You are on page 1of 13

EUROPEAN UROLOGY 68 (2015) 267–279

available at www.sciencedirect.com
journal homepage: www.europeanurology.com

Platinum Priority – Review – Bladder Cancer


Editorial by Tom Powles on pp. 280–282 of this issue

A Systematic Review of Immunotherapy in Urologic Cancer:


Evolving Roles for Targeting of CTLA-4, PD-1/PD-L1, and HLA-G

Edgardo D. Carosella a,b,*, Guillaume Ploussard c, Joel LeMaoult a,b, Francois Desgrandchamps a,b,c
a
CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, Paris,
b c
France; University Paris Diderot, Sorbonne Paris Cité, UMR E_5 Institut Universitaire d’Hematologie, Saint-Louis Hospital, Paris, France; Urology
Department, Saint-Louis Hospital, Paris, France

Article info Abstract

Article history: Context: Overexpression of immune checkpoint molecules affects tumor-specific T-cell
Accepted February 25, 2015 immunity in the cancer microenvironment, and can reshape tumor progression and
metastasis. Antibodies targeting checkpoints could restore antitumor immunity by
blocking the inhibitory receptor-ligand interaction.
Keywords: Objective: To analyze data and current trends in immune checkpoint targeting therapy
Cancer immunotherapy for urologic cancers.
Monoclonal antibodies Evidence acquisition: Systematic literature search for clinical trials in the PubMed and
Cochrane databases up to August 2014 according to Preferred Reporting Items for
Checkpoint
Systematic Reviews and Meta-analyses guidelines. Endpoints included oncologic
CTLA-4 results, tumor response rates, safety, and tolerability.
PD-1 Evidence synthesis: Anti-CTLA-4 monotherapy has demonstrated biochemical responses
HLA-G in prostate cancer. One phase 3 trial assessing ipilimumab efficacy in castration-resistant
disease was negative overall. Nevertheless, ipilimumab may significantly improve overall
survival compared with placebo in subgroups of patients with favorable prognostic
features. In renal cancer, phase 1 trials showed interesting stabilization or long-lasting
objective response rates approaching 50% using anti-PD-1/PD-L1 drugs in heavily pre-
treated metastatic patients. In bladder cancer, one phase 2 trial indicated a good safety
profile for ipilimumab as a neoadjuvant drug before radical cystectomy. Overall, immune-
Please visit related effects such as colitis and dermatitis were common and well tolerated.
www.eu-acme.org/ Conclusions: Our systematic review shows that antibodies blocking immune check-
points offer interesting and long-lasting response rates in heavily pretreated patients
europeanurology to read and
with advanced urologic cancers. More promising results are currently provided by anti-
answer questions on-line. CTLA-4 antibodies in prostate cancer and by PD-1/PD-L1 inhibitors in renal cancer. These
The EU-ACME credits will should encourage new clinical trials of immune therapy combinations and immuno-
then be attributed therapy monotherapy combined with conventional anticancer drugs. In bladder cancer,
the use of targeted immunotherapy still remains underevaluated; however, preliminary
automatically.
results reported at recent conferences seem encouraging.
Patient summary: Data from studies support the activity and safety of immune check-
point inhibitors in urologic cancers, alone or in combination with conventional cancer
therapies. Encouraging data in other oncologic fields could translate into interesting
responses in urological cancers.
# 2015 European Association of Urology. Published by Elsevier B.V. All rights reserved.

* Corresponding author. CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI),
Research Division in Hematology and Immunology (SRHI), Saint-Louis Hospital, 1 Avenue Claude
Vellefaux, 75475 Paris Cedex 10, France. Tel. +33 1 57276778; Fax: +33 1 47276780.
E-mail address: edgardo.carosella@cea.fr (E.D. Carosella).
http://dx.doi.org/10.1016/j.eururo.2015.02.032
0302-2838/# 2015 European Association of Urology. Published by Elsevier B.V. All rights reserved.
268 EUROPEAN UROLOGY 68 (2015) 267–279

1. Introduction association for immune checkpoints and/or its functional


consequences were developed. In animal models, such
To survive and proliferate, tumors adopt active immune antibodies were successful at restoring antitumor immuni-
escape strategies that protect them from antitumor ty when tumor escape depended on the particular immune
immunity. To restore antitumor immune efficiency, im- checkpoint considered. Thus, immunotherapeutic clinical
mune escape mechanisms must be bypassed, overridden, or trials were initiated, some of which are currently ongoing.
cancelled. Current cancer immunotherapy strategies mostly To date, antibodies blocking immune checkpoints have
aim at restoring T-cell–mediated antitumor immunity. T- exhibited oncologic efficacy, including prolongation of
cell–mediated immunity includes multiple sequential overall survival (OS), in various malignancies, mainly breast
steps: clonal selection of antigen-specific cells, activation, cancer and melanomas. Here we review immune check-
proliferation, trafficking, and execution of direct effector points related to urologic cancers with a focus on CTLA-4/B7,
function. Each step is regulated by a balance between PD-1/PD-L1, and HLA-G/ILT-2/4 because of their specific
stimulatory and inhibitory signals. Immune checkpoints are relevance to immunotherapy and clinical trials to modulate
the inhibitory pathways that physiologically counterbal- them have been conducted.
ance the co-stimulatory pathways to fine-tune the immune
responses. In other words, immune checkpoints are normal 2. Evidence acquisition
immune signals capable of stopping an immune response.
They involve inhibitory receptors and their ligands (Fig. 1 A systematic literature search in the PubMed and Cochrane
and Fig. 2): one is expressed by a putative target cell and the databases was performed to identify clinical and random-
other is expressed by effector cells, in particular T cells. ized controlled trials (RCTs) published up to August
Under normal physiological conditions, immune check- 2014. Various algorithms including the following terms
point pathways are crucial for self-tolerance maintenance, were used: bladder cancer, prostate cancer, renal cancer,
modulation of the duration and strength of normal immunotherapy, CTLA-4, PD-1, PD-L1, MHC-II, B7-H1, B7-H3,
physiological immune responses, and minimization of B7-H4, TIM3, HLA-G, and each molecule under development
collateral damage to healthy tissues. Thus, overexpression (Table 1). Given that targeted immunotherapy is a fast-
of immune checkpoint molecules by tumor cells profoundly moving field, we also searched abstracts from the major
affects tumor-specific T-cell immunity in the cancer oncology conferences during 2012–2014. Inclusion criteria
microenvironment. This effectively marks tumor cells as used were published full articles, clinical trials, retrospec-
not for elimination, and can therefore reshape tumor tive series, meta-analyses, and English language. Our
progression and metastasis. Since most tumor immune systematic review focused on published clinical trials
escape mechanisms that use immune checkpoints block according to Preferred Reporting Items for Systematic
effector cell functions, antitumor immunity may be restored Reviews and Meta-analyses (PRISMA) guidelines. Primary
by antibodies that block the inhibitory receptor-ligand outcomes included oncologic results and tumor response
interaction and thus inactivate the immune checkpoints. rates. Secondary outcomes included safety and tolerability.
On the basis of these immunology data, monoclonal The flow diagram for article selection is shown in
antibodies capable of disrupting the ligand-receptor Figure 3. Each identified article was analyzed and classified.

[(Fig._1)TD$IG]
T cell ac!va!on
Prolifera!on
IFNG secre!on
Res!ng Cytotoxic func!on Ac!vated
T cell T cell

PD1

+ + BTLA

TCR CD28
TCR CD28 TIM-3
B7-H1/PDL1
LILRB1/ILT2

CTLA4/CD152
LAG-3/CD223

LILRB1/ILT2

APC/tumor cell MHC-I / II B7-1, B7-2

Fig. 1 – Upregulation of immune checkpoint receptors on T cell activation. With the exception of ILT2, most immune checkpoint receptors are absent
from the resting T-cell surface. T cell activation is initiated by combined signals from TCR:MHC and BD28:B7 engagement. This leads to proliferation,
upregulation of proinflammatory cytokine expression, acquisition of effector cell function, which is cytotoxicity in the case of CD8+ cells, and
provision of help for CD4+ T cells. Activated T cells are effector T cells. They express immune checkpoint receptors, and have therefore become
sensitive to inhibition by ligands expressed by tumor cells or within the microenvironment. APC = antigen-presenting cell; IFNG = interferon-
gamma.
EUROPEAN UROLOGY 68 (2015) 267–279 269

Fig. 2 – Schematic representation of possible engagement of checkpoint proteins leading to T cell inhibition. On engagement of immune checkpoint
protein receptors (expressed by activated T cells) by immune checkpoint ligands (expressed by APC or tumor cells), the activated T-cell functions
(proliferation, IFNG secretion, and cytotoxic function shown here) are inhibited. Furthermore, immune checkpoint signaling may induce T cell anergy
and the differentiation of regulatory T cells whose functions contribute to further inhibition of antitumor immunity. APC = antigen-presenting cell;
IFNG = interferon-gamma.

