Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

Annals of Oncology 29: 84–91, 2018

doi:10.1093/annonc/mdx755
Published online 7 December 2017

REVIEW

Comprehensive analysis of the clinical


immuno-oncology landscape

J. Tang, A. Shalabi & V. M. Hubbard-Lucey*


The Anna-Maria Kellen Clinical Accelerator, Cancer Research Institute, New York, USA

*Correspondence to: Dr Vanessa M. Hubbard-Lucey, Anna-Maria Kellen Clinical Accelerator, Cancer Research Institute, 29 Broadway, 4th Floor, New York, NY 10006, USA.
Tel: þ1-212-6887515; E-mail: vlucey@cancerresearch.org

Advances from immuno-oncology (IO) are changing the standard of care of many types of cancer, and the paradigm of cancer
treatments and drug development is being rewritten on a regular basis. Moreover, an unprecedented number of new
investigational agents and companies are entering the field of IO. As such, it has become challenging for oncology physicians
conducting clinical trials, industry veterans developing IO drugs, and even regulators reviewing novel IO agents to keep track of
the rapidly evolving landscape. To help the key stake holders in the field understand the latest IO landscape, we sought to
present an unbiased, neutral, scientifically curated, and timely updated analysis of all the current IO agents in clinical
development and the clinical trials testing these agents. We based our analyses on information collected from numerous
trusted and publicly available sources. We have developed two databases. One database tracks 2004 IO agents (940 in clinical
stage and 1064 in preclinical stage) against 303 targets, from 864 companies; the other tracks 3042 active clinical trials of these
agents with a target enrollment of 577 076 patients. This report provides key analyses of these data. Furthermore, we will discuss
a number of important and actionable trends in the current IO landscape: a large number of companies developing agents
against the same IO targets; a rapid increase in the number of anti-PD-1/L1 combination studies, many of which are testing the
same combinations and following inefficient patterns; and a significant increase in the number of small, investigator-initiated
studies. For each of the findings, we speculate the causes and discuss a few initiatives that aim to address some of these
challenges. Finally, by making these landscape analyses available, we aspire to inform the cancer community as they seek to
strive for efficiencies and innovation while avoiding duplication.
Key words: immuno-oncology, cancer immunotherapy, landscape analysis, clinical trials, tumor immunology

Introduction CD3-targeted bispecific antibody (also targeting CD19) have


Advances in immuno-oncology (IO) are revolutionizing the been approved [4–7]. These novel immunotherapies, in most
standard of care for many types of cancer. As of our data cut-off cases, are designed to treat advanced, refractory, or relapsed
date (September 2017), 26 immunotherapies have been cancers that did not respond to standard-of-care cancer treat-
approved, and 17 types of cancer have at least one approved im- ments (Table 1).
munotherapy as a treatment option. The first approval of modern Since the approval of ipilimumab, the pipeline of IO agents in
cancer immunotherapy was interferon-a in 1986 for hairy cell clinical and preclinical development has become very crowded
leukemia, and later for chronic myelogenous leukemia, follicular [8]. As of September 2017, there were 940 IO agents in clinical de-
non-Hodgkin lymphoma, melanoma, and AIDS-related Kaposi’s velopment, with another 1064 in preclinical phase. We also found
sarcoma [1]. Several other agents have been approved since then, that 3042 interventional active clinical trials are evaluating these
but a transformation in the landscape of IO started with the ap- clinical-stage immunotherapies with a target of enrolling 577 076
proval of ipilimumab—a checkpoint inhibitor targeting CTLA- patients (supplementary Table S1, available at Annals of Oncology
4—for advanced melanoma in 2011 [2, 3]. In the past 3 years online). These IO trials cover all common cancer types and the
alone, five new checkpoint inhibitors (targeting PD-1 or PD-L1), majority of the less common ones, and promising results are
two new cell therapies (targeting CD19), and one new being reported on a daily basis in major conferences and clinical

