immune Therapies

Nivolumab + Pembrolizumab + Avelumab + Axitinib Atezolizumab + Ipilimumab Axitinib Bevacizumab

Trial CheckMate 214 Keynote 426 Javelin 101 ImMotion 151

N 1096 861 1096 915

PD-L1+ (%)* 23.0% 59.3% 61.0% 40.0%

PD-L1 Assay/Cutoff Dako 28-8 (≥1% TC) Dako 22C-3 (CPS≥1%) Ventana SP263 (≥ 1% IC) Ventana SP-142 (>1% IC)

Favorable 23.0% 31.9% 21.3% 20.0%

Risk Intermediate 61.0% 55.1% 61.3% 69.0% Category Poor 17.0% 13.0% 16.3% 12.0%

Liver Metastases (%) 24.5% 15.0% NR 17.0%

Median Follow-Up (m) 32.4 12.8 12.0 24.0

ORR** 41.0% 59.3% 51.4% 37.0%

CR** 11.0% 5.8% 3.4% 5.0%

PFS (m) Combination Arm 9.7 15.1 13.8 11.2

Sunitinib Arm 9.7 11.1 8.4 8.4

HR (CI) 0.85 0.69 0.69 0.83 (95.0% CI 0.73 – 0.98) (95% CI 0.57 – 0.84) (95.0% CI 0.56 – 0.84) (95.0% CI 0.70 – 0.97)

OS (m) Combination Arm NR NR NR 33.6

Sunitinib Arm 37.9 NR NR 34.9

HR (CI) 0.71 0.53 0.78 0.93 (95.0% CI 0.59 – 0.86) (95% CI 0.38 – 0.74) (95% CI 0.55 – 1.08) (95% CI 0.76 – 1.14)

*Percent PD-L1 positive in combination I/O arm. PD-L1 cutoff and compartment evaluated differs in each trial. **ORR and CR rate in combination I/O arm TC= Tumor cells, IC= Immune Cells, CPS= Combined positive score calculated as the number of (total PD-L1 + TC and IC)/divided by total number of TC x 100 NR= Not reported

24

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

HR with statistically significant confidence intervals are in Bold.

Table 2. Prospects and Limitations of Anti-PD-1 and Anti-PD-L1 Immunotherapies for

Combination Therapy in RCC

Advantages Disadvantages

- Targets T cells directly - Distinct metabolic and epigenetic changes upon PD1 binding reverse T cell exhaustion - No direct tumor effects from antibody Anti-PD-1 - Does not require tumor PD-L1 expression - In vitro VEGF TKIs increase PD-1 expression on immune for activity cells providing a potential resistance mechanism - PD-1 occupancy on T cells observed within 3 weeks on treatment

- Does not block PD-1 binding to PD-L2 - PD-L1 exists on exosomes and in soluble forms which may act as a “decoy” receptor for antibody therapy - VEGF TKI treatment permits PD-L1 upregulation on tumor cells providing a potential resistance mechanism - Targets tumor cells directly - Limited engagement of anti-PD-L1 with PD-L1+ tumor T Anti-PD-L1 - May permit ADCC of tumor cells cells allows for continued T cell dysfunction - May also target immunosuppressive TAMs - Potential for ADCC and elimination of PD-L1+ positive that express PD-L1 in the TME immune cells subsets (NK cells, DCs, anti-tumor TAM) - Kinetics of PD-L1 occupancy are not yet defined and complete saturation to block PD-1 ligation may not be possible

25

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Table 3. On-going Immunotherapy Trials in Renal Cell Carcinoma

Therapy Number Phase Trial ID Estimated Completion Date

First Line Metastatic RCC Trials Pembrolizumab + Lenvatinib or Everolimus + Lenvatinib vs. Sunitinib (CLEAR) 1050 III NCT02811861 February 2021

Nivolumab + Ipilimumab Followed by Nivolumab +/- Cabozantinib (PDIGREE) 1046 III NCT03793166 September 2021

Nivolumab + Ipilimumab +/- Cabozantinib (COSMIC-313) 676 III NCT03937219 November 2021

Nivolumab + Cabozantinb vs. Sunitinib 638 III NCT03141177 May 2024

Nivolumab + Bempegalesleukin (CD122 agonist) vs. Cabozantinib or Sunitinib 600 III NCT03729245 June 2024 Nivolumab + Ipilimumab vs. Nivo/IDO vs. Nivo/Anti-Lag3 () vs. Nivolumab + 200 Ib/II NCT02996110 January 2022 CCR2/CCR5 dual agonist (BMS936558) FRACTION-RCC Nivolumab + Cabozantinib +/- Ipilimumab 152 I NCT02496208 Early 2020

Nivolumab + Ipilimumab or Pazopanib or Sunitinib (BIONIKK Biomarker Guided Trial) 150 II NCT02960906 May 2020

Nivolumab with Salvage Nivolumab + Ipilimumab 120 II NCT03117309 February 2021

Nivolumab + (CD122 agonist) +/- Ipilimumab 90 Ib/II NCT02983045 June 2021

Pembrolizumab + Cabozantinib 55 Ib/II NCT03149822 June 2020

Advanced (2nd Line or later) Metastatic RCC Trials Arginase Inhibitor (INCB001158) + Pembrolizumab 424 Ib/II NCT02903914 January 2020

TLR 7/8 agonist (NKTR 262) + Bempegalesleukin +/- Nivolumab 393 Ib/II NCT03435640 December 2023