3. Evidence synthesis occur via its association with the tyrosine phosphatases SHP1,
SHP2, and PP2A.
3.1. Antitumor activity CTLA-4 is expressed by activated CD8+ effector T cells.
However, its major physiological role seems to be through
3.1.1. CTLA-4 distinct effects on CD4+ T cells, including downregulation of
Cytotoxic T-lymphocyte–associated antigen 4 (CTLA-4; also helper T (TH) cell activity and enhancement of regulatory
known as CD152) is expressed exclusively on T cells. CTLA-4 T cell (TReg) immunosuppressive activity [4,5]. CTLA-4
primarily regulates the amplitude of early-stage T-cell blockade results in broad enhancement of the immune
activation. One of its mechanisms of action involves responses that depend on TH cells.
antagonism of B7-CD28–mediated co-stimulatory signals,
which occurs because CTLA-4 has a much higher affinity 3.1.1.1. Prostate cancer. Preclinical studies have supported the
for B7 than CD28 does: binding of CTLA-4 to CD80/86 is potential role of an anti–CTLA-4 antibody (ipilimumab) as
500–2500 times greater than that of CD28. Signaling an antitumor agent in prostate cancer [6–8]. However, only
through CD28 promotes mRNA expression of the cytokine six phase 1/2 trials and one phase 3 trial have been
IL-2 and entry into the cell cycle, T-cell survival, T-helper-cell published to date. Outcome data are listed in Table 2.
differentiation, and immunoglobulin isotype switching. Thus, In phase 1/2 trials, anti–CTLA-4 blockade monotherapy
signaling through CTLA-4 inhibits IL-2 mRNA production led to biochemical responses (prostate-specific antigen
and inhibits cell cycle progression. This mechanism only decline >50%) in approximately 15% of cases [9,10]. A
depends on the extracellular domain of CTLA-4 [1,2]. A second combination of ipilimumab with granulocyte-macrophage
mechanism by which CTLA-4 can inactivate T cells involves colony-stimulating factor or vaccines seemed to improve the
delivery of a negative signal. This mechanism requires the biochemical tumor response rate by up to 25–50% [11–15].
cytoplasmic tail of CTLA-4 and occurs at low levels of surface Use of a vaccine that enhances co-stimulation of the immune
expression [3]. In most cases, the inhibitory actions of CTLA-4 system did not seem to exacerbate the immune-related
270 EUROPEAN UROLOGY 68 (2015) 267–279

Table 1 – Immune checkpoint targets in cancer immunotherapy

Target Function Antibody or Ig Description Indication Clinical Company


fusion protein development

CTL-4 Inhibitory Ipilimumab Fully human IgG1 mAb Unresectable metastatic melanoma FDA approval Bristol Myers
receptor Squibb
Urothelial carcinoma Phase 2
Metastatic CRPC Phase 2
RCC Phase 1b and 2
Tremelimumab Fully human IgG2 mAb Advanced melanoma Phase 3 Pfizer
PSA-recurrent prostate cancer and RCC Phase 1
PD-1 Inhibitory Nivolumab Fully human IgG4 mAb Metastatic melanoma Approval for treatment Bristol-Myers
receptor (BMS-936558; NSCLC Phase 3 Squibb
ONO-4538; RCC Phase 3
MDX-1106) CRPC Phase 1
Lambrolizumab Humanized IgG4 mAb NSCLC, RCC, melanoma Phase 3 Merck
(MK-3475) Triple-negative breast cancer, Phase 1
metastatic bladder cancer, and head
and neck cancer
Pidilizumab Humanized IgG1 mAb DLBCL, colon cancer, multiple Phase 2 CureTech
(CT-011) myeloma, acute leukemia, pancreatic
and bladder cancers
AMP-224 PD-L2 (B7-DC) human Solid tumors, cutaneous T-cell Phase 1 GlaxoSmithKline/
IgG1 fusion protein lymphoma Amplimmune
PD-L1 Ligand for BMS-936559 Fully human IgG4 mAb NSCLC, melanoma, RCC, pancreatic, Phase 1 Bristol Myers
PD-1 (MDX-1105) gastric, and breast cancers Squibb
RG7446/ Fully human IgG1 mAb Bladder cancer Phase 1 Roche/Genentech
MPDL3280 (engineered Fc NSCLC Phase 3
domain optimized)
MEDI4736 Fully human IgG1 mAb NSCLC Phase 3 Medimmune
(B7-H1 a)
b
MHCII Enhancing ImmuFact Soluble LAG-3 Ig Breast cancer Phase 3 Immutep
TReg cell IMP321 fusion protein Metastatic RCC Phase 1/2a
B7-H3 Inhibitory MGA271 Fully human IgG1 mAb Solid tumors in multiple cancers Phase 1 MacroGenics
(CD276) ligand (engineered Fc domain
optimized)
B7-H4 Inhibitory Fully human IgG mAb Preclinical
ligand development
TIM3 Inhibitory Mouse mAb Preclinical
receptor development
HLA-G Inhibitory Mouse mAb Preclinical
ligand development

CRPC = castration-resistant prostate cancer; DLBCL = diffuse large B-cell lymphoma; FDA = US Food and Drug Administration; Ig = immunoglobulin; LAG-
3 = lymphocyte activation gene 3; mAb = monoclonal antibody; NSCLC = non-small-cell lung carcinoma; PSA = prostate-specific antigen; RCC = renal cell
carcinoma; TIM3 = T-cell membrane protein 3.
a
B7 homology 1, PD-L1.
b
LAG-3 (CD223) ligand of MHCII antigen-presenting cell activation.

[(Fig._3)TD$IG] adverse events associated with ipilimumab. Most patients


included in these trials had very advanced castration-
Prostate cancer Kidney cancer Bladder cancer resistant prostate cancer (CRPC) that was refractory to
various therapies including chemotherapy. Moreover, when
Ar!cle selec!on obtained, objective responses tended to have a sustainable
120 142 29
n
effect over time.
One phase 3 RCT has been recently reported (Table 2)
Editorials 13 11 2 [16]. This trial assessed the efficacy of ipilimumab at the
Reviews 40 34 6
Other cancers 38 31 5 dose of 10 mg/kg every 3 wk for up to four cycles in
Case report 1 1 0
Preclinical 14 47 13
comparison with placebo in metastatic CRPC (mCRPC).
Language 3 7 1 After a mid-term follow-up of less than 12 mo, this study
was negative overall (median survival 11.2 vs 10.0 mo;
Eligible studies
p = 0.053). However, subgroup analyses suggested that
11 7 1
n ipilimumab might provide an OS benefit for patients with
favorable prognostic features and who do not have a heavy
Fig. 3 – Flow chart for article selection. Clinical trials reporting oncologic
metastatic burden. For the subgroup of patients with
results, tumor response rates, safety, and tolerability for use of antibodies
blocking immune checkpoints in prostate, kidney, and bladder cancer. favorable prognostic features, ipilimumab significantly
Table 2 – Outcome data from clinical trials of immune checkpoint inhibitors in urologic cancers

Clinical trial Phase Treatment Patients Population Endpoints Follow-up OS Factors favoring drug Conclusion
(mo) (mo) (subgroup analysis)

Prostate cancer
CTLA-4
Ipilimumab [16] 3 10 mg/kg vs 799 mCRPC (i) OS 9.6 11.2 vs 10.0 Race (white) (i) NS (p = 0.053)
placebo (1:1) Post-CT ECOG score 0 OS 0–5 mo: HR 1.46 (1.1–1.9)
(ii) PFS ALP <1.5 times the upper OS 5–12 mo: HR 0.65 (0.5–0.8)
Bone-directed RT limit of normal (ULN) OS >12 mo: HR 0.60 (0.4–0.9)
8 Gy before inclusion Gleason score>7
Normal LDH level (ii) S (p<0.0001)
No visceral metastases b
Hemoglobin >110 g/L
Not North America region
Low pain score
c
Ipilimumab [9] 1/2 10 mg/kg 50 mCRPC Safety 15.7 17.4 N/A PSA decline >50%: 16%
!RT (n = 34) PSA response RECIST criteria:

EUROPEAN UROLOGY 68 (2015) 267–279


RECIST Complete (3.6%)
Partial (7.1%)
Stable (21.4%)
Ipilimumab [10] 1 3 mg/kg 14 mCRPC (i) Safety N/A N/A N/A PSA decline >50%: 14.3%
(ii) PSA response
d
Ipilimumab + GM-CSF [11] 1 Ipilimumab 3 mg/kg 6 mCRPC (i) Safety N/A N/A N/A PSA decline >50%: 50%
+ GM-CSF (ii) PSA response RECIST criteria:
(ii) RECIST Complete (0)
Partial (16.7%)
Ipilimumab + vaccines [12,13] 1 1, 3, 5, 10 mg/kg 30 mCRPC (i) Safety N/A 34.4 No previous CT PSA decline>50%: 50%
+ PROSTVAC (ii) PSA response Immune biomarkers
(PD1+CD4+, CTLA-4/TReg)
e
Ipilimumab + vaccines [14,15] 1 3 mg/kg 16 mCRPC (i) Safety N/A 29.2 PSMA antibody response PSA decline >50%: 25%
+ GVAX (ii) PSA response Immune biomarkers (including e)
(T-cell regulation, low TReg
levels, CD4+/CTLA-4+)
Tremelimumab [17] 1 3–6 mg/kg 11 Rising PSA after (i) Safety N/A N/A N/A No significant increase
+ bicalutamide local treatment (ii) PSA kinetics in PSA doubling time
150 mg 6 mo
PD-1
Nivolumab [29,35] 1 10 mg/kg 17 CRPC (i) Safety N/A N/A N/A No objective response
(ii) RECIST
Bladder cancer
CTLA-4
Ipilimumab [22] 2 3–10 mg/kg 12 Neoadjuvant (i) Safety 7-33 N/A N/A Correlation between OS
precystectomy (ii) Immune and increase in CD4+ICOS+ T cells
monitoring
Kidney cancer
CTLA-4
a
Ipilimumab [18] 2 3 then 1 mg/kg 61 mRCC (i) Safety N/A N/A Severity of immune-related Response rate 12.5%
3 then 3 mg/kg (ii) RECIST adverse effects
No previous IL-2 therapy
Tremelimumab [20] 1 6, 10, 15 mg/kg 28 mRCC (i) Safety Partial response 42%
+ sunitinib (ii) RECIST

271
272 EUROPEAN UROLOGY 68 (2015) 267–279

improved OS compared with placebo (hazard ration

ALP = alkaline phosphatase; CT = chemotherapy; ECOG = Eastern Cooperative Oncology Group; GM-CSF = granulocyte/macrophage colony-stimulating factor; HR = hazard ratio; LDH = lactate dehydrogenase; mCRPC = metastatic
castration-resistant prostate cancer; mRCC = metastatic renal cell carcinoma; NA = not applicable (or not reported); NS = not significant; OS = overall survival; PFS = progression-free survival; RECIST = Response Evaluation Criteria
[HR] 0.64; p = 0.0038). Treatment with ipilimumab also
significantly improved progression-free survival com-

Objective response 11.8%

Objective response: 27%


Conclusion

pared with placebo in the overall cohort (4.0 vs 3.1 mo;

No objective response
Stabilization 34.5%
p < 0.001).

Stabilization: 27%
Stabilization 41%
Another key point was the delayed benefit reported in
patients receiving ipilimumab. Whereas short-term OS did
not differ between the ipilimumab and placebo arms,
survival curves began to diverge after 5 mo. The 2-yr OS rate
was 26.2% in the ipilimumab group and only 15.0% in the
placebo group. Continuing survival follow-up is warranted
Factors favoring drug
(subgroup analysis)

to draw strong conclusions and address whether blocking of


PD-L1-positive tumors

this immune checkpoint provides a survival advantage in


(response rate 36%
vs 0%; p = 0.0006)

mCRPC patients. Studies in advanced melanoma patients


Dose >6 mg

have suggested that the length of follow-up is fundamental


when assessing the sustainability of survival benefit of
ipilimumab treatment. In chemotherapy-naı̈ve prostate
NA

cancer and neoadjuvant settings, numerous clinical studies


assessing ipilimumab at an early stage are also under way
(NCT01194271, NCT01057810).
(mo)
OS

Another anti-CTLA4 antibody, tremelimumab, was eval-


NA

NA

NA

uated in a phase 1 trial [17]. Only 11 patients treated for


biochemical recurrence after local therapy were included.
Follow-up

Although the safety profile was good, no oncologic data are


(mo)

NA

NA

NA

yet available.

3.1.1.2. Kidney cancer. Immune-based therapy using IL-2 has


(i) Pharmacokinetics
(ii) Safety, RECIST

an objective response rate of 20% in renal cancer. Despite


Endpoints

this relatively low rate, a minority of patients have been


(ii) RECIST

(ii) RECIST

cured by this therapy and the majority of tumor regression


(i) Safety

(i) Safety

was long-lasting, leading to continued interest in immuno-


therapy for renal cancer management.
To date, three clinical trials have tested ipilimumab or
tremelimumab as antitumor agents in patients with
Population

metastatic renal cancer [18–20].


The first phase 1 trial of ipilimumab only reported the
mRCC

mRCC

mRCC

incidence of autoimmune hypophisitis in a mixed cohort of


Prior dose-escalation phase involving 12 patients (drug doses 0.3, 1. and 3 mg/kg).
Prior dose-escalation phase involving 18 patients (drug doses 0.5 and 1.5 mg/kg).

patients treated for melanoma (n = 113) or renal cancer


Prior dose-escalation phase involving 21 patients (drug doses 3 and 5 mg/kg).
Patients

(n = 50) [19]. A more mature phase 2 trial investigated the


17

34

21

safety of ipilimumab and the response rate in 61 patients


with metastatic renal cell carcinoma (RCC) [18]. The
objective partial response rate according to Response
0.3, 1, 3, 10 mg/kg

Evaluation Criteria in Solid Tumors (RECIST) was 12.5%.


Treatment

No complete response was noted. The duration of the


0.05–30 mg

response ranged from 7 to 21 mo. The absence of a


10 mg/kg

In the 40 patients receiving 3 mg/kg at each dose.

previous IL-2 regimen and the severity of immune-related


in Solid Tumors; RT = radiotherapy; S = significant.

toxicity were associated with tumor regression and its


sustainability.
Phase

A phase 1 dose-escalation trial investigated the combi-


1

Significant in multivariate analysis.

nation of sunitinib and tremelimumab in 28 patients with


metastatic RCC [20]. Three doses of tremelimumab (6, 10,
and 15 mg/kg) were investigated in combination with
sunitinib. Twenty-one patients were evaluable for tumor
Table 2 (Continued )

response, of whom nine (43%) showed objective partial


Nivolumab [29,36]
BMS-936559 [35]

responses. A median response duration was not achieved.