C The Author 2017. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
V
All rights reserved. For Permissions, please email: journals.permissions@oup.com.
Annals of Oncology Review
Table 1. The list of 26 approved immuno-oncology agents, company origins, and targets

Therapy type Therapy name Company Target

T-cell-targeted immunomodulatory (six in total) Ipilimumab Bristol-Myers Squibb Co. CTLA-4


Nivolumab Bristol-Myers Squibb Co. PD-1
Pembrolizumab Merck & Co Inc. PD-1
Atezolizumab Roche/Genentech Ltd PD-L1
Avelumab Merck KGaA PD-L1
Durvalumab AstraZeneca/MedImmune LLC PD-L1
Other immunomodulatory (eight in total) Aldesleukin Novartis AG IL2R
Imiquimod Valeant Pharmaceuticals Intl Inc. TLR7
Interferon alfa Sumitomo Dainippon Pharma Co Ltd IFNAR1; IFNAR2
Interferon alfa-1b Shenzhen Kexing Biotech Co Ltd IFNAR1
Interferon alfa-2a Cadila Healthcare Ltd IFNAR1; IFNAR2
Interferon alfa-2b Merck & Co Inc. IFNAR1; IFNAR2
Interferon beta Toray Industries Inc. IFNAR1
Interferon gamma-1a Otsuka Pharmaceutical Co Ltd IFNAR1
Cancer vaccine (seven in total) BCG Live Shire Plc TLR
ImmuCyst Sanofi TLR
Immuno BCG Ataulpho Paiva Foundation TLR
Mycidac-C Cadila Pharmaceuticals Ltd TLR2
Sipuleucel-T Dendreon Unspecified TAA
TICE BCG Merck & Co Inc. TLR
Uro-BCG Medac Inc. TLR
Cell therapy (two in total) Tisagenlecleucel Novartis AG CD19
Axicabtagene ciloleucel Gilead CD19
Oncolytic virus (two in total) Oncorine Shanghai Sunway Biotech Co Ltd CD40L
Talimogene laherparepvec Amgen Inc. GMCSFR
CD3-targeted bispecific ab Blinatumomab Amgen Inc. CD19 X CD3

journals. The large numbers of promising experimental therapies, PD-1, CD40, and GITR); (ii) other immunomodulators that act
active clinical trials, and planned patient enrollment suggest that on other immune cells or the tumor immune microenvironment
we are entering a new era that could change the standard-of-care to unleash antitumor immunity (e.g. agents modulating IFNAR,
in even more cancer types. CSF1R, IDO1, A2AR, and KIR); (iii) cancer vaccines that induce
However, the fast-expanding and ever-changing IO field could antigen-specific antitumor immunity (e.g. sipuleucel-T); (iv) cell
overwhelm even the most experienced clinical investigators as therapies that engineer immune cells such as T cells to directly
they seek treatment options for their patients. To help the cancer attack cancer cells (e.g. anti-CD19 CAR-T); (v) oncolytic viruses
community stay informed of the current IO landscape, the Anna- that rely on both direct tumor killing and activation of antitumor
Maria Kellen Clinical Accelerator of the Cancer Research immunity (e.g. T-VEC); and (vi) CD3-targeted bispecific anti-
Institute (CRI), the clinical research program of CRI, provides bodies that bring T cell to the targeted tumor cells for direct kill-
herein an unbiased, neutral, comprehensive, and scientifically ing (e.g. blinatumomab) (Figure 1).
curated landscape analysis that aims to inform the physician-
scientists, drug developers, regulators, and other stakeholders of
the latest trends in IO clinical development. Concentration of agents on a few targets. The 940 clinical-stage
IO agents are owned by 462 different companies or academic in-
stitutes (supplementary Table S2, available at Annals of Oncology
online), and these agents modulate 271 different targets.
The landscape of clinical immuno-oncology Interestingly, a closer investigation of the 940 agents revealed that
almost half of them modulate only 40 targets (Figure 2). For ex-
A large number of IO drugs in clinical development ample, we found 164 agents targeting PD-1 or PD-L1 (PD-1/L1),
with significant duplication with 50 clinical-stage agents targeting PD-1/L1, 34 of which are
monoclonal antibodies (Figure 3). This confirms our assumption
Overview of the Clinical Accelerator IO database. Our database of significant duplication, despite the fact that five anti-PD-1/L1
included 2004 IO agents as of September 2017, 940 of which are monoclonal antibodies have already been approved [9].
in clinical development. On the basis of different mechanisms of Although it is still unclear if targeting the PD-1 receptor or its lig-
actions, we classified these agents into six categories: (i) T-cell- and PD-L1 will result in any differences in efficacy and safety,
targeted immunomodulators that act on inhibitory or activating none of the agents or studies in our database show any head-to-
molecules expressed by T cells (e.g. agents targeting CTLA-4, head evaluation in patients. In another example, agents targeting