Anti-CD73 (CPI-006) +/- A2AR Antagonist or Pembrolizumab 378 Ib/II NCT03454451 December 2023

Glutaminase Inhibitor (CB-839) + Nivolumab 299 Ib/II NCT02771626 Early 2020

Anti-TIM3 (MBG453) +/- Spartalizumab (Anti-PD1) 250 Ib/II NCT02608268 Early 2020

Durvalumab +/- or Savolitinib 195 II NCT02819596 Early 2020

ApoE Agonist (RGX104) + Nivolumab 150 Ib/II NCT02922764 Early 2020

Anti-CSF1R (Cabiralizumab) + Anti-CD40 (APX005M) +/- Nivolumab 120 Ib/II NCT03502330 October 2024

HIF-2a Inhibitor (PT2977) + Cabozantinib 118 II NCT03634540 September 2022

Axitinib + Nivolumab 98 Ib/II NCT03172754 April 2024

Sitravitanib + Nivolumab 60 Ib/II NCT03015740 April 2023

Angiopoietin-2 inhibitor (Trebananib) + Pembrolizumab 60 Ib/II NCT03239145 August 2024

Anti-IL1 () + Cabozantinib 60 Ib NCT03798626 December 2023 Guadecitabine + Durvalumab 58 Ib/II NCT03308396 December 2020

177Lu-J591 Anti-PSMA Radiolabeled Antibody 50 I NCT00967577 December 2019

Anti-CD25 pyrrolobenzodiazepine toxin conjugate () 50 I NCT03621982 July 2021

26

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

IL-2 (Aldesleukin) + Pembrolizumab 27 I NCT03260504 March 2021

Perioperative (Neoadjuvant RCC Trials) Nivolumab – PROSPER RCC 805 III NCT03055013 November 2023

MSKCC 29 Pilot NCT02595918 August 2020

Royal Marsden 19 Pilot NCT02446860 Late 2019

Avelumab + Axitinib 40 Pilot NCT03341845 August 2025 Durvalumab +/- Tremelimumab 45 Ib NCT02762006 January 2020 Nivolumab + Sitravitinib 25 II NCT03680521 December 2019 Anti-IL1 () + Spartalizumab (Anti-PD1) 14 Pilot NCT04028245 2021

Adjuvant RCC Immunotherapy Trials Durvalumab vs. Durvalumab/Tremelimumab vs. Observation 1750 III NCT03288532 December 2037

Pembrolizumab vs. Observation 950 III NCT03142334 December 2025

Nivolumab + Ipilimumab vs. Observation 800 III NCT03138512 July 2023

Atezolizumab vs. Observation 778 III NCT03024996 April 2024

27

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 1:

CD8 T cell

PD-1 Anti-PD-1 TAM PD-L2 Pembrolizumab DC Nivolumab Spartalizumab TCR PD-1

VEGF PepMHC PD-L1 B7.1/2 B7.1/2 Pep Anti-PD-L1 MHC Avelumab CD28 CTLA4 Atezolizumab TCR Durvalumab

Anti-CTLA4 Tumor lpilimumab CD4 cell VEGF Tremelimumab VEGF TKIs Axitinib Cabozantinib Sunitinib Bevacizumab Pazopanib Tavozanib Sitravitanib

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Figure 2: A Prevents PD-L1 interaction B Prevents PD-1 interaction C ADCC on tumor cell D ADCC on immune cell PD-1 Blockade PD-L1 blockade

Direct T-cell engagement Direct tumor engagement Direct tumor engagement Direct T-cell engagement

T cell = Anti-PD-1 T cell = Anti-PD-L1 = Anti-PD-L1 = Anti-PD-L1

TAM TAM PD-1 B7.1 Fc (CD80) PD-1 PD-1 receptor Fc TCR PD-1 PD-L2 PD-L2 TCR receptor pep pep pep MHC TCR PD-L1 DC MHC PD-L1 DC Lytic enzymes MHC Peforin Lytic enzymes PD-L1 Granzymes Peforin TNF Granzymes T cell TNF Tumor Tumor Tumor cell cell cell

• Allows metabolic and • Blocks interaction with PD-1 on • Anti-PD-L1 binds to tumor • Anti-PD-L1 binds to immune epigenetic reprogramming immunocytes and B7.1 (CD80) cells expressing PD-L1 cells expressing PD-L1 of T cells • Does not interact directly with T • Tumor cell killing via Fc-receptor • Potential for immune cell • Blocks interaction between cells mediated antibody-dependent killing and depletion of either PD-1 and PD-L1/PD-L2 cellular cytotoxicity effector T cells or immunosuppressive cells (M2- like TAMs, MDSCs)

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research. Author Manuscript Published OnlineFirst on January 16, 2020; DOI: 10.1158/1078-0432.CCR-19-3323 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Targeting PD-1 or PD-L1 in Metastatic Kidney Cancer - Combination Therapy in the First Line Setting

David H Aggen, Charles G. Drake and Brian I. Rini

Clin Cancer Res Published OnlineFirst January 16, 2020.

Updated version Access the most recent version of this article at: doi:10.1158/1078-0432.CCR-19-3323

Supplementary Access the most recent supplemental material at: Material http://clincancerres.aacrjournals.org/content/suppl/2020/01/16/1078-0432.CCR-19-3323.DC1

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

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

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://clincancerres.aacrjournals.org/content/early/2020/01/16/1078-0432.CCR-19-3323. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from clincancerres.aacrjournals.org on September 26, 2021. © 2020 American Association for Cancer Research.