Clinical trial

IMP321 [69]

Nevertheless, from comparisons with data available from


phase 3 trials of sunitinib alone, the authors concluded that
MHC II
PD-L1

PD-1

the objective response rate did not differ obviously with the
d
b
a

addition of tremelimumab.
c
EUROPEAN UROLOGY 68 (2015) 267–279 273

3.1.1.3. Bladder cancer. Few preclinical studies have investi- although T cells surrounding the prostate are positive for
gated the role of CTLA-4 in bladder cancer carcinogenesis or PD-1 [30]. All CRPC cases stained for PD-L1 expression in the
aggressiveness. A recent report highlighted a significant study by Topalian et al [29] were negative, in accordance
association between CTLA-4 polymorphism and bladder with previous immunohistochemistry cohorts [31]. Phase
cancer risk in a case-control study [21]. 1b/2 trials are investigating the use of these PD-1/PD-L1
Given the ongoing trials assessing ipilimumab in other agents in prostate cancer (NCT01420965, NCT00730639).
urologic malignancies, one phase 2 trial investigated the
safety and efficacy of ipilimumab as a neoadjuvant drug 3.1.2.2. Kidney cancer. PD-1 expression in RCC, blood, tumors,
before radical cystectomy in muscle-invasive bladder and tumor-infiltrating lymphocytes has been independent-
cancers (Table 2) [22]. Twelve patients receiving ipilimumab ly associated with an increased risk of overall and cancer-
at a dose of 3–10 mg/kg for two cycles were included. In this specific mortality [32–34].
presurgical setting, the safety profile was acceptable and In kidney cancer, drugs targeting the PD-1/PD-L1 pathway
measurable immunologic effects were noted. Further trials have been tested in the metastatic setting [29,35,36].
are warranted to assess the oncologic and pathologic impact Tumor response to an anti–PD-L1 antibody was assessed
of such effects. as a secondary endpoint in a phase 1 trial in 17 patients with
metastatic RCC [36]. A total of 207 patients with advanced
3.1.2. PD-1/PD-L1 cancers (melanoma, lung, colorectal, ovarian, pancreatic)
Programmed death 1 (PD-1) is more broadly expressed than were included to test anti–PD-L1 drug safety. Before
CTLA-4: it is induced on other activated non–T-lymphocyte inclusion, 94%, 82%, and 41% of RCC patients had been
subsets, including B cells and natural killer (NK) cells, in treated using nephrectomy, antiangiogenic therapy, and
which it limits lytic activity. Therefore, PD-1 blockade, immunotherapy, respectively. An objective response
which enhances the activity of effector T cells, should also according to RECIST criteria was observed in two patients
enhance NK cell activity in tumors and tissues, and antibody (12%) with a durable response lasting 4 and 17 mo. Stable
production. PD-1 ligand 1 (PD-L1; also known as B7-H1 or disease lasting at least 24 wk was observed in seven
CD274) and PD-1 ligand 2 (PD-L2; also known as B7-DC or additional patients (41%). No survival data were available.
CD273) are the two ligands of PD-1 [23,24]. In a phase 1 trial assessing nivolumab (anti–PD-1), an
In contrast to CTLA-4, which regulates T-cell activation, objective response rate of 27% was estimated in 34 RCC
the major role of PD-1 [25] is to limit the activity of T cells in patients [29,35]. At a dose of 10 mg/kg, the response
peripheral tissues at the time of an inflammatory response duration ranged from 8 to 22 mo. In line with the anti–PD-
to infection, and to limit autoimmunity [26]. This translates L1 antibody trial, patients were heavily pretreated. Inter-
into a major immune resistance mechanism within the estingly, objective responses were observed in various
tumor microenvironment [27]. PD-1 expression is induced metastasis sites. Disease stabilization was also noted in 27%
when T cells are activated (Fig. 1) [25]. Engagement of PD-1 of patients, suggesting that more than half of tumors
by a ligand during antigen recognition induces cross- responded to nivolumab with prolonged disease control.
linkage of the antigen-receptor complex with PD-1. This The 6-mo progression-free survival rate was 56%.
results in phosphorylation of the tyrosine residue in the The superiority of one agent over the other should be
immunoreceptor tyrosine-based switch motif of PD-1 and evaluated in a dedicated direct comparison trial.
recruits the tyrosine phosphatase SHP-2, which depho- It has been noted in other malignancies that anti-PD-1/
sphorylates and inactivates proximal effector molecules PD-L1 therapy may induce transient worsening of disease
such as Syk in B cells and Zap70 in T cells. The immediate progression and that tumor responses may be delayed.
outcome of stimulation via PD-1 is inhibition of cell growth Nevertheless, the objective response rate and the prolonged
and cytokine secretion [28]. disease stabilization, even after discontinuation of immune
PD-1 is highly expressed on TReg cells, where it may therapy, encouraged further clinical trials. Several phase 1/2
enhance their proliferation in the presence of a ligand. Thus, trials are studying the use of anti–PD-1 and anti–PD-L1 agents
because many tumors are infiltrated with TReg cells, in renal cell cancers, and updated analyses from already
blockade of the PD-1 pathway may also enhance antitumor published trials confirmed durable clinical activity and
immune responses by diminishing the number and/or interesting response rates (NCT01472081, NCT01668784,
suppressive activity of intratumoral TReg cells. NCT01354431, NCT01441765, NCT01984242). Preliminary
results from NCT01375842 assessing the PD-L1 antibody
3.1.2.1. Prostate cancer. A recent multicenter phase 1 trial MPDL3280a have been presented, revealing 6-mo progres-
assessed the efficacy and safety profile of nivolumab, a fully sion-free survival of 50% [37]. Results from a randomized
human anti–PD-1 antibody, in a cohort of 296 patients with dose-ranging trial of nivolumab for metastatic renal cancer
advanced cancer (Table 2) [29]. Only 17 CRPC patients were were presented at the 2014 European Society for Medical
enrolled and treated with nivolumab. Whereas antitumor Oncology (ESMO) conference [38]. Overall, more than half of
activity was seen in melanoma, lung cancer, and RCC patients had objective responses lasting for >1 yr. Combina-
patients, no objective response was reported for CRPC. tions of nivolimab with pazopanib or sunitinb are also being
Immunostaining for PD-L1 expression has been suggested tested and show comparable 50% objective response rates
to explain these discrepancies in antitumor activity. [39]. Further clinical investigations should also focus on
Prostate cancer shows low levels of PD-L1 expression, theranostic factors. Preliminary data demonstrated that
274 EUROPEAN UROLOGY 68 (2015) 267–279