Volume 29 | Issue 1 | 2018 doi:10.1093/annonc/mdx755 | 85


Review Annals of Oncology
Clinical
T-cell targeted immunomodulator agent count
99
Other immunomodulator
170
Cancer vaccine
344
Cell therapy Clinical stage
Approved 224
Oncolytic virus Phase III
Phase II 69
Phase I/II
CD3-targeted bispecific antibody Phase I
Preclinical & Discovery 34
350 300 250 200 150 100 50 0 50 100 150 200 250 300 350
Number of agents

Figure 1. The overview of 2004 immuno-oncology (IO) agents. Six classes of IO agents are identified on the basis of different mechanisms of
actions.

Target
Unspecified TAA
CD19
PD-1
HER2
NY-ESO-1
PD-L1
CD20
CD40
EGFR
CSF1R
GD2
WT1
IFNAR1
MUC1
PSMA
TERT
Survivin
HPV E6
Personalization
BCMA
CEA
LAG-3 Therapy type
OX40
TLR T-cell targeted immunomodulator
TLR7
CD123 Other immunomodulator
GITR
GPC3 Cancer vaccine
HPV E7
IDO1 Cell therapy
IL2
mesothelin Oncolytic virus
PSA CD3-targeted bispecific mAb
TLR9
CD22
CD30
CD47
MAGE-A3
ADORA2A
CD25
0 10 20 30 40 50 60 70 80 90 100 110 120
Number of clinical agents

Figure 2. The top 40 targets with the most clinical-stage immuno-oncology agents. Unspecified tumor-associated antigen is defined as the
target of unspecified antigens from either individual patients and cell lines. In this sense, therapies against this target mainly consist of cell
line-based vaccines, autologous tumor vaccines, autologous DC vaccines, and tumor-infiltrating T-cell therapies. Personalization is defined as
the targets discovered by algorithm-based approaches from, e.g. the individual patients’ tumor samples or blood circulating DNA.

unspecified tumor-associated antigens (unspecified TAA) rep- Rapid proliferation of CAR-T-cell therapy, especially in China.
resent the largest number of clinical IO agents. There are now CAR-T-cell therapy has delivered unprecedented results, with
114 agents in the clinic, 98 of which are cancer vaccines. Of the two recent approvals of CD19-targeted CAR-T-cell therapies
114 unspecific TAA-targeted IO agents, only sipuleucel-T (an au- [11, 12]. CD19 is also the most common CAR-T target, with
tologous dendritic cell vaccine) has been approved for prostate 56 clinical-stage therapies against it (supplementary Table S3,
cancer [10]. This number of agents raises questions on efficien- available at Annals of Oncology online). Surprisingly, by interrog-
cies of resources and, importantly, the patients who enroll into ating our IO drug database, we found 291 different CAR-T thera-
clinical trials. The concentration of IO development and patient pies, with 162 of them being tested clinically. These agents are
resources on a few targets, some with already approved drugs, being developed by 136 different companies and academic cen-
could potentially be stalling future innovation. Rather, investing ters. Interestingly, companies and academic centers from China
these precious resources more efficiently could help accelerate seem to lead the total number of clinical-stage CAR-T therapies,
needed progress in finding cures to this deadly disease. with 98 clinical-stage agents owned by 46 companies from China