PD-L1 expression might be an interesting predictive bio- tumor grade and worse prognosis [52]. Animal models have
marker for response to PD-1 and PD-L1 inhibitors [40,41]. In demonstrated that anti–HLA-G blocking antibodies restore
the nivolumab trial, an objective response rate of 36% was antitumor immunity against HLA-G–expressing tumor cells
observed for PD-L1-positive tumors, in contrast to none for in vivo. Furthermore, HLA-G expression was associated with
PD-L1-negative tumors. tumor metastasis and poor survival in a humanized mouse
model of ovarian cancer [53].
3.1.2.3. Bladder cancer. PD-L1 immunohistochemistry in HLA-G plays a key role in the induction of immune
urothelial cancer has suggested that PD-L1 expression is tolerance and constitutes a novel immune escape mechanism
associated with tumor aggressiveness [42,43]. PD-1 and of tumor cells. Tissue HLA-G and soluble HLA-G were
PD-L1 were altered in a large proportion of urothelial upregulated in RCC [54,55]. However, the therapeutic role
cancers and, more importantly, PD-L1 expression predicted of HLA-G peptide-based immunotherapy requires further
overall mortality after cystectomy for patients with organ- investigation. HLA-G is also expressed in bladder cancer
confined tumors, suggesting that PD-L1 manipulation may [56,57], and therapeutic anti–HLA-G antibodies are currently
be a beneficial immunotherapy target in muscle-invasive in preclinical development for non–muscle-invasive bladder
bladder cancers [44,45]. cancer.
At least one phase 1 trial is under way to assess the safety
of a fully human anti–PD-L1 antibody, as well as two phase 3.1.4. Other immune checkpoint molecules
1/2 trials of anti–PD-1 drugs (lambrolizumab, pidilizumab). The molecules described above are those that have been
Preliminary results from the phase 1b KEYNOTE-012 studied the most and, in the case of CTLA-4 and PD-1, for
(NCT01848834) study were presented at the last ESMO which clinical developments are the most advanced.
conference [46]. Thirty-three heavily pretreated (at least Nevertheless, other immune checkpoint proteins have been
two prior therapies) patients with advanced urothelial identified in basic studies, some of which are under clinical
cancer were enrolled. A response rate of 24% was reported. evaluation. We briefly describe B7-H3, B7-H4, LAG3, and
Inhibition of PD-L1 also showed encouraging results, with a TIM3.
43% response rate for PD-L1 positivity on tumor biopsy B7-H3 protein (also known as CD276 [58]) is physiologi-
[47]. In a recent phase 1 expansion study with an adaptive cally expressed by activated monocytes, T cells, B cells, and
design that allowed for biomarker-positive enriched NK cells. The receptors for B7-H3 have not been identified
cohorts, tumors expressing PD-L1–positive tumor-infiltrat- and it has been reported that B7-H3 has both stimulatory and
ing immune cells had particularly high response rates [48]. inhibitory functions. Thus, B7-H3 is thought to have several
ligands that can trigger several, even antagonist functions.
3.1.3. HLA-G B7-H3 expression has been reported for tumor cell lines
HLA-G is a nonclassical HLA class I molecule whose primary and/or patient specimens including prostate cancer. It often
function is to protect the fetus from destruction by the correlated with increased tumor size. B7-H3 expression is
mother’s immune system, playing a role in maternal-fetal also associated with decreased numbers of tumor-infiltrating
tolerance [49–52]. Thus, HLA-G is one of the few proteins lymphocytes and suppression of antitumor T-cell responses.
whose physiological immune checkpoint function is B7-H4 [59] is a ligand of the B7 family whose receptor is
exerted primarily towards foreign tissues. Under normal still unknown. Its engagement decreases T-cell proliferation
physiological conditions, HLA-G is hardly expressed by and IL-2 production, as well as expansion of neutrophil
adult tissues. By contrast, most tumors neoexpress HLA-G at progenitors. Besides its immunoinhibitory function, B7-H4
different stages of their evolution, either on their cell may also directly promote the growth of cancer cells. B7-H4
surface or released as soluble forms. In vitro and in vivo overexpression has been identified in multiple solid
studies of the function of HLA-G revealed a broad malignancies, including renal cancer. It has been consis-
immunoregulating function that affects both innate and tently correlated with increasing tumor burden, neovascu-
adaptive immune responses. Through its receptors LILRB1, larization, and poor patient outcomes [60–62].
LILRB2 (also known as ILT2/CD85j and ILT4/CD85d), and LAG3 (lymphocyte-activation gene 3, also known as
KIR2DL4, which are differentially expressed by immune cells, CD223 [63]) is an inhibitory receptor that binds HLA class
HLA-G inhibits the cytolytic function of uterine and II molecules. It may be upregulated on tumor cells, and it is
peripheral blood NK cells, the antigen-specific cytolytic expressed at high levels by tumor-infiltrating antigen-
function of cytotoxic T lymphocytes (CTLs) and g/dT cells, the presenting cells and macrophages. LAG3 has a dual function:
alloproliferative response of CD4+ T cells, the proliferation of it inhibits T cell functions, in particular those of CD8+ T cells,
T cells and peripheral blood NK cells, and the maturation and and enhances the immunoinhibitory functions of TReg cells. A
function of dendritic cells (DCs). HLA-G also induces TReg cells synergic tumor escape function of PD-1 and LAG3 was
and myeloid suppressive cells. Thus, HLA-G is capable of recently shown in mice.
inhibiting all actors in antitumor responses and, in contrast to TIM3 [64] is a receptor for galectin 9, which is upregulated
both CTLA-4 and PD-1, of blocking all stages of such an in various types of cancer. TIM3 inhibits TH1 cell responses
antitumor response, from APC activation and effector and is coexpressed with PD-1 on tumor-specific CD8+ T cells.
priming, to the function of fully activated CTLs or NK cells. In animal models, blockade of both PD-1 and TIM3 enhanced
In cancer, HLA-G is expressed on tumor cells and tumor- antitumor immune responses and tumor rejection to a
infiltrating cells. HLA-G expression is associated with higher greater extent than blockade of each individual molecule.
EUROPEAN UROLOGY 68 (2015) 267–279 275

3.1.4.1. Prostate cancer. Molecules targeting other immune adverse clinicopathologic features and with metastatic
checkpoints are underevaluated in prostate cancer. Howev- status. In particular, tumor vasculature B7-H3 expression
er, preclinical and prognostic studies may lead to the was significantly associated with an increased risk of death
promotion of new clinical trials. A high level of B7-H3 from renal cancer [71]. Similar findings have been
expression in prostate cancer has been correlated with tumor suggested for B7-H4 expression [34].
progression, proliferation markers, and poor oncologic out-
comes, suggesting it is a promising therapeutic target 3.1.4.3. Bladder cancer. No clinical trial investigating the
[65–68]. usefulness of other immune blockades in bladder cancer
has been published. A target of interest might be B7-H3,
3.1.4.2. Kidney cancer. A recombinant soluble LAG-3 fusion whose expression is largely altered in bladder tumors.
protein, IMP 321, which agonizes MHC class II–driven The aim of the ongoing NCT01391143 phase 1 trial is to
dendritic cell activation, was safe and stabilized advanced assess the safety of the inhibitory ligand MGA271 in various
renal cancer in a phase 1 trial [69]. Disease stabilization was malignancies, including prostate, kidney, and bladder
observed in a third of patients, and this rate reached 87% cancers.
when the IMP 321 dose was >6 mg/injection. However, the
primary endpoints of this study were evaluations of 3.2. Safety
pharmacokinetics and pharmacodynamics, and further
clinical investigation is warranted. Safety was the primary endpoint in most phase 1/2 trials
To date, no clinical trial evaluating other drugs targeting assessing targeted immune therapy in urologic cancers.
immune checkpoints has been published. Several molecules Overall, immune therapy was relatively well tolerated.
are in development and preclinical data support their However, drug-related adverse events, including immune-
potential role in renal cancer therapy. related effects, were frequent. The frequency and severity of
B7-H3 expression is approximately 15% in RCC but drug-related adverse events are listed in Table 3.
reaches 95% in blood vessels surrounding such tumors The toxicity reported for CTLA-4 blockade includes
[70,71]. This vascular expression has been associated with immune-related adverse effects such as colitis, mostly

Table 3 – Toxicity of checkpoint inhibitors used in urologic cancers

Clinical trial Cancer Patients ECOG Grade 3/4 Discontinuation Most frequent immune-related adverse
score rate (%) effects

CTLA-4
Ipilimumab 10 mg/kg [16] Prostate 400 0: 42% Grade 3: 59% (vs 41%) 35 Diarrhea (39%), pruritus (20%), rash (17%),
1: 55% Grade 4: 17% (vs 11%) colitis (7%)
2: 1%
Ipilimumab 10 mg/kg [9] Prostate 50 0: 38% 46% 16 Diarrhea (54%), pruritus (20%), rash (32%),
1: 56% colitis (22%)
2: 0%
Ipilimumab 10 mg/kg [12] Prostate 15 0–1 18% 43.3 Diarrhea/colitis (26.7%), rash (53.3%),
pan-hypopituitarism (13.3%)
Ipilimumab 10 mg/kg [22] Urothelial 12 NA 33.3% 8.3 Diarrhea/colitis (58.3%), pruritus (25%),
rash (58.3%), uveitis (8.3%)
Ipilimumab 3 mg/kg [11] Prostate 6 0–1 50% N/A G3 diarrhea (16.7%), G3
pan-hypopituitarism (16.7%),
G3 temporal arteritis (16.7%)
Ipilimumab 3 mg/kg [10] Prostate 14 0: 71% Grade 3: 21% N/A Diarrhea (21.4%), rash (14.3%),
1: 28% Grade 4: 0 pruritus (7.1%)
Ipilimumab 3 mg/kg [14] Prostate 16 0: 94% Grade 3: 29% 19 Colitis (19%), hypophysitis (13%),
1: 6% Grade 4: 4% hepatitis (6%)
Ipilimumab 3 mg/kg [18] RCC 61 0: 61% Grade 3: 27.9% N/A Colonic perforation (5%), diarrhea/ colitis
1: 40% Grade 4: 3.3% (27.8%), hypopituitarism (3.3%),
Grade 5: 1.6% adrenal insufficiency (1.6%)
Tremelimumab [17] Prostate 11 0: 91% Grade 3: 27.3% 9.1 Diarrhea (27.3%), pruritus (27.3%),
1: 9% Grade 4: 0 rash (36.4%), colitis (9.1%)
Tremelimumab + mRCC 28 0: 64% 61% Colitis
sunitinib [20] 1: 36%
PD-1
Nivolumab 10 mg/kg [29] Prostate 17 0-2 14% N/A Diarrhea (9%), rash (8%), pruritus (7%),
RCC 34 dysthyroidism (3%)
Other cancers 245
PD-L1
BMS-936559 [35] RCC 17 0-2 9% 11 Diarrhea/colitis (9%), pruritus (6%),
Other cancers 190 rash (7%), dysthyroidism (4%)
MHC II
IMP 321 [69] RCC 21 0-1 0 0 Infusion reaction (10%)