86 | Tang et al. Volume 29 | Issue 1 | 2018


Annals of Oncology Review
compared with 51 clinical agents owned by 22 companies from The landscape of anti-PD-1/L1 combination trials
the United States, with a much smaller number in other countries
combined (Figure 4 and supplementary Table S4, available at Trends in the landscape of 1105 PD-1/L1 combo therapy trials.
Annals of Oncology online). Despite the approval of five anti-PD-1/L1 antibody agents, our
database shows that there are numerous clinical trial programs
120 114
that are ongoing and actively recruiting patients. The clinical-stage
PD-1/PD-L1 agents are evaluating clinical questions across 1502
100 different studies. Of these, 1105 are combination trials that com-
Molecule type
bine anti-PD-1/L1 agents with other IO therapies, targeted thera-
Monoclonal antibody
Number of records

80
Bispecific antibody pies, chemotherapies, radiotherapies, or chemoradiotherapies. It is
Small molecule worth noting that 49 of the combination trials are testing agents
60 Other biologic that have not been approved, whereas the majority of combination
trials focus on the five approved agents (Figure 5).
40
30
20 Fragmentation and lack of coordination in PD-1/L1 combination
9 5 trial landscape. A deeper analysis of the 1105 trials reveals some
3 3
0 interesting observations. First, there has been a rapid increase in
Preclinical & Phase I Phase I/II Phase II Phase III Approved
Discovery new combination studies in the past 5 years. Analysis of our data-
base shows that in 2017 alone, 469 new studies were started, with a
Figure 3. The overview of 164 anti-PD-1/L1 agents. Of note, 164 target enrollment of 52 539 patients (Figure 6). Second, agents
agents are currently in development, 50 of whom are in clinical against 165 different targets are being combined with anti-PD-1/L1
stages. agents (supplementary Table S5, available at Annals of Oncology

China Clinical agent count

98

US
Clinical stage
51 Approved
Phase III
Other
Phase II
13 Phase I/II
Phase I
80 60 40 20 0 20 40 60 80 100 Preclinical & Discovery
Number of agents

Figure 4. The landscape of CAR-T-cell therapy. China leads the total number of clinical-stage CAR-T-cell therapies, whereas United States has
the largest number of preclinical agents.

399 Combination type


400
IO
Targeted
350 340
Chemotherapy
Radiotherapy
300 Chemoradiotherapy
Multi-way combo
Number of total trials

250

200
161
150
124

100

49
50 32

0
Pembrolizumab Nivolumab Durvalumab Atezolizumab Avelumab Others

Figure 5. Analysis of PD-1/L1 combination trial types. The majority of combination trials focus on the five approved agents. Combination
with other immuno-oncology agents, targeted therapies, and chemotherapies are the top common strategies.

Volume 29 | Issue 1 | 2018 doi:10.1093/annonc/mdx755 | 87


Review Annals of Oncology
Trial start year 2009 2010 2011 2012 2013 2014 2015 2016 2017
Clinical phase
Phase 4
Phase 3
Phase 2
Phase 1/2
Phase 1

Number
1 5 2 13 20 58 190 329 469
of new trials
Planned new
136 2473 582 4867 5031 11 276 39 821 46 153 52 539
enrollment
Planned
enrollment 136 495 291 374 252 194 210 140 112
per new trial

Figure 6. The rapid increase of new anti-PD-1/L1combination trials in the past 5 years. The size of the bubble correlates to the target enroll-
ment of patients. Multiple bubbles of the same color represent multiple cancer types that are being tested in these trials.