ECOG = Eastern Cooperative Oncology Group; mRCC = metastatic renal cell carcinoma; NA = not applicable.
276 EUROPEAN UROLOGY 68 (2015) 267–279

Table 4 – Ongoing clinical trials

Clinical trial Drug Phase Cancer Estimated Population Primary endpoint Arms Estimated completion
enrollment date

CTLA-4
NCT01057810 Ipilimumab 3 Prostate 600 CT-naı̈ve mCRPC OS Versus placebo February 2016
NCT01530984 Ipilimumab 2 Prostate 54 CT-naı̈ve mCRPC PSA decline Versus ipilimumab + December 2018
GM-CSF
NCT01688492 Ipilimumab 1/2 Prostate 25 CT-naı̈ve mCRPC PSA decline With abiraterone September 2015
NCT01498978 Ipilimumab 2 Prostate 30 mCRPC PSA decline April 2018
NCT01194271 Ipilimumab 2a Prostate 20 Localized Immune response Neoadjuvant September 2015
NCT01804465 Ipilimumab 2 Prostate 66 CT-naı̈ve mCRPC Immune response With sipuleucel-T December 2016
NCT02231749 Ipilimumab 3 RCC 1070 mRCC OS, PFS With nivolumab October 2019
versus sunitinib
NCT01524991 Ipilimumab 2 Bladder 36 M OS With GC June 2016
PD-1
NCT01354431 Nivolumab 2 RCC 150 mRCC RECIST April 2015
NCT01668784 Nivolumab 3 RCC 822 mRCC RECIST Versus everolimus September 2016
NCT01441765 Pidilizumab 2 RCC 44 mRCC RECIST ! dendritic cell vaccine November 2015
NCT01420965 Pidilizumab 2 Prostate 57 mCRPC Immune response With sipuleucel ! December 2018
cyclophosphamide
NCT02210117 Nivolumab 2 RCC 45 mCRPC Safety ! sunitinib versus ! January 2019
ipilimumab
NCT01928394 Nivolumab 1/2 Bladder 410 M Response rate ! ipilimumab March 2017
PD-L1
NCT01984242 MPDL3280A 2 RCC 150 mRCC RECIST ! sunitinib versus January 2016
bevacizumab
NCT02108652 MPDL3280A 2 Bladder 330 M Response rate March 2016
B7-H3
NCT01391143 MGA271 1 Prostate 151 M Safety February 2016
Bladder
RCC

CRPC = castration-resistant prostate cancer; RCC = renal cell carcinoma; m = metastatic; GM-CSF = granulocyte/macrophage colony-stimulating factor;
OS = overall survival; PSA = prostate-specific antigen; PFS = progression-free survival; CT = chemotherapy; RECIST = Response Evaluation Criteria in Solid
Tumors; GC = gemcitabine and cisplatin.

manifested as diarrhea, dermatitis (pruritus, rash), uveitis, with anti–PD-L1 therapy (10%). It has been suggested that the
and hypophysitis. Arthritis, hepatitis, iritis, and vitiligo were level of PD-L1 expression in tumor tissue is correlated with
also seen. Manifestations of immune toxicity are generally the severity of toxicity.
treated with systemic steroids. Table 3 shows the rate of Toxicity induced by agents targeted at other immune
grade 3/4 events and the most frequent immune-related checkpoints is expected to be quite similar to that reported
adverse effects. Rash and diarrhea are frequent, occurring in for anti–CTLA-4 and anti–PD-1/PD-L1 antibodies. However,
approximately 30–40% of patients. The overall rate of grade the lack of published reports for phase 1/2 trials currently
3/4 toxicity ranged from 20% to >50% among studies. Grade precludes strong conclusions for urologic cancer patients.
5 adverse effects have also been noted, with a significant
rate of colonic perforation due to severe colitis in renal 4. Conclusions
cancer patients [18,20].
It has been suggested that the immune-related adverse An improved understanding of the molecular mechanisms
effects of anti–CTLA-4 antibodies and their severity are that govern interactions between a tumor and the host
correlated with objective tumor responses [18,72]. immune response has led to major advances in targeted
The type of immune-related adverse events for anti–PD-1/ immunotherapy and cancer treatments. Our systematic
PD-L1 agents was similar to that seen with ipilimumab review demonstrates that immune checkpoint inhibitors
[73]. Dermatological adverse events, diarrhea, and fatigue offer interesting and long-lasting response rates in heavily
occurred in approximately 20%, 20%, and 30% of cases, pretreated patients with advanced urologic cancers.
respectively. Severe colitis was infrequent. Pneumonitis has In prostate cancer, a growing body of data supports the
also been observed after the use anti-PD-1 agents [35]. oncologic role of anti–CTLA-4 antibodies, alone or in
Although anti–CTLA-4 and anti–PD-1/PD-L1 antibodies have combination with other immune therapies. A recent phase
not been directly compared, immune-related effects associ- 3 trial assessing ipilimumab in castration-resistant prostate
ated with anti–CTLA-4 drugs were more common and of cancer is negative; nevertheless, interesting response rates
higher grade. Thus, approximately 10% of patients experi- in cancers with favorable features encourage further
enced grade 3/4 adverse effects after using anti–PD-1/PD-L1 investigations and longer follow-up. Despite better tolera-
antibodies, compared with 20–50% after using CTLA-4 bility, PD-1/PD-L1 inhibitors have not yet shown satisfac-
inhibitors. Infusion reactions were more frequently observed tory oncologic outcomes in prostate cancer.
EUROPEAN UROLOGY 68 (2015) 267–279 277

In renal cancer, the most encouraging findings have References


been observed for PD-1/PD-L1 inhibitors given their safety
[1] Collins AV, Brodie DW, Gilbert RJC, et al. The interaction properties
and antitumor activity. Recent presentations at 2014 on-
of costimulatory molecules revisited. Immunity 2002;17:201–10.
cology conferences highlighted interesting and long- [2] Rudd CE, Taylor A, Schneider H. CD28 and CTLA-4 coreceptor
lasting tumor response rates ("50%). Final publications expression and signal transduction. Immunol Rev 2009;229:12–26.
for these trials are awaited. The superiority of one agent [3] Teft WA, Kirchhof MG, Madrenas J. A molecular perspective of
over the other should be evaluated in a dedicated head-to- CTLA-4 function. Annu Rev Immunol 2006;24:65–97.
head comparison trial. In bladder cancer, the use of [4] Lenschow DJ, Walunas TL, Bluestone JA. CD28/B7 system of T cell
targeted immunotherapy remains underevaluated and costimulation. Annu Rev Immunol 1996;14:233–58.
further trials are awaited. [5] Wing K, Onishi Y, Prieto-Martin P, et al. CTLA-4 control over Foxp3+
The field of immunotherapy in urologic cancer treatment regulatory T cell function. Science 2008;322:271–5.
[6] Hurwitz AA, Foster BA, Kwon ED, et al. Combination immunother-
is evolving. Oncologic efficacy, including prolongation of
apy of primary prostate cancer in a transgenic mouse model using
overall survival, has already been observed for immune
CTLA-4 blockade. Cancer Res 2000;60:2444–8.
checkpoint inhibitors in various malignancies, mainly in
[7] Demaria S, Kawashima N, Yang AM, et al. Immune-mediated inhi-
breast cancer and melanomas. Several ongoing trials are bition of metastases after treatment with local radiation and CTLA-
studying immune therapy combinations and immune 4 blockade in a mouse model of breast cancer. Clin Cancer Res
monotherapy combined with conventional anticancer 2005;11:728–34.
drugs (Table 4). An anti–CTLA-4 antibody and vaccine [8] Wada S, Jackson CM, Yoshimura K, et al. Sequencing CTLA-4 block-
combination has already been tested with interesting ade with cell-based immunotherapy for prostate cancer. J Transl
outcomes. Preclinical studies also support the synergistic Med 2013;11:89.
role of CTLA-4 and PD-1 blockade [74,75]. [9] Slovin SF, Higano CS, Hamid O, et al. Ipilimumab alone or in
The continual development of new drugs highlights the combination with radiotherapy in metastatic castration-resistant
prostate cancer: results from an open-label, multicenter phase I/II
need to improve the prediction of response to immune
study. Ann Oncol 2013;24:1813–21.
therapies and to optimize sequential therapeutic modali-
[10] Small EJ, Tchekmedyian NS, Rini BI, Fong L, Lowy I, Allison JP. A pilot
ties. Identification of activity and marker correlates should trial of CTLA-4 blockade with human anti-CTLA-4 in patients with
be included in newly opened clinical trials to guide hormone-refractory prostate cancer. Clin Cancer Res 2007;13:
treatment decision-making. In this light, the preliminary 1810–5.
data suggest that PD-L1 expression in tumor tissue might be [11] Fong L, Kwek SS, O’Brien S, et al. Potentiating endogenous antitu-
a predictive marker of tumor response to anti-PD-1 mor immunity to prostate cancer through combination immuno-
antibodies and might guide treatment choice. therapy with CTLA4 blockade and GM-CSF. Cancer Res 2009;69:
609–15.
Author contributions: Edgardo D. Carosella had full access to all the data [12] Madan RA, Mohebtash M, Arlen PM, et al. Ipilimumab and a
in the study and takes responsibility for the integrity of the data and the poxviral vaccine targeting prostate-specific antigen in metastatic
accuracy of the data analysis. castration-resistant prostate cancer: a phase 1 dose-escalation
trial. Lancet Oncol 2012;13:501–8.
Study concept and design: Carosella, Ploussard, LeMaoult, Desgrand- [13] Jochems C, Tucker JA, Tsang KY, et al. A combination trial of vaccine
champs. plus ipilimumab in metastatic castration-resistant prostate cancer
Acquisition of data: Carosella, Ploussard, LeMaoult, Desgrandchamps. patients: immune correlates. Cancer Immunol Immunother 2014;63:
Analysis and interpretation of data: Carosella, Ploussard, LeMaoult, 407–18.
Desgrandchamps. [14] van den Eertwegh AJM, Versluis J, van den Berg HP, et al. Combined
Drafting of the manuscript: Carosella, Ploussard, LeMaoult, Desgrand- immunotherapy with granulocyte-macrophage colony-stimulating
champs. factor-transduced allogeneic prostate cancer cells and ipilimumab
Critical revision of the manuscript for important intellectual content: in patients with metastatic castration-resistant prostate cancer: a
Carosella, Desgrandchamps. phase 1 dose-escalation trial. Lancet Oncol 2012;13:509–17.
Statistical analysis: None. [15] Santegoets SAM, Stam AM, Lougheed S, et al. T cell profiling reveals
Obtaining funding: None. high CD4+CTLA-4+ T cell frequency as dominant predictor for
Administrative, technical, or material support: None. survival after prostate GVAX/ipilimumab treatment. Cancer Immu-
Supervision: Carosella. nol Immunother 2013;62:245–56.
Other: None. [16] Kwon ED, Drake CG, Scher HI, et al. Ipilimumab versus placebo after
radiotherapy in patients with metastatic castration-resistant pros-
Financial disclosures: Edgardo D. Carosella certifies that all conflicts of
tate cancer that had progressed after docetaxel chemotherapy
interest, including specific financial interests and relationships and
(CA184-043): a multicentre, randomised, double-blind, phase 3 tri-
affiliations relevant to the subject matter or materials discussed in the
al. Lancet Oncol 2014;15:700–12.
manuscript (eg, employment/affiliation, grants or funding, consul-
[17] McNeel DG, Smith HA, Eickhoff JC, et al. Phase I trial of tremeli-
tancies, honoraria, stock ownership or options, expert testimony,
mumab in combination with short-term androgen deprivation in
royalties, or patents filed, received, or pending), are the following:
patients with PSA-recurrent prostate cancer. Cancer Immunol
None.
Immunother 2012;61:1137–47.
Funding/Support and role of the sponsor: None. [18] Yang JC, Hughes M, Kammula U, et al. Ipilimumab (anti-CTLA4
antibody) causes regression of metastatic renal cell cancer asso-
Acknowledgments: We thank Dr. Adrien Riviere, Urology Department,
ciated with enteritis and hypophysitis. J Immunother 2007;30:
APHP, Saint-Louis Hospital, for his help with the data collection for this
825–30.
review.
278 EUROPEAN UROLOGY 68 (2015) 267–279