Numbers of trials using common combo strategies:

1. Anti-CTLA-4 agents: 251

2. Chemotherapies: 170

3. Radiotherapies: 64

4. Anti-VEGFA agents: 43

5. Chemoradiotherapy combos: 42

Figure 7. The landscape analysis of targets of anti-PD-1/L1 combination trials. The size of the bubble correlates to the number of trials.

online). Many of these studies we observed have significant over- New trial designs should be considered for IO
lap and duplication of the combination partner targets. For in- development
stance, anti-CTLA-4 agents and chemotherapies are the two
most prevalent classes of therapies being combined with anti- Breakthrough treatments sooner. The early activity observed with
PD-1/L1 agents, with 251 and 170 studies, respectively (Figure anti-PD-1/L1 agents across multiple tumor types has accelerated
7). This could possibly be driven by the evidence of preclinical the use of novel trial designs, several of which have led to regula-
synergy of the combination [13], or the recent approvals of anti- tory approvals. There are several examples of novel study designs
PD-1/anti-CTLA-4 combination for advanced melanoma and where drugs have leaped through the drug development phases:
anti-PD-1/chemotherapy combination as the first-line treat- from first-in-human, dose selection, safety, efficacy, biomarker
ment of advanced non-small-cell lung cancer (NSCLC) [14, 15]. selection, all the way to regulatory approval, in one single study.
However, it should be questioned if 251 and 170 studies are Particularly, the proliferation of agents has resulted in a competi-
needed to answer clinically relevant questions. Other prevalent tive drug development landscape with a need to answer clinical
combination partners are anti-VEGF agents (predominantly questions faster within a shorter timeframe by using these novel
with bevacizumab) and radiochemotherapy, with some having trial designs. One example is KEYNOTE-001, a phase I trial that
already shown clinical benefits [16]. This wide coverage of dis- led to two FDA approvals, one in melanoma and the other in
tinct targets, in our opinion, reflects a fragmented and uncoor- advanced NSCLC [17]. Moreover, the PD-L1 biomarker was
dinated approach to drug development across the anti-PD-1/L1 evaluated and confirmed all within the same study. KEYNOTE-
combination trial landscape. 001 is arguably the largest phase I trial, which finally recruited

88 | Tang et al. Volume 29 | Issue 1 | 2018


Annals of Oncology Review
1245 patients. Its adaptive design allowed the trial to seamlessly approvals (FDA sub-part H), which require confirmatory trials
transition from generating safety data to producing regulatory to validate the results [23, 24]. These novel trial designs, coupled
submission-quality efficacy data rather than starting separate with highly flexible and cooperative attitudes from regulators
studies for each clinical question [17]. Speed of development was globally have significantly accelerated the delivery of innovative
also demonstrated in this innovative trial design. In <4 years, and clinically beneficial immunotherapies to patients with
KEYNOTE-001 went from IND filing to the approval of pembro- cancer.
lizumab for ipilimumab-resistant metastatic melanoma [18].
One year after the first approval, results from this same trial led to
A notable shift in study sponsorship from
the second approval in advanced NSCLC [19].
companies to investigators
Another example of novel study design involves biomarker-
based, histology-agnostic basket trial design. It was known that Rapid increase of investigator-initiated trials in the early develop-
tumors with high level of microsatellite instability (MSI-H) or ment cycle. Traditionally, companies conduct most early- and
deficiency in mismatch DNA repair (dMMR) have very high level mid-stage development of a compound themselves—through
of nonsynonymous mutations and high immunogenicity, which multicenter, industry-sponsored trials. Once the agent is closer to
associates with a high response rate to anti-PD-1 therapy [20, regulatory approval, the companies would allow more ‘investiga-
21]. In KEYNOTE-059, a phase II basket trial, patients with solid tor-initiated’ exploratory studies to be conducted. However, for
tumors tested positive with MSI-H or dMMR were enrolled, re- IO agents, our analysis shows that this traditional development
gardless of their cancer types. Indeed, 149 enrolled patients with paradigm is no longer followed. First, we found that of the 1105
MSI-H or dMMR tumors demonstrated a 39.6% objective re- anti-PD-1/L1 combo trials, 60% (655 out of 1105) are sponsored
sponse rate, which led to the first histology-agnostic approval of a by non-industry sources, such as individual academic centers,
cancer treatment in the United States [22]. Notably, most ap- government agencies, collaborative groups, or nonprofit entities
provals in IO today are based on unprecedented benefit–risk (Figure 8). Investigator-initiated studies are deemed more ex-
ratios, with results from relatively small datasets. However, most ploratory and have an enrollment target of 76 patients per trial on
of these early approvals in the United States are accelerated average, compared with 257 patients per industry-sponsored trial
(Figure 9). Furthermore, as most of the investigator-initiated tri-
Number of trials Planned enrollment als are being conducted at single centers, it is unrealistic that these
(1105 in total) (1 65 215 in total)
655 non-industry trials will ever achieve the overall target enroll-
ment of 50 000 patients (Figure 8). This enormous recruitment
49 693
target, in our opinion, is also a challenging hurdle with many du-
450
plicate questions being addressed and limited patient resources.

655
1 15 522 Summary and discussion
Sponsorship
In summary, the current clinical landscape of IO presents enor-
mous enthusiasm from academia and industry, which is exempli-
Non-industry-sponsored trials
fied by 940 clinical-stage IO agents, 3042 immunotherapy trials,
Industry-sponsored trials
and 1 105 anti-PD-1/L1 combination trials. The field is very
Figure 8. Comparison of total trial numbers and target enrollment be- promising and has the potential to deliver many breakthroughs
tween industry-sponsored and non-industry-sponsored anti-PD-1/L1 to change the standard of care of many cancer types. However, it
combination trials. is also very crowded, fragmented, and uncoordinated with

Industry-sponsored trials 700


Average target enrollment per new trial

400 Non-industry-sponsored trials


600
Number of new trials

300 500

400

200
300

200
100
100

0 0
2009 2010 2011 2012 2013 2014 2015 2016 2017

Figure 9. Comparison of average target enrollment between trials with different sponsorship.

Volume 29 | Issue 1 | 2018 doi:10.1093/annonc/mdx755 | 89


Review Annals of Oncology
significant duplication. Our research has identified and con- nonprofit organizations work together. This cross-sector collab-
firmed several opportunities and challenges: many IO agents oration, deep commitment to following the science, and adopting
concentrate on a few targets such as PD-1; anti-PD-1/L1 combin- novel study design would be the best ways to bring the true prom-
ation trial space is very fragmented and uncoordinated; trad- ise of cancer immunotherapies to patients, sooner than later.
itional trial designs are not suited for the fast-changing landscape
of clinical IO development; and a significant proportion of the
increase in studies can be attributed to investigator initiated IO Methods
trials—small, single-center trials that may fail to recruit enough
patients and produce high-quality results. We now discuss several All the information collected for these analyses came from publicly avail-
able sources, press releases, quarterly reports, web-based company pipe-
ongoing trends that could address these challenges.
lines, news alerts, and GlobalData (London, UK). All data were manually
There are clear limitations to traditional trial designs, namely curated and analyzed using Tableau Software (v10.4).
single-center studies and the pursuit of the same biological targets
by multiple companies. From our research of the IO landscape,
there is an urgent need to enhance efficiencies. In a recent publi- Acknowledgement
cation led by the FDA, the authors provided an eloquent sum-
The authors would like to thank the generous donors to the
mary and examples of collaborative and novel trial designs,
Cancer Research Institute, particularly those who give philan-
which could allow more questions to be answered more effi-
thropically to the Anna-Maria Kellen Clinical Accelerator, espe-
ciently in a single multicenter trial [25]. These umbrella, basket,
cially Michael Kellen.
or platform designs create a unique opportunity to drive collab-
oration, coordination, and accelerated innovation.
Although many biopharmaceutical companies are adopting
such study designs, they still tend to include drugs only from their Funding
own portfolio. The nonprofit and public sectors can play an im- None declared.
portant role in facilitating and conducting these innovative trials
across multiple companies and research centers. There are now
several examples of nonprofit organizations leading these novel
study designs that have made significant contributions to the Disclosure