[19] Blansfield JA, Beck KE, Tran K, et al. Cytotoxic T-lymphocyte-asso- [39] Amin A, Plimack ER, Infante JR, et al. Nivolumab in combination
ciated antigen-4 blockage can induce autoimmune hypophysitis in with sunitinib or pazopanib in patients with metastatic renal cell
patients with metastatic melanoma and renal cancer. J Immun- carcinoma. Ann Oncol 2014;25(Suppl 4), iv362–3.
other 2005;28:593–8. [40] Choueiri TK, Figueroa DJ, Fay AP, et al. Correlation of PD-L1 tumor
[20] Rini BI, Stein M, Shannon P, et al. Phase 1 dose-escalation trial of expression and treatment outcomes in patients with renal cell
tremelimumab plus sunitinib in patients with metastatic renal cell carcinoma receiving sunitinib or pazopanib: results from COM-
carcinoma. Cancer 2011;117:758–67. PARZ, a randomized controlled trial. Clin Cancer Res 2015;21:
[21] Wang L, Su G, Zhao X, et al. Association between the cytotoxic T- 1071–7.
lymphocyte antigen 4 +49A/G polymorphism and bladder cancer [41] Herbst RS, Soria J-C, Kowanetz M, et al. Predictive correlates of
risk. Tumour Biol 2014;35:1139–42. response to the anti-PD-L1 antibody MPDL3280A in cancer
[22] Carthon BC, Wolchok JD, Yuan J, et al. Preoperative CTLA-4 block- patients. Nature 2014;515:563–7.
ade: tolerability and immune monitoring in the setting of a pre- [42] Nakanishi J, Wada Y, Matsumoto K, Azuma M, Kikuchi K, Ueda S.
surgical clinical trial. Clin Cancer Res 2010;16:2861–71. Overexpression of B7-H1 (PD-L1) significantly associates with
[23] Freeman GJ, Long AJ, Iwai Y, et al. Engagement of the PD-1 immu- tumor grade and postoperative prognosis in human urothelial
noinhibitory receptor by a novel B7 family member leads to negative cancers. Cancer Immunol Immunother 2007;56:1173–82.
regulation of lymphocyte activation. J Exp Med 2000;192:1027–34. [43] Inman BA, Sebo TJ, Frigola X, et al. PD-L1 (B7-H1) expression by
[24] Tseng S-Y, Otsuji M, Gorski K, et al. B7-Dc, a New dendritic cell urothelial carcinoma of the bladder and BCG-induced granulomata:
molecule with potent costimulatory properties for T cells. J Exp associations with localized stage progression. Cancer 2007;109:
Med 2001;193:839–46. 1499–505.
[25] Ishida Y, Agata Y, Shibahara K, Honjo T. Induced expression of PD-1, [44] Xylinas E, Robinson BD, Kluth LA, et al. Association of T-cell co-
a novel member of the immunoglobulin gene superfamily, upon regulatory protein expression with clinical outcomes following
programmed cell death. EMBO J 1992;11:3887–95. radical cystectomy for urothelial carcinoma of the bladder. Eur J
[26] Keir ME, Butte MJ, Freeman GJ, Sharpe AH. PD-1 and its ligands in Surg Oncol 2014;40:121–7.
tolerance and immunity. Annu Rev Immunol 2008;26:677–704. [45] Boorjian SA, Sheinin Y, Crispen PL, et al. T-cell coregulatory mole-
[27] Dong H, Strome SE, Salomao DR, et al. Tumor-associated B7-H1 cule expression in urothelial cell carcinoma: clinicopathologic
promotes T-cell apoptosis: a potential mechanism of immune correlations and association with survival. Clin Cancer Res
evasion. Nat Med 2002;8:793–800. 2008;14:4800–8.
[28] Okazaki T, Chikuma S, Iwai Y, Fagarasan S, Honjo T. A rheostat for [46] Plimack ER, Gupta S, Bellmunt J, et al. A phase Ib study of pem-
immune responses: the unique properties of PD-1 and their advan- brolizumab in patients with advanced urothelial tract cancer. Ann
tages for clinical application. Nat Immunol 2013;14:1212–8. Oncol 2014;25(Suppl 4):mdu438.24.
[29] Topalian SL, Hodi FS, Brahmer JR, et al. Safety, activity, and immune [47] Bellmunt J, Petrylak DP, Powles T, et al. Inhibition of PD-L1
correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012;366: by MPDL3208a leads to clinical activity in patients with meta-
2443–54. static urothelial bladder cancer. Ann Oncol 2014;25(Suppl
[30] Sfanos KS, Bruno TC, Meeker AK, De Marzo AM, Isaacs WB, Drake 4):iv280.
CG. Human prostate-infiltrating CD8+ T lymphocytes are oligoclo- [48] Powles T, Eder JP, Fine GD, et al. MPDL3280A (anti-PD-L1) treat-
nal and PD-1+. Prostate 2009;69:1694–703. ment leads to clinical activity in metastatic bladder cancer. Nature
[31] Roth I, Corry DB, Locksley RM, Abrams JS, Litton MJ, Fisher SJ. 2014;515:558–62.
Human placental cytotrophoblasts produce the immunosuppres- [49] Carosella ED, Favier B, Rouas-Freiss N, Moreau P, Lemaoult J.
sive cytokine interleukin 10. J Exp Med 1996;184:539–48. Beyond the increasing complexity of the immunomodulatory
[32] Tang PA, Heng DY. Programmed death 1 pathway inhibition in HLA-G molecule. Blood 2008;111:4862–70.
metastatic renal cell cancer and prostate cancer. Curr Oncol Rep [50] Carosella ED, Gregori S, LeMaoult J. The tolerogenic interplay(s)
2013;15:98–104. among HLA-G, myeloid APCs, and regulatory cells. Blood 2011;118:
[33] Thompson RH, Dong H, Kwon ED. Implications of B7-H1 expression 6499–505.
in clear cell carcinoma of the kidney for prognostication and [51] Carosella ED, Moreau P, Lemaoult J, Rouas-Freiss N. HLA-G: from
therapy. Clin Cancer Res 2007;13, 709s–15s. biology to clinical benefits. Trends Immunol 2008;29:125–32.
[34] Krambeck AE, Dong H, Thompson RH, et al. Survivin and B7-H1 are [52] Rouas-Freiss N, Moreau P, LeMaoult J, Carosella ED. The dual role of
collaborative predictors of survival and represent potential thera- HLA-G in cancer. J Immunol Res 2014:359748.
peutic targets for patients with renal cell carcinoma. Clin Cancer [53] Lin A, Zhang X, Xu HH, Xu DP, Ruan YY, Yan WH. HLA-G expression
Res 2007;13:1749–56. is associated with metastasis and poor survival in the Balb/c nu/nu
[35] Brahmer JR, Drake CG, Wollner I, et al. Phase I study of single-agent murine tumor model with ovarian cancer. Int J Cancer 2012;131:
anti-programmed death-1 (MDX-1106) in refractory solid tumors: 150–7.
safety, clinical activity, pharmacodynamics, and immunologic cor- [54] Li BL, Lin A, Zhang XJ, et al. Characterization of HLA-G expression in
relates. J Clin Oncol 2010;28:3167–75. renal cell carcinoma. Tissue Antigens 2009;74:213–21.
[36] Brahmer JR, Tykodi SS, Chow LQ, et al. Safety and activity of anti- [55] Bukur J, Rebmann V, Grosse-Wilde H, et al. Functional role of
PD-L1 antibody in patients with advanced cancer. N Engl J Med human leukocyte antigen-G up-regulation in renal cell carcinoma.
2012;366:2455–65. Cancer Res 2003;63:4107–11.
[37] Cho DC, Sosman JA, Sznol M, et al. Clinical activity, safety, and [56] El-Chennawi FA, Auf FA, El-Diasty AM, et al. Expression of HLA-G in
biomarkers of MPDL3280A, an engineered PD-L1 antibody in cancer bladder. Egypt J Immunol 2005;12:57–64.
patients with metastatic renal cell carcinoma (mRCC). ASCO Meet- [57] Gan LH, Huang LF, Zhang X, et al. Tumor-specific upregulation of
ing Abstr 2013;31:4505. human leukocyte antigen-G expression in bladder transitional cell
[38] Motzer RJ, Rini BI, McDermott DF, et al. Randomized, dose-ranging carcinoma. Hum Immunol 2010;71:899–904.
phase II trial of nivolumab for metastatic renal cell carcinoma. Ann [58] Wang L, Kang F-B, Shan B-E. B7-H3-mediated tumor immunology:
Oncol 2014;25(Suppl 4):iv281. friend or foe? Int J Cancer 2014;134:2764–71.
EUROPEAN UROLOGY 68 (2015) 267–279 279

[59] Ceeraz S, Nowak EC, Noelle RJ. B7 family checkpoint regulators radiation therapy for recurrent prostate cancer. Int J Radiat Oncol
in immune regulation and disease. Trends Immunol 2013;34: Biol Phys 2011;79:1343–9.
556–63. [68] Roth TJ, Sheinin Y, Lohse CM, et al. B7-H3 ligand expression by
[60] Jiang J, Zhu Y, Wu C, et al. Tumor expression of B7-H4 predicts poor prostate cancer: a novel marker of prognosis and potential target
survival of patients suffering from gastric cancer. Cancer Immunol for therapy. Cancer Res 2007;67:7893–900.
Immunother 2010;59:1707–14. [69] Brignone C, Escudier B, Grygar C, Marcu M, Triebel F. A phase I
[61] Krambeck AE, Thompson RH, Dong H, et al. B7-H4 expression in pharmacokinetic and biological correlative study of IMP321, a
renal cell carcinoma and tumor vasculature: associations with novel MHC class II agonist, in patients with advanced renal cell
cancer progression and survival. Proc Natl Acad Sci U S A carcinoma. Clin Cancer Res 2009;15:6225–31.
2006;103:10391–6. [70] Qin X, Zhang H, Ye D, Dai B, Zhu Y, Shi G. B7-H3 is a new cancer-
[62] Tringler B, Zhuo S, Pilkington G, et al. B7-H4 is highly expressed specific endothelial marker in clear cell renal cell carcinoma. Onco
in ductal and lobular breast cancer. Clin Cancer Res 2005;11: Targets Ther 2013;6:1667–73.
1842–8. [71] Crispen PL, Sheinin Y, Roth TJ, et al. Tumor cell and tumor vascu-
[63] Norde WJ, Hobo W, van der Voort R, Dolstra H. Coinhibitory lature expression of B7-H3 predict survival in clear cell renal cell
molecules in hematologic malignancies: targets for therapeutic carcinoma. Clin Cancer Res 2008;14:5150–7.
intervention. Blood 2012;120:728–36. [72] Beck KE, Blansfield JA, Tran KQ, et al. Enterocolitis in patients with
[64] Ngiow SF, Teng MWL, Smyth MJ. Prospects for TIM3-targeted cancer after antibody blockade of cytotoxic T-lymphocyte-associ-
antitumor immunotherapy. Cancer Res 2011;71:6567–71. ated antigen 4. J Clin Oncol 2006;24:2283–9.
[65] Liu Y, Vlatkovic L, Saeter T, et al. Is the clinical malignant phenotype [73] Dolan DE, Gupta S. PD-1 pathway inhibitors: changing the land-
of prostate cancer a result of a highly proliferative immune-evasive scape of cancer immunotherapy. Cancer Control 2014;21:231–7.
B7-H3-expressing cell population? Int J Urol 2012;19:749–56. [74] Wolchok JD, Kluger H, Callahan MK, et al. Nivolumab plus ipilimu-
[66] Yuan H, Wei X, Zhang G, Li C, Zhang X, Hou J. B7-H3 over expression mab in advanced melanoma. N Engl J Med 2013;369:122–33.
in prostate cancer promotes tumor cell progression. J Urol [75] Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4
2011;186:1093–9. combination blockade expands infiltrating T cells and reduces
[67] Parker AS, Heckman MG, Sheinin Y, et al. Evaluation of B7-H3 regulatory T and myeloid cells within B16 melanoma tumors. Proc
expression as a biomarker of biochemical recurrence after salvage Natl Acad Sci U S A 2010;107:4275–80.

You might also like