field. Notably the I-SPY program for breast cancer [26] is a JT and VHL have nothing to disclose. AS has an advisory role
unique nonprofit-led collaborative that has recruited over 1000 with AstraZeneca.
patients and has graduated multiple agents into pivotal develop-
ment. In another program, the LUNG-MAP for lung cancer [27]
was a multi-party collaboration between Friends of Cancer References
Research, Foundation for NIH, National Cancer Institute, the 1. Talpaz M, Kantarjian HM, McCredie K et al. Hematologic remission and
Southwest Oncology group, and multiple biopharmaceutical and cytogenetic improvement induced by recombinant human interferon
diagnostic companies. Despite a slow start and operational com- alpha A in chronic myelogenous leukemia. N Engl J Med 1986; 314(17):
1065–1069.
plexities, the study is now open with multiple arms at hundreds
2. Hodi FS, O’Day SJ, McDermott DF et al. Improved survival with ipili-
of sites. There is an opportunity to learn from these experiences mumab in patients with metastatic melanoma. N Engl J Med 2010;
and apply them to future collaborative opportunities to help 363(8): 711–723.
avoid further fragmentation and duplication. 3. Ledford H. Melanoma drug wins US approval. Nature 2011; 471(7340):
Public IO-focused information hubs would help coordinate ef- 561.
forts, optimize research resources, and identify rising opportuni- 4. Topalian SL, Hodi FS, Brahmer JR et al. Safety, activity, and immune
correlates of anti-PD-1 antibody in cancer. N Engl J Med 2012; 366(26):
ties in the field. In basic research, the online cancer-immunity
2443–2454.
cycle (https://cancer-immunity.nature.com/pages/map), which 5. Grupp SA, Kalos M, Barrett D et al. Chimeric antigen receptor-modified
evolved from a recent landmark review paper, serves as a central- T cells for acute lymphoid leukemia. N Engl J Med 2013; 368(16):
ized information hub for new IO targets and factors that affect 1509–1518.
tumor immunology [28]. In clinical development, we realized the 6. Locke FL, Neelapu SS, Bartlett NL et al. Phase 1 results of ZUMA-1: a
urgent need of an updated, scientifically curated landscape for multicenter study of KTE-C19 anti-CD19 CAR T cell therapy in refrac-
the clinical IO community. Along with this publication, we will tory aggressive lymphoma. Mol Ther 2017; 25(1): 285–295.
7. Topp MS, Kufer P, Gokbuget N et al. Targeted therapy with the T-cell-
make all related resources available on CRI’s website (www.can
engaging antibody blinatumomab of chemotherapy-refractory minimal
cerresearch.org/IO-landscape). These analyses will be updated on residual disease in B-lineage acute lymphoblastic leukemia patients re-
a quarterly basis. Working with the IO community, we aim to sults in high response rate and prolonged leukemia-free survival. J Clin
build an unbiased, neutral, comprehensive information hub for Oncol 2011; 29: 2493–2498.
clinical IO. 8. Emens LA, Ascierto PA, Darcy PK et al. Cancer immunotherapy: oppor-
tunities and challenges in the rapidly evolving clinical landscape. Eur J
Cancer.2017; 81: 116–129.
Conclusion 9. Nishino M, Ramaiya NH, Hatabu H, Hodi FS. Monitoring immune-
checkpoint blockade: response evaluation and biomarker development.
It is probably the best time for progress in oncology in the past Nat Rev Clin Oncol 2017; 14(11): 655–668.
decades. The historic opportunity would be maximally capital- 10. DeFrancesco L. Landmark approval for Dendreon’s cancer vaccine. Nat
ized if people from academia, industry, regulatory agencies, and Biotechnol 2010; 28(6): 531–532.

90 | Tang et al. Volume 29 | Issue 1 | 2018


Annals of Oncology Review
11. First-ever CAR T-cell therapy approved in U.S. Cancer Discov 2017; 7: 19. Garon EB, Rizvi NA, Hui R et al. Pembrolizumab for the treatment of
OF1. non-small-cell lung cancer. N Engl J Med 2015; 372(21): 2018–2028.
12. Golubovskaya V. CAR-T cell therapy: from the bench to the bedside. 20. Le DT, Uram JN, Wang H et al. PD-1 blockade in tumors with
Cancers (Basel) 2017; 9(11): 150. mismatch-repair deficiency. N Engl J Med 2015; 372(26): 2509–2520.
13. Curran MA, Montalvo W, Yagita H, Allison JP. PD-1 and CTLA-4 com- 21. Le DT, Durham JN, Smith KN et al. Mismatch repair deficiency predicts re-
bination blockade expands infiltrating T cells and reduces regulatory T sponse of solid tumors to PD-1 blockade. Science 2017; 357(6349): 409–413.
and myeloid cells within B16 melanoma tumors. Proc Natl Acad Sci U S 22. Mullard A. Genetic biomarker trumps tissue type in landmark oncology
A 2010; 107(9): 4275–4280. approval. Nat Rev Drug Discov 2017; 16(7): 447.
14. Larkin J, Chiarion-Sileni V, Gonzalez R et al. Combined nivolumab and 23. Johnson JR, Ning YM, Farrell A et al. Accelerated approval of oncology
ipilimumab or monotherapy in untreated melanoma. N Engl J Med products: the food and drug administration experience. J Natl Cancer
2015; 373(1): 23–34. Inst 2011; 103(8): 636–644.
15. Langer CJ, Gadgeel SM, Borghaei H et al. Carboplatin and pemetrexed 24. Dagher R, Johnson J, Williams G et al. Accelerated approval of oncology
with or without pembrolizumab for advanced, non-squamous non- products: a decade of experience. J Natl Cancer Inst 2004; 96(20):
small-cell lung cancer: a randomised, phase 2 cohort of the open-label 1500–1509.
KEYNOTE-021 study. Lancet Oncol 2016; 17(11): 1497–1508. 25. Woodcock J, LaVange LM. Master protocols to study multiple therapies,
16. Antonia SJ, Villegas A, Daniel D et al. Durvalumab after chemoradio- multiple diseases, or both. N Engl J Med 2017; 377(1): 62–70.
therapy in stage III non-small-cell lung cancer. N Engl J Med 2017; 26. Barker AD, Sigman CC, Kelloff GJ et al. I-SPY 2: an adaptive breast can-
377(20): 1919–1929. cer trial design in the setting of neoadjuvant chemotherapy. Clin
17. Kang SP, Gergich K, Lubiniecki GM et al. Pembrolizumab KEYNOTE- Pharmacol Ther 2009; 86(1): 97–100.
001: an adaptive study leading to accelerated approval for two indica- 27. Herbst RS, Gandara DR, Hirsch FR et al. Lung master protocol (Lung-
tions and a companion diagnostic. Ann Oncol 2017; 28(6): 1388–1398. MAP)—a biomarker-driven protocol for accelerating development of
18. Robert C, Ribas A, Wolchok JD et al. Anti-programmed-death-receptor- therapies for squamous cell lung cancer: SWOG S1400. Clin Cancer Res
1 treatment with pembrolizumab in ipilimumab-refractory advanced 2015; 21(7): 1514–1524.
melanoma: a randomised dose-comparison cohort of a phase 1 trial. 28. Chen DS, Mellman I. Elements of cancer immunity and the cancer-
Lancet 2014; 384(9948): 1109–1117. immune set point. Nature 2017; 541(7637): 321–330.

Volume 29 | Issue 1 | 2018 doi:10.1093/annonc/mdx755 | 91

You might